Next Article in Journal
The Upper Bound Theorem in Forging Processes: Model of Triangular Rigid Zones on Parts with Horizontal Symmetry
Next Article in Special Issue
Diarylureas as Antitumor Agents
Previous Article in Journal
4D Printing of Origami Structures for Minimally Invasive Surgeries Using Functional Scaffold
Previous Article in Special Issue
Ibrutinib Could Suppress CA-125 in Ovarian Cancer: A Hypothesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Updated Review of Smac Mimetics, LCL161, Birinapant, and GDC-0152 in Cancer Treatment

by
Yung-Chieh Chang
1 and
Chun Hei Antonio Cheung
1,2,*
1
Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No. 1 University Road, Tainan 701, Taiwan
2
Department of Pharmacology, College of Medicine, National Cheng Kung University, No. 1 University Road, Tainan 701, Taiwan
*
Author to whom correspondence should be addressed.
Appl. Sci. 2021, 11(1), 335; https://doi.org/10.3390/app11010335
Submission received: 5 December 2020 / Revised: 26 December 2020 / Accepted: 27 December 2020 / Published: 31 December 2020
(This article belongs to the Special Issue Anticancer Drugs Activity and Underlying Mechanisms)

Abstract

:
Inhibitor of apoptosis proteins (IAPs) are suggested as therapeutic targets for cancer treatment. Smac/DIABLO is a natural IAP antagonist in cells; therefore, Smac mimetics have been developed for cancer treatment in the past decade. In this article, we review the anti-cancer potency and novel molecular targets of LCL161, birinapant, and GDC-0152. Preclinical studies demonstrated that Smac mimetics not only induce apoptosis but also arrest cell cycle, induce necroptosis, and induce immune storm in vitro and in vivo. The safety and tolerance of Smac mimetics are evaluated in phase 1 and phase 2 clinical trials. In addition, the combination of Smac mimetics and chemotherapeutic compounds was reported to improve anti-cancer effects. Interestingly, the novel anti-cancer molecular mechanism of action of Smac mimetics was reported in recent studies, suggesting that many unknown functions of Smac mimetics still need to be revealed. Exploring these currently unknown signaling pathways is important to provide hints for the modification and combination therapy of further compounds.

1. Introduction

Inhibitor of apoptosis proteins (IAPs), including cellular inhibitor of apoptosis protein 1 (cIAP1), cellular inhibitor of apoptosis protein 2 (cIAP2), melanoma inhibitor of apoptosis (ML-IAP/Livin), testis-specific inhibitor of apoptosis (Ts-IAP/ILP-2), neuronal apoptosis inhibitory protein (NAIP), X-linked inhibitor of apoptosis protein (XIAP), survivin, and BIR repeat containing ubiquitin-conjugating enzyme (BRUCE), are known for their anti-apoptotic effects [1,2]. Members of the IAP family are characterized by the presence of the baculoviral IAP repeat (BIR) domain, which physically interacts with caspase proteins and inhibits the activity of caspases. BIR domains are grouped into two types, based on the presence or absence of the IAP binding motif (IBM) on the BIR domain [3]. Only type II BIR domain, which is with IBM, can interact with caspases. The BIR1-BIR2 linker of XIAP interacts with caspase-3 and -7 [3]. The BIR3 of XIAP inhibits the activity of caspase-9 by interacting with the N-terminal tetrapeptide of caspase-9 [4]. IAPs play important roles in mediating a variety of cellular processes, including apoptosis, mitosis, autophagy, and DNA damage in cancer cells [5,6,7,8,9,10,11,12,13,14]. Therefore, the dysregulation of IAPs promotes tumorigenesis, metastasis, angiogenesis, and therapeutic resistance, including chemotherapy and radiotherapy [15,16,17,18]. Currently, many IAP-targeting treatments, such as small-molecule inhibitors (i.e., ASTX660, Embelin, and YM155) [19,20,21], anti-sense oligonucleotides (i.e., LY2181308) [22], and Smac mimetic compounds (i.e., birinapant, LCL161, and GDC-0152) [23,24,25], have been developed. Despite the anti-cancer potency of birinapant, LCL161, and GDC-0152, they are still under investigation in preclinical and clinical studies, and these compounds have already received much attention in recent years. In this review, we mainly focus on describing the current development of LCL161, birinapant, and GDC-0152 as anti-cancer agents, and we discuss the potential of using these agents for the treatment of cancer in the future.

2. Smac/DIABLO Inhibits IAPs in Cancer

Second mitochondria-derived activator of apoptosis/direct inhibitor of apoptosis-binding protein with low pI (Smac/DIABLO) physically interacts with IAPs and antagonizes the anti-apoptotic activity of IAPs in cells, resulting in apoptosis (Figure 1) [26,27,28]. In the presence of apoptosis stimuli, mature Smac/DIABLO is released from the mitochondria to cytosol [26,27]. Smac/DIABLO interacts with the BIR domain of IAPs by its particular NH2-terminal motif consisting of four amino acids (Ala-Val-Pro-Ile), and it releases caspases from IAPs, thereby inducing caspase-dependent apoptosis [26]. Previous studies demonstrated that Smac/DIABLO interacts with the BIR2 and BIR3 domains of XIAP, and caspase-3 and -9 are released from XIAP, respectively (Figure 1) [4,29]. Smac/DIABLO not only mediates the cellular function of XIAP but also regulates cIAP1 and cIAP2. Smac/DIABLO induces the ubiquitination and degradation of cIAP1 and cIAP2 (Figure 1) [30]. However, Smac/DIABLO does not degrade XIAP [31]. Interestingly, a recent study found that a Smac/DIABLO isoform, Smac3, induces the autoubiquitination and degradation of XIAP [32]. On the other hand, cIAP1 and cIAP2 can ubiquitinate receptor-interacting kinase 1 (RIPK1), resulting in the inhibition of caspase-dependent apoptosis and necroptosis [1,33,34]. Moreover, cIAP1 and cIAP2 promote cell proliferation, migration, and invasion by activating the canonical nuclear factor kappa-light-chain-enhancer of the activated B cell (NF-κB) signaling pathway [1]. Therefore, upregulation of Smac/DIABLO can induce caspase-dependent apoptosis through the de-ubiquitination of receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and necroptosis by activating the RIPK1/receptor-interacting serine/threonine-protein kinase 3 (RIPK3)/mixed lineage kinase domain-like protein (MLKL) signaling pathway [35]. Pathologically, the protein expression level of Smac is frequently downregulated in renal carcinoma, colorectal cancer, bladder cancer, lung cancer, hepatocellular carcinoma, testicular germ cell tumors, and pancreatic cancer compared with normal tissues, but not in cervical cancer [36,37,38,39,40,41,42,43,44,45]. For these reasons, the use of Smac mimetics was suggested as a potential approach for cancer treatment.

3. Smac Mimetics for Cancer Treatment

The first Smac mimetic compound with eight amino acids was studied in 2000 [46]. Currently, eight Smac mimetics have been developed, and their anti-cancer potency has been evaluated in different preclinical and clinical studies. Smac mimetics are classified into two groups based on the number of Smac-mimicking moieties; for example, monovalent compounds contain one Smac-mimicking moiety (i.e., LCL161, AT-406 (Debio1143), GDC-0152, and GDC-0917 (CUDC-427)), and bivalent compounds are Smac-mimicking elements connected via a linker (i.e., birinapant (TL32711), BV6, and SM-164) (Figure 2) [23,47,48,49,50,51,52]. The anti-cancer potency of six of them has been elucidated in clinical trials. Bivalent compounds exhibit higher anti-cancer potency than monovalent compounds because the former possess better binding affinity with IAPs and higher potency to induce caspase-dependent apoptosis than the latter [53]. The anti-cancer potency of Smac mimetics is also dependent on their specificity to IAPs. For example, birinapant and AT406 preferentially target cIAP1 and cIAP2 rather than XIAP. LCL161 and GDC-0152 are pan-IAP inhibitors that have similar affinities to XIAP, cIAP1, and cIAP2 [23,48,49,51]. Among these Smac mimetic compounds, LCL161, birinapant, and GDC-0152 are currently the most popular, in which their therapeutic effectiveness and the molecular mechanism of actions have been studied extensively in pre-clinical and clinical studies. Therefore, we summarize current LCL161, birinapant, and GDC-0152-related findings in the following sections.

4. Anti-Cancer Molecular Mechanism of LCL161

LCL161 is an orally bioavailable monovalent Smac mimetic developed by Novartis Pharmaceuticals. LCL161 shows both pro-apoptotic and anti-proliferation effects in cancer cells [47]. In many preclinical studies, the anti-cancer potency of LCL161 has been established in multiple myeloma, glioblastoma, hepatocellular carcinoma, oral squamous carcinoma, neuroblastoma, osteosarcoma, sarcoma, triple-negative breast cancer (TNBC), leukemia, cervical cancer, non-small cell lung cancer, and head and neck cancer cells [24,54,55,56]. At the molecular level, LCL161 binds to the BIR3 domain of cIAP1 and cIAP2 with high affinity, and induces cIAP1 and cIAP2 autoubiquitination and proteasome degradation, resulting in the activation of non-canonical NF-κB signaling pathways and production of tumor necrosis factor α (TNF-α) (Figure 3) [57]. LCL161 also induces apoptosis and necroptosis through the RIP1/RIP3/MLKL signaling pathway (Figure 3) [58,59]. Surprisingly, a few studies showed that LCL161 does not induce apoptosis, but instead it induces G2/M phase arrest by downregulating cIAP1 and activating the p21 signaling pathway in medulloblastoma and neuroblastoma cells (Figure 3) [60,61]. Moreover, it has been shown that LCL161 does not kill cancer cells directly, but it promotes the activity of immune cells and enhances cytokine secretion instead. Mechanistically, LCL161 promotes the activity of T cells by inducing cytokine secretion and dendritic cell maturation (Figure 3) [62,63,64]. In multiple myeloma, cIAP1 and cIAP2 are usually deleted, but LCL161 still inhibits the growth of multiple myeloma through the upregulation of tumor cell autonomous type I interferon signaling, and strong acute inflammatory signaling (promoting macrophagy, M-spike, and dendritic cell maturation) in transgenic Vk*MYC mice [65]. Recent studies demonstrated the anti-cancer potency of LCL161 is correlated with the protein expression levels of B-cell lymphoma 2 (BCL-2) and TNF-α and NF-κB activity in cancer cells. In neuroblastoma, Langemann et al. suggested that the anti-cancer potency of LCL161 is based on the cells’ sensitivity to TNF-α and modulation of NF-κB [61]. Interestingly, in our recent study, we found that LCL161 interacts with p-glycoprotein (also called multidrug resistance protein 1 (MDR1)) and inhibits the MDR1 multi-drug efflux activity, resulting in increased sensitivity to the MDR1 substrates, such as paclitaxel and YM155 in cervical and bladder cancer cells [66]. Moreover, we found that LCL161 downregulates the protein expression level of survivin in MDR1-expressing cervical and bladder cancer cells [66].

5. LCL161 Combination Treatment

Many preclinical studies have explored the anti-cancer effects of LCL161 in combination with different anti-cancer agents. It has been demonstrated that LCL161 sensitizes cancer cells to paclitaxel, Fas ligand, vincristine, and obatoclax (BCL-2 inhibitor) in different types of cancer [54,55,60,61,67,68,69,70]. Moreover, LCL161 improves the anti-cancer effects of radiotherapy in head and neck squamous and esophageal carcinoma cells [71,72]. Immunotherapy is a popular treatment for cancer. Chesi et al. noted improvements in the anti-cancer effects of immunotherapy when combined with LCL161 in multiple myeloma in vivo [65]. Maintaining the balance of reactive oxygen species (ROS) plays an important role in tumorigenesis. Targeting redox homeostasis with erastin and auranofin, which are ROS inducers, can improve LCL161-induced cell death in acute lymphoblastic leukemia cells [73].

6. Current Status of LCL161 in Clinical Trials

LCL161 is currently involved in many phase 1 and phase 2 clinical trials (Table 1), and results showed that it has favorable pharmacological properties, such as good tolerability and minor toxicity. Infante et al. showed that LCL161 is well tolerated at doses of up to 1800 mg [74]. Some LCL161-treated patients may present with vomiting, nausea, asthenia, and anorexia side effects, but these effects are not severe after treatment once a week for 21 days [74]. Only 3 out of 53 patients studied demonstrated cytokine release syndrome [74]. LCL161 was rapidly absorbed at the time to reach maximum plasma concentration (i.e., 0.5–2 h), and the plasma concentration declined within the range of 4–16 h [74].
Combination therapy of LCL161 with chemotherapeutic drugs such as paclitaxel and gemcitabine has been evaluated in patients with cancer (Table 1). Bardia et al. found that (1) LCL161 combined with paclitaxel can improve the pathologic complete response of TNF-α-positive TNBC, (2) the expression level of TNF-α might be a biomarker for predicting the anti-cancer effects of LCL161 and paclitaxel combination treatment, and (3) the anti-cancer potency of this combination is not correlated with the alternation of breast cancer type 1 susceptibility protein (BRCA1) and breast cancer type 2 susceptibility protein (BRCA2) in TNBC [75]. Some adverse effects were observed during combination treatment, including pyrexia (5 of 209 patients studied), pneumonia (4 of 209 patients studied), and pneumonitis (4 of 209 patients studied).

7. Anti-Cancer Molecular Mechanisms of Birinapant (TL32711)

Birinapant is a second-generation bivalent Smac mimetic, first synthesized in 2014 [76,77]. Birinapant has better tolerability for treating solid tumors compared with the first-generation bivalent Smac mimetic, and it preferentially targets cIAP1, relative to cIAP2 and XIAP [76]. The anti-cancer potency of birinapant has been investigated in acute myeloid leukemia, melanoma, colorectal cancer, ovarian cancer, breast cancer, head and neck squamous cell carcinoma, hepatocellular carcinoma, glioblastoma, and breast cancer [23,77,78,79,80,81,82,83,84]. At the molecular level, birinapant binds the BIR domains of IAPs and promotes the degradation of TNF receptor-associated factor (TRAF)-bound IAPs, resulting in the induction of caspase-8-dependent apoptosis (Figure 3) [85]. In addition, birinapant degrades IAPs, activates the non-canonical NF-κB signaling pathway once the IAPs are degraded, and stabilizes mitogen-activated protein kinase 14 (MAP3K14) (NF-κB-inducing kinase), resulting in the activation of the non-canonical NF-κB signaling pathway [84,86]. Mak et al. demonstrated that apoptosis repressor with caspase recruitment domain, an apoptosis suppressor, decreases the anti-cancer effect of birinapant by inhibiting the cIPA1/MAP3K14 signaling pathway in acute myeloid leukemia [87]. Birinapant induces cell cycle G2/M arrest in head and neck squamous cell carcinoma; however, the underlying molecular mechanism of action remains unclear (Figure 3) [79,88]. In acute lymphoblastic leukemia, birinapant induces RIP-1-dependent necroptosis (Figure 3) [83]. When birinapant is combined with emricasan, a caspase-8 inhibitor, necroptosis is induced in acute myeloid leukemia [89]. Kearney et al. demonstrated that birinapant-induced TNF activates CD8+ T cells and natural killer cells, resulting in cancer cell death (Figure 3) [90]. Targeting programmed death-ligand 1 (PD-1) upregulates TNF production; therefore, the combination of birinapant and PD-1 blockade increases TNF production and promotes the anti-cancer potency of immune therapy [90].

8. Combination Therapy with Birinapant for Cancer Treatment

Overexpression of IAPs induces chemotherapeutic drug resistance in many cancer types. Targeting IAPs sensitizes drug-resistant cancer cells to chemotherapeutic drugs. Birinapant as an IAP antagonist has been combined with many chemotherapeutic drugs for cancer treatment. Birinapant has been combined with norcanthanidin, docetaxel, or TNF-related apoptosis-inducing ligand (TRAIL) for breast cancer treatment [23,91,92]. In addition, birinapant has been combined with paclitaxel, demethylating agent (5-AZ), TNF-α, Fas ligand (FasL), gemcitabine, 5-flurouracie (5-FU), and oxaliplatin for different types of cancer treatment, such as pancreatic cancer, head and neck squamous cell carcinoma, melanoma, acute myeloid leukemia, and colon cancer [79,80,84,86,88,93,94,95,96,97]. Birinapant improves the anti-cancer effects of radiotherapy for treating Fas-associated protein with death domain (FADD) and cIAP1-overexpressed head and neck cancers [88]. Kearney et al. demonstrated that birinapant activates CD8 T-cell and natural killer cells, resulting in cancer cell death [90]. The anti-cancer effects of chimeric antigen receptor (CAR) T-cell therapy correlate directly with the level of TNF. To improve the anti-cancer potency of CAR T cells, Michie et al. combined CAR T-cell therapy with birinapant for cancer treatment. Results showed that this combination approach significantly reduced cancer growth [98].

9. Current Status of Birinapant in Clinical Trials

Clinical trials of birinapant are listed in Table 2. A phase 1 trial study reported the maximum tolerated dose, safety, and pharmacokinetic properties of birinapant in solid tumors [99]. The maximum tolerated dose of birinapant is 47 mg/m2 [99]. The half-life of birinapant is 30–35 h [99]. A phase 2 clinical trial demonstrated that birinapant has a plasma half-life of 31 h and a tumor tissue half-life of 52 h [100]. Birinapant accumulates in tumor tissues, resulting in the downregulated protein expression level of cIAP1 and apoptosis induced in peripheral blood mononuclear cells and cancer cells [99]. However, birinapant does not increase the protein level of TNF, monocyte chemoattractant protein-1 (MCP-1), or interleukin 1, 6, and 8 [99]. For treating patients with cancer, birinapant is used as a single agent or in combination therapy with chemotherapeutic drugs (i.e., pembrolizumab, carboplatin, docetaxel, and gemcitabine) and radiotherapy. However, some birinapant clinical trials were terminated because birinapant lacked anti-cancer efficacy (NCT02147873, NCT02587962, NCT01681368), or the sponsors did not fund further work (NCT01573780).

10. GDC-0152 for Cancer Treatment

The development of GDC-0152, a pan-IAP antagonist, as an anti-cancer agent was first reported in 2012 [49]. The anti-cancer potency of GDC-0152 was evaluated in glioblastoma, leukemia, osteosarcoma, and glioblastoma [49,101,102,103,104]. At the molecular level, GDC-0152 induces intrinsic caspase-dependent apoptosis and inhibits the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway, resulting in cancer cell death [101,102]. Tchoghandjian et al. demonstrated that GDC-0152 not only downregulates the protein expression level of cIAP1, cIAP2, and XIAP, but it also downregulates ML-IAP in glioblastoma cells in vitro and in vivo [104]. Mantik et al. found that the pharmacological properties of GDC-0152 are correlated with pH [105]. They demonstrated that the presence of succinic acid and hydroxypropyl-β-cyclodextrin can increase the solubility of GDC-0152, thereby improving blood compatibility, reducing hemolysis, and increasing the maximum tolerated dose [105]. Additionally, they remain stable in human plasma for up to 25 h [106]. Only one phase 1 clinical trial has studied the safety and pharmacokinetics of GDC-0152 (NCT00977067); however, it was terminated for reasons unrelated to patient safety or anti-cancer activity in 2017.

11. Conclusions and Future Directions

As dysregulation of IAPs has been found in a variety of tumors, and overexpression of IAPs promotes tumorigenesis and tumor metastasis, the use of Smac mimetics is suggested as a potential therapeutic approach for the treatment of cancer. The anti-cancer potency of Smac mimetics has been demonstrated in different types of malignant tumors in preclinical studies. Smac mimetics induce apoptosis and the non-canonical NF-kB signaling pathway by downregulating the protein expression level of IAPs. However, many unknown anti-cancer molecular mechanisms of action of Smac mimetics are still yet to be discovered. Exploring these signaling pathways is important to provide hints for the modification and combination therapy of further compounds.
The safety and tolerance of Smac mimetics are currently investigated in many clinical studies. Most studies report that Smac mimetics, such as LCL161, are safe and have good tolerance in patients with cancer. However, other Smac mimetics have shown low anti-cancer potency in the treatment of patients with cancer. The underlying molecular mechanism of action is still unclear. The anti-cancer potency of most Smac mimetics is on the micromolar level. Thus, many studies have combined Smac mimetics with chemotherapeutic drugs, radiotherapy, and immune therapy for cancer treatment. Results showed that the synergistic effect of these combinations can decrease the dosage of Smac mimetics. With the advancement of nanotechnology, delivering chemotherapeutic drugs with various types of nanoparticles can improve anti-cancer potency and cancer-targeting specificity through active or passive cancer targeting. For instance, Nikkhoo et al. synthesized a chitosan-based nanoparticle for co-delivering STAT3 siRNA and BV6, a bivalent Smac mimetic, to treat breast cancer cells, colorectal carcinoma cells, and melanoma cells, and their nanoparticles were shown to suppress cancer cell progression through caspase-dependent apoptosis in vitro and in vivo [107]. Therefore, nanotechnology can potentially be applied to improve the anti-cancer potency of Smac mimetics.

Author Contributions

Literature research: Y.-C.C. and C.H.A.C.; Figure and table preparation: Y.-C.C.; Manuscript writing and proofreading: Y.-C.C. and C.H.A.C. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by Ministry of Science and Technology, Taiwan (MOST 109-2320-B-006-031).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

IAPs: Inhibitor of apoptosis proteins; BRCA1: breast cancer type 1 susceptibility protein; BRCA2: breast cancer type 2 susceptibility protein; BRUCE: BIR repeat containing ubiquitin-conjugating enzyme; cIAP1: cellular inhibitor of apoptosis protein 1; cIAP2: cellular inhibitor of apoptosis 2; DIABLO: direct inhibitor of apoptosis-binding protein with low pI; ML-IAP: melanoma inhibitor of apoptosis; Smac: second mitochondria-derived activator of apoptosis; NAIP: neuronal apoptosis inhibitory protein; TNBC: triple-negative breast cancer; TNF-α: tumor necrosis factor α; Ts-IAP: testis-specific inhibitor of apoptosis.

References

  1. Fulda, S.; Vucic, D. Targeting IAP proteins for therapeutic intervention in cancer. Nat. Rev. Drug Discov. 2012, 11, 109–124. [Google Scholar] [CrossRef] [PubMed]
  2. Wei, Y.; Fan, T.; Yu, M. Inhibitor of apoptosis proteins and apoptosis. Acta Biochim. Biophys. Sin. 2008, 40, 278–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Eckelman, B.P.; Drag, M.; Snipas, S.J.; Salvesen, G.S. The mechanism of peptide-binding specificity of IAP BIR domains. Cell Death Differ. 2008, 15, 920–928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Shiozaki, E.N.; Chai, J.; Rigotti, D.J.; Riedl, S.J.; Li, P.; Srinivasula, S.M.; Alnemri, E.S.; Fairman, R.; Shi, Y. Mechanism of XIAP-mediated inhibition of caspase-9. Mol. Cell 2003, 11, 519–527. [Google Scholar] [CrossRef]
  5. Lin, T.Y.; Chan, H.H.; Chen, S.H.; Sarvagalla, S.; Chen, P.S.; Coumar, M.S.; Cheng, S.M.; Chang, Y.C.; Lin, C.H.; Leung, E.; et al. BIRC5/Survivin is a novel ATG12-ATG5 conjugate interactor and an autophagy-induced DNA damage suppressor in human cancer and mouse embryonic fibroblast cells. Autophagy 2020, 16, 1296–1313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Fulda, S.; Debatin, K.M. Extrinsic versus intrinsic apoptosis pathways in anticancer chemotherapy. Oncogene 2006, 25, 4798–4811. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Gyrd-Hansen, M.; Meier, P. IAPs: From caspase inhibitors to modulators of NF-kappaB, inflammation and cancer. Nat. Rev. Cancer 2010, 10, 561–574. [Google Scholar] [CrossRef] [PubMed]
  8. Wertz, I.E.; Dixit, V.M. Regulation of death receptor signaling by the ubiquitin system. Cell Death Differ. 2010, 17, 14–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Dogan, T.; Harms, G.S.; Hekman, M.; Karreman, C.; Oberoi, T.K.; Alnemri, E.S.; Rapp, U.R.; Rajalingam, K. X-linked and cellular IAPs modulate the stability of C-RAF kinase and cell motility. Nat. Cell Biol. 2008, 10, 1447–1455. [Google Scholar] [CrossRef] [PubMed]
  10. Uren, A.G.; Beilharz, T.; O’Connell, M.J.; Bugg, S.J.; van Driel, R.; Vaux, D.L.; Lithgow, T. Role for yeast inhibitor of apoptosis (IAP)-like proteins in cell division. Proc. Natl. Acad. Sci. USA 1999, 96, 10170–10175. [Google Scholar] [CrossRef] [Green Version]
  11. Ebner, P.; Poetsch, I.; Deszcz, L.; Hoffmann, T.; Zuber, J.; Ikeda, F. The IAP family member BRUCE regulates autophagosome-lysosome fusion. Nat. Commun. 2018, 9, 599. [Google Scholar] [CrossRef] [PubMed]
  12. Sauer, M.; Reiners, K.S.; Hansen, H.P.; Engert, A.; Gasser, S.; von Strandmann, E.P. Induction of the DNA damage response by IAP inhibition triggers natural immunity via upregulation of NKG2D ligands in Hodgkin lymphoma in vitro. Biol. Chem. 2013, 394, 1325–1331. [Google Scholar] [CrossRef] [PubMed]
  13. Cheng, S.M.; Chang, Y.C.; Liu, C.Y.; Lee, J.Y.; Chan, H.H.; Kuo, C.W.; Lin, K.Y.; Tsai, S.L.; Chen, S.H.; Li, C.F.; et al. YM155 down-regulates survivin and XIAP, modulates autophagy and induces autophagy-dependent DNA damage in breast cancer cells. Br. J. Pharmacol. 2015, 172, 214–234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ghobrial, I.M.; Witzig, T.E.; Adjei, A.A. Targeting apoptosis pathways in cancer therapy. CA Cancer J. Clin. 2005, 55, 178–194. [Google Scholar] [CrossRef] [PubMed]
  15. Mehrotra, S.; Languino, L.R.; Raskett, C.M.; Mercurio, A.M.; Dohi, T.; Altieri, D.C. IAP regulation of metastasis. Cancer Cell 2010, 17, 53–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Liu, J.; Zhang, D.; Luo, W.; Yu, Y.; Yu, J.; Li, J.; Zhang, X.; Zhang, B.; Chen, J.; Wu, X.R.; et al. X-linked inhibitor of apoptosis protein (XIAP) mediates cancer cell motility via Rho GDP dissociation inhibitor (RhoGDI)-dependent regulation of the cytoskeleton. J. Biol. Chem. 2011, 286, 15630–15640. [Google Scholar] [CrossRef] [Green Version]
  17. Tran, J.; Rak, J.; Sheehan, C.; Saibil, S.D.; LaCasse, E.; Korneluk, R.G.; Kerbel, R.S. Marked induction of the IAP family antiapoptotic proteins survivin and XIAP by VEGF in vascular endothelial cells. Biochem. Biophys. Res. Commun. 1999, 264, 781–788. [Google Scholar] [CrossRef]
  18. Rathore, R.; McCallum, J.E.; Varghese, E.; Florea, A.M.; Busselberg, D. Overcoming chemotherapy drug resistance by targeting inhibitors of apoptosis proteins (IAPs). Apoptosis 2017, 22, 898–919. [Google Scholar] [CrossRef]
  19. Ward, G.A.; Lewis, E.J.; Ahn, J.S.; Johnson, C.N.; Lyons, J.F.; Martins, V.; Munck, J.M.; Rich, S.J.; Smyth, T.; Thompson, N.T.; et al. ASTX660, a Novel Non-peptidomimetic Antagonist of cIAP1/2 and XIAP, Potently Induces TNFalpha-Dependent Apoptosis in Cancer Cell Lines and Inhibits Tumor Growth. Mol. Cancer Ther. 2018, 17, 1381–1391. [Google Scholar] [CrossRef] [Green Version]
  20. Ahn, K.S.; Sethi, G.; Aggarwal, B.B. Embelin, an inhibitor of X chromosome-linked inhibitor-of-apoptosis protein, blocks nuclear factor-kappaB (NF-kappaB) signaling pathway leading to suppression of NF-kappaB-regulated antiapoptotic and metastatic gene products. Mol. Pharmacol. 2007, 71, 209–219. [Google Scholar] [CrossRef]
  21. Nakahara, T.; Kita, A.; Yamanaka, K.; Mori, M.; Amino, N.; Takeuchi, M.; Tominaga, F.; Hatakeyama, S.; Kinoyama, I.; Matsuhisa, A.; et al. YM155, a novel small-molecule survivin suppressant, induces regression of established human hormone-refractory prostate tumor xenografts. Cancer Res. 2007, 67, 8014–8021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Talbot, D.C.; Ranson, M.; Davies, J.; Lahn, M.; Callies, S.; Andre, V.; Kadam, S.; Burgess, M.; Slapak, C.; Olsen, A.L.; et al. Tumor survivin is downregulated by the antisense oligonucleotide LY2181308: A proof-of-concept, first-in-human dose study. Clin. Cancer Res. 2010, 16, 6150–6158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Allensworth, J.L.; Sauer, S.J.; Lyerly, H.K.; Morse, M.A.; Devi, G.R. Smac mimetic Birinapant induces apoptosis and enhances TRAIL potency in inflammatory breast cancer cells in an IAP-dependent and TNF-alpha-independent mechanism. Breast Cancer Res. Treat. 2013, 137, 359–371. [Google Scholar] [CrossRef] [PubMed]
  24. Houghton, P.J.; Kang, M.H.; Reynolds, C.P.; Morton, C.L.; Kolb, E.A.; Gorlick, R.; Keir, S.T.; Carol, H.; Lock, R.; Maris, J.M.; et al. Initial testing (stage 1) of LCL161, a SMAC mimetic, by the Pediatric Preclinical Testing Program. Pediatr. Blood Cancer 2012, 58, 636–639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Zhen, M.C.; Wang, F.Q.; Wu, S.F.; Zhao, Y.L.; Liu, P.G.; Yin, Z.Y. Identification of mTOR as a primary resistance factor of the IAP antagonist AT406 in hepatocellular carcinoma cells. Oncotarget 2017, 8, 9466–9475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Du, C.; Fang, M.; Li, Y.; Li, L.; Wang, X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 2000, 102, 33–42. [Google Scholar] [CrossRef] [Green Version]
  27. Verhagen, A.M.; Ekert, P.G.; Pakusch, M.; Silke, J.; Connolly, L.M.; Reid, G.E.; Moritz, R.L.; Simpson, R.J.; Vaux, D.L. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 2000, 102, 43–53. [Google Scholar] [CrossRef] [Green Version]
  28. Kominsky, D.J.; Bickel, R.J.; Tyler, K.L. Reovirus-induced apoptosis requires mitochondrial release of Smac/DIABLO and involves reduction of cellular inhibitor of apoptosis protein levels. J. Virol. 2002, 76, 11414–11424. [Google Scholar] [CrossRef] [Green Version]
  29. Scott, F.L.; Denault, J.B.; Riedl, S.J.; Shin, H.; Renatus, M.; Salvesen, G.S. XIAP inhibits caspase-3 and -7 using two binding sites: Evolutionarily conserved mechanism of IAPs. EMBO J. 2005, 24, 645–655. [Google Scholar] [CrossRef]
  30. Darding, M.; Feltham, R.; Tenev, T.; Bianchi, K.; Benetatos, C.; Silke, J.; Meier, P. Molecular determinants of Smac mimetic induced degradation of cIAP1 and cIAP2. Cell Death Differ. 2011, 18, 1376–1386. [Google Scholar] [CrossRef]
  31. Yang, Q.H.; Du, C. Smac/DIABLO selectively reduces the levels of c-IAP1 and c-IAP2 but not that of XIAP and livin in HeLa cells. J. Biol. Chem. 2004, 279, 16963–16970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Fu, J.; Jin, Y.; Arend, L.J. Smac3, a novel Smac/DIABLO splicing variant, attenuates the stability and apoptosis-inhibiting activity of X-linked inhibitor of apoptosis protein. J. Biol. Chem. 2003, 278, 52660–52672. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Tenev, T.; Bianchi, K.; Darding, M.; Broemer, M.; Langlais, C.; Wallberg, F.; Zachariou, A.; Lopez, J.; MacFarlane, M.; Cain, K.; et al. The Ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs. Mol. Cell 2011, 43, 432–448. [Google Scholar] [CrossRef] [PubMed]
  34. Feoktistova, M.; Geserick, P.; Kellert, B.; Dimitrova, D.P.; Langlais, C.; Hupe, M.; Cain, K.; MacFarlane, M.; Hacker, G.; Leverkus, M. cIAPs block Ripoptosome formation, a RIP1/caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Mol. Cell 2011, 43, 449–463. [Google Scholar] [CrossRef] [Green Version]
  35. Akara-Amornthum, P.; Lomphithak, T.; Choksi, S.; Tohtong, R.; Jitkaew, S. Key necroptotic proteins are required for Smac mimetic-mediated sensitization of cholangiocarcinoma cells to TNF-alpha and chemotherapeutic gemcitabine-induced necroptosis. PLoS ONE 2020, 15, e0227454. [Google Scholar] [CrossRef] [PubMed]
  36. Mizutani, Y.; Nakanishi, H.; Yamamoto, K.; Li, Y.N.; Matsubara, H.; Mikami, K.; Okihara, K.; Kawauchi, A.; Bonavida, B.; Miki, T. Downregulation of Smac/DIABLO expression in renal cell carcinoma and its prognostic significance. J. Clin. Oncol. 2005, 23, 448–454. [Google Scholar] [CrossRef]
  37. Kempkensteffen, C.; Hinz, S.; Christoph, F.; Krause, H.; Magheli, A.; Schrader, M.; Schostak, M.; Miller, K.; Weikert, S. Expression levels of the mitochondrial IAP antagonists Smac/DIABLO and Omi/HtrA2 in clear-cell renal cell carcinomas and their prognostic value. J. Cancer Res. Clin. Oncol. 2008, 134, 543–550. [Google Scholar] [CrossRef]
  38. Endo, K.; Kohnoe, S.; Watanabe, A.; Tashiro, H.; Sakata, H.; Morita, M.; Kakeji, Y.; Maehara, Y. Clinical significance of Smac/DIABLO expression in colorectal cancer. Oncol. Rep. 2009, 21, 351–355. [Google Scholar] [CrossRef] [Green Version]
  39. Mizutani, Y.; Katsuoka, Y.; Bonavida, B. Prognostic significance of second mitochondria-derived activator of caspase (Smac/DIABLO) expression in bladder cancer and target for therapy. Int. J. Oncol. 2010, 37, 503–508. [Google Scholar] [CrossRef] [Green Version]
  40. Mizutani, Y.; Katsuoka, Y.; Bonavida, B. Low circulating serum levels of second mitochondria-derived activator of caspase (Smac/DIABLO) in patients with bladder cancer. Int. J. Oncol. 2012, 40, 1246–1250. [Google Scholar] [CrossRef] [Green Version]
  41. Sekimura, A.; Konishi, A.; Mizuno, K.; Kobayashi, Y.; Sasaki, H.; Yano, M.; Fukai, I.; Fujii, Y. Expression of Smac/DIABLO is a novel prognostic marker in lung cancer. Oncol. Rep. 2004, 11, 797–802. [Google Scholar] [CrossRef] [PubMed]
  42. Bao, S.T.; Gui, S.Q.; Lin, M.S. Relationship between expression of Smac and Survivin and apoptosis of primary hepatocellular carcinoma. Hepatobiliary Pancreat Dis. Int. 2006, 5, 580–583. [Google Scholar] [PubMed]
  43. Kempkensteffen, C.; Jager, T.; Bub, J.; Weikert, S.; Hinz, S.; Christoph, F.; Krause, H.; Schostak, M.; Miller, K.; Schrader, M. The equilibrium of XIAP and Smac/DIABLO expression is gradually deranged during the development and progression of testicular germ cell tumours. Int. J. Androl. 2007, 30, 476–483. [Google Scholar] [CrossRef] [PubMed]
  44. Liang, W.; Liao, Y.; Zhang, J.; Huang, Q.; Luo, W.; Yu, J.; Gong, J.; Zhou, Y.; Li, X.; Tang, B.; et al. Heat shock factor 1 inhibits the mitochondrial apoptosis pathway by regulating second mitochondria-derived activator of caspase to promote pancreatic tumorigenesis. J. Exp. Clin. Cancer Res. 2017, 36, 64. [Google Scholar] [CrossRef]
  45. Arellano-Llamas, A.; Garcia, F.J.; Perez, D.; Cantu, D.; Espinosa, M.; De la Garza, J.G.; Maldonado, V.; Melendez-Zajgla, J. High Smac/DIABLO expression is associated with early local recurrence of cervical cancer. BMC Cancer 2006, 6, 256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Chai, J.; Du, C.; Wu, J.W.; Kyin, S.; Wang, X.; Shi, Y. Structural and biochemical basis of apoptotic activation by Smac/DIABLO. Nature 2000, 406, 855–862. [Google Scholar] [CrossRef]
  47. Weisberg, E.; Ray, A.; Barrett, R.; Nelson, E.; Christie, A.L.; Porter, D.; Straub, C.; Zawel, L.; Daley, J.F.; Lazo-Kallanian, S.; et al. Smac mimetics: Implications for enhancement of targeted therapies in leukemia. Leukemia 2010, 24, 2100–2109. [Google Scholar] [CrossRef] [Green Version]
  48. Cai, Q.; Sun, H.; Peng, Y.; Lu, J.; Nikolovska-Coleska, Z.; McEachern, D.; Liu, L.; Qiu, S.; Yang, C.Y.; Miller, R.; et al. A potent and orally active antagonist (SM-406/AT-406) of multiple inhibitor of apoptosis proteins (IAPs) in clinical development for cancer treatment. J. Med. Chem. 2011, 54, 2714–2726. [Google Scholar] [CrossRef] [Green Version]
  49. Flygare, J.A.; Beresini, M.; Budha, N.; Chan, H.; Chan, I.T.; Cheeti, S.; Cohen, F.; Deshayes, K.; Doerner, K.; Eckhardt, S.G.; et al. Discovery of a potent small-molecule antagonist of inhibitor of apoptosis (IAP) proteins and clinical candidate for the treatment of cancer (GDC-0152). J. Med. Chem. 2012, 55, 4101–4113. [Google Scholar] [CrossRef]
  50. Wong, H.; Gould, S.E.; Budha, N.; Darbonne, W.C.; Kadel, E.E., III; La, H.; Alicke, B.; Halladay, J.S.; Erickson, R.; Portera, C.; et al. Learning and confirming with preclinical studies: Modeling and simulation in the discovery of GDC-0917, an inhibitor of apoptosis proteins antagonist. Drug Metab. Dispos. 2013, 41, 2104–2113. [Google Scholar] [CrossRef] [Green Version]
  51. Varfolomeev, E.; Blankenship, J.W.; Wayson, S.M.; Fedorova, A.V.; Kayagaki, N.; Garg, P.; Zobel, K.; Dynek, J.N.; Elliott, L.O.; Wallweber, H.J.; et al. IAP antagonists induce autoubiquitination of c-IAPs, NF-kappaB activation, and TNFalpha-dependent apoptosis. Cell 2007, 131, 669–681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Lu, J.; Bai, L.; Sun, H.; Nikolovska-Coleska, Z.; McEachern, D.; Qiu, S.; Miller, R.S.; Yi, H.; Shangary, S.; Sun, Y.; et al. SM-164: A novel, bivalent Smac mimetic that induces apoptosis and tumor regression by concurrent removal of the blockade of cIAP-1/2 and XIAP. Cancer Res. 2008, 68, 9384–9393. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Fulda, S. Promises and Challenges of Smac Mimetics as Cancer Therapeutics. Clin. Cancer Res. 2015, 21, 5030–5036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ramakrishnan, V.; Painuly, U.; Kimlinger, T.; Haug, J.; Rajkumar, S.V.; Kumar, S. Inhibitor of apoptosis proteins as therapeutic targets in multiple myeloma. Leukemia 2014, 28, 1519–1528. [Google Scholar] [CrossRef] [Green Version]
  55. Brands, R.C.; Herbst, F.; Hartmann, S.; Seher, A.; Linz, C.; Kubler, A.C.; Muller-Richter, U.D.A. Cytotoxic effects of SMAC-mimetic compound LCL161 in head and neck cancer cell lines. Clin. Oral. Investig. 2016, 20, 2325–2332. [Google Scholar] [CrossRef]
  56. Ren, K.; Ma, L.; Chong, D.; Zhang, Z.; Zhou, C.; Liu, H.; Zhao, S. Effects of LCL161, a Smac mimetic on the proliferation and apoptosis in hepatocellular carcinoma cells. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2016, 41, 898–904. [Google Scholar]
  57. Shekhar, T.M.; Miles, M.A.; Gupte, A.; Taylor, S.; Tascone, B.; Walkley, C.R.; Hawkins, C.J. IAP antagonists sensitize murine osteosarcoma cells to killing by TNFalpha. Oncotarget 2016, 7, 33866–33886. [Google Scholar] [CrossRef] [Green Version]
  58. Jin, G.; Lan, Y.; Han, F.; Sun, Y.; Liu, Z.; Zhang, M.; Liu, X.; Zhang, X.; Hu, J.; Liu, H.; et al. Smac mimeticinduced caspaseindependent necroptosis requires RIP1 in breast cancer. Mol. Med. Rep. 2016, 13, 359–366. [Google Scholar] [CrossRef] [Green Version]
  59. Jin, G.; Liu, Y.; Xu, P.; Jin, G. Induction of Necroptosis in Human Breast Cancer Drug-Resistant Cells by SMAC Analog LCL161 After Caspase Inhibition Requires RIP3. Pharmazie 2019, 74, 363–368. [Google Scholar]
  60. Chen, S.M.; Lin, T.K.; Tseng, Y.Y.; Tu, C.H.; Lui, T.N.; Huang, S.F.; Hsieh, L.L.; Li, Y.Y. Targeting inhibitors of apoptosis proteins suppresses medulloblastoma cell proliferation via G2/M phase arrest and attenuated neddylation of p21. Cancer Med. 2018, 7, 3988–4003. [Google Scholar] [CrossRef]
  61. Langemann, D.; Trochimiuk, M.; Appl, B.; Hundsdoerfer, P.; Reinshagen, K.; Eschenburg, G. Sensitization of neuroblastoma for vincristine-induced apoptosis by Smac mimetic LCL161 is attended by G2 cell cycle arrest but is independent of NFkappaB, RIP1 and TNF-alpha. Oncotarget 2017, 8, 87763–87772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Dougan, M.; Dougan, S.; Slisz, J.; Firestone, B.; Vanneman, M.; Draganov, D.; Goyal, G.; Li, W.; Neuberg, D.; Blumberg, R.; et al. IAP inhibitors enhance co-stimulation to promote tumor immunity. J. Exp. Med. 2010, 207, 2195–2206. [Google Scholar] [CrossRef] [PubMed]
  63. Knights, A.J.; Fucikova, J.; Pasam, A.; Koernig, S.; Cebon, J. Inhibitor of apoptosis protein (IAP) antagonists demonstrate divergent immunomodulatory properties in human immune subsets with implications for combination therapy. Cancer Immunol. Immunother. 2013, 62, 321–335. [Google Scholar] [CrossRef] [PubMed]
  64. Muller-Sienerth, N.; Dietz, L.; Holtz, P.; Kapp, M.; Grigoleit, G.U.; Schmuck, C.; Wajant, H.; Siegmund, D. SMAC mimetic BV6 induces cell death in monocytes and maturation of monocyte-derived dendritic cells. PLoS ONE 2011, 6, e21556. [Google Scholar] [CrossRef] [PubMed]
  65. Chesi, M.; Mirza, N.N.; Garbitt, V.M.; Sharik, M.E.; Dueck, A.C.; Asmann, Y.W.; Akhmetzyanova, I.; Kosiorek, H.E.; Calcinotto, A.; Riggs, D.L.; et al. IAP antagonists induce anti-tumor immunity in multiple myeloma. Nat. Med. 2016, 22, 1411–1420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Chang, Y.C.; Kondapuram, S.K.; Yang, T.H.; Syed, S.B.; Cheng, S.M.; Lin, T.Y.; Lin, Y.C.; Coumar, M.S.; Chang, J.Y.; Leung, E.; et al. The SMAC mimetic LCL161 is a direct ABCB1/MDR1-ATPase activity modulator and BIRC5/Survivin expression down-regulator in cancer cells. Toxicol. Appl. Pharmacol. 2020, 401, 115080. [Google Scholar] [CrossRef] [PubMed]
  67. Tian, A.; Wilson, G.S.; Lie, S.; Wu, G.; Hu, Z.; Hebbard, L.; Duan, W.; George, J.; Qiao, L. Synergistic effects of IAP inhibitor LCL161 and paclitaxel on hepatocellular carcinoma cells. Cancer Lett. 2014, 351, 232–241. [Google Scholar] [CrossRef]
  68. Scheurer, M.J.J.; Seher, A.; Steinacker, V.; Linz, C.; Hartmann, S.; Kubler, A.C.; Muller-Richter, U.D.A.; Brands, R.C. Targeting inhibitors of apoptosis in oral squamous cell carcinoma in vitro. J. Craniomaxillofac. Surg. 2019, 47, 1589–1599. [Google Scholar] [CrossRef]
  69. Najem, S.; Langemann, D.; Appl, B.; Trochimiuk, M.; Hundsdoerfer, P.; Reinshagen, K.; Eschenburg, G. Smac mimetic LCL161 supports neuroblastoma chemotherapy in a drug class-dependent manner and synergistically interacts with ALK inhibitor TAE684 in cells with ALK mutation F1174L. Oncotarget 2016, 7, 72634–72653. [Google Scholar] [CrossRef] [Green Version]
  70. Ramakrishnan, V.; Gomez, M.; Prasad, V.; Kimlinger, T.; Painuly, U.; Mukhopadhyay, B.; Haug, J.; Bi, L.; Rajkumar, S.V.; Kumar, S. Smac mimetic LCL161 overcomes protective ER stress induced by obatoclax, synergistically causing cell death in multiple myeloma. Oncotarget 2016, 7, 56253–56265. [Google Scholar] [CrossRef] [Green Version]
  71. Yang, L.; Kumar, B.; Shen, C.; Zhao, S.; Blakaj, D.; Li, T.; Romito, M.; Teknos, T.N.; Williams, T.M. LCL161, a SMAC-mimetic, Preferentially Radiosensitizes Human Papillomavirus-negative Head and Neck Squamous Cell Carcinoma. Mol. Cancer Ther. 2019, 18, 1025–1035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Qin, Q.; Zuo, Y.; Yang, X.; Lu, J.; Zhan, L.; Xu, L.; Zhang, C.; Zhu, H.; Liu, J.; Liu, Z.; et al. Smac mimetic compound LCL161 sensitizes esophageal carcinoma cells to radiotherapy by inhibiting the expression of inhibitor of apoptosis protein. Tumour. Biol. 2014, 35, 2565–2574. [Google Scholar] [CrossRef] [PubMed]
  73. Hass, C.; Belz, K.; Schoeneberger, H.; Fulda, S. Sensitization of acute lymphoblastic leukemia cells for LCL161-induced cell death by targeting redox homeostasis. Biochem. Pharmacol. 2016, 105, 14–22. [Google Scholar] [CrossRef] [PubMed]
  74. Infante, J.R.; Dees, E.C.; Olszanski, A.J.; Dhuria, S.V.; Sen, S.; Cameron, S.; Cohen, R.B. Phase I dose-escalation study of LCL161, an oral inhibitor of apoptosis proteins inhibitor, in patients with advanced solid tumors. J. Clin. Oncol. 2014, 32, 3103–3110. [Google Scholar] [CrossRef]
  75. Bardia, A.; Parton, M.; Kummel, S.; Estevez, L.G.; Huang, C.S.; Cortes, J.; Ruiz-Borrego, M.; Telli, M.L.; Martin-Martorell, P.; Lopez, R.; et al. Paclitaxel With Inhibitor of Apoptosis Antagonist, LCL161, for Localized Triple-Negative Breast Cancer, Prospectively Stratified by Gene Signature in a Biomarker-Driven Neoadjuvant Trial. J. Clin. Oncol. 2018, JCO2017748392. [Google Scholar] [CrossRef] [PubMed]
  76. Condon, S.M.; Mitsuuchi, Y.; Deng, Y.; LaPorte, M.G.; Rippin, S.R.; Haimowitz, T.; Alexander, M.D.; Kumar, P.T.; Hendi, M.S.; Lee, Y.H.; et al. Birinapant, a smac-mimetic with improved tolerability for the treatment of solid tumors and hematological malignancies. J. Med. Chem. 2014, 57, 3666–3677. [Google Scholar] [CrossRef] [PubMed]
  77. Benetatos, C.A.; Mitsuuchi, Y.; Burns, J.M.; Neiman, E.M.; Condon, S.M.; Yu, G.; Seipel, M.E.; Kapoor, G.S.; Laporte, M.G.; Rippin, S.R.; et al. Birinapant (TL32711), a bivalent SMAC mimetic, targets TRAF2-associated cIAPs, abrogates TNF-induced NF-kappaB activation, and is active in patient-derived xenograft models. Mol. Cancer Ther. 2014, 13, 867–879. [Google Scholar] [CrossRef] [Green Version]
  78. Po Yee Mak, D.H.M.; Ruvolo, V.; Jacamo, R.; Steven, M.K.; Andreeff, M.; Bing, Z. Carter, Apoptosis suppressor ARC modulates SMAC mimetic-induced cell death through BIRC2/MAP3K14 signalling in acute myeloid leukaemia. Br. Kournal Haematol. 2014, 163, 9. [Google Scholar]
  79. Eytan, D.F.; Snow, G.E.; Carlson, S.G.; Schiltz, S.; Chen, Z.; Van Waes, C. Combination effects of SMAC mimetic birinapant with TNFalpha, TRAIL, and docetaxel in preclinical models of HNSCC. Laryngoscope 2015, 125, E118–E124. [Google Scholar] [CrossRef] [Green Version]
  80. Krepler, C.; Chunduru, S.K.; Halloran, M.B.; He, X.; Xiao, M.; Vultur, A.; Villanueva, J.; Mitsuuchi, Y.; Neiman, E.M.; Benetatos, C.; et al. The novel SMAC mimetic birinapant exhibits potent activity against human melanoma cells. Clin. Cancer Res. 2013, 19, 1784–1794. [Google Scholar] [CrossRef] [Green Version]
  81. Ding, J.; Qin, D.; Zhang, Y.; Li, Q.; Li, Y.; Li, J. SMAC mimetic birinapant inhibits hepatocellular carcinoma growth by activating the cIAP1/TRAF3 signaling pathway. Mol. Med. Rep. 2020, 21, 1251–1257. [Google Scholar] [CrossRef] [PubMed]
  82. Zakaria, Z.; Tivnan, A.; Flanagan, L.; Murray, D.W.; Salvucci, M.; Stringer, B.W.; Day, B.W.; Boyd, A.W.; Kogel, D.; Rehm, M.; et al. Patient-derived glioblastoma cells show significant heterogeneity in treatment responses to the inhibitor-of-apoptosis-protein antagonist birinapant. Br. J. Cancer 2016, 114, 188–198. [Google Scholar] [CrossRef] [PubMed]
  83. McComb, S.; Aguade-Gorgorio, J.; Harder, L.; Marovca, B.; Cario, G.; Eckert, C.; Schrappe, M.; Stanulla, M.; von Stackelberg, A.; Bourquin, J.P.; et al. Activation of concurrent apoptosis and necroptosis by SMAC mimetics for the treatment of refractory and relapsed ALL. Sci. Transl. Med. 2016, 8, 339ra70. [Google Scholar] [CrossRef] [PubMed]
  84. Crawford, N.; Salvucci, M.; Hellwig, C.T.; Lincoln, F.A.; Mooney, R.E.; O’Connor, C.L.; Prehn, J.H.; Longley, D.B.; Rehm, M. Simulating and predicting cellular and in vivo responses of colon cancer to combined treatment with chemotherapy and IAP antagonist Birinapant/TL32711. Cell Death Differ. 2018, 25, 1952–1966. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hamacher, R.; Schmid, R.M.; Saur, D.; Schneider, G. Apoptotic pathways in pancreatic ductal adenocarcinoma. Mol. Cancer 2008, 7, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Carter, B.Z.; Mak, P.Y.; Mak, D.H.; Shi, Y.; Qiu, Y.; Bogenberger, J.M.; Mu, H.; Tibes, R.; Yao, H.; Coombes, K.R.; et al. Synergistic targeting of AML stem/progenitor cells with IAP antagonist birinapant and demethylating agents. J. Natl. Cancer Inst. 2014, 106, djt440. [Google Scholar] [CrossRef] [Green Version]
  87. Mak, D.Y.; Fraser, I.; Ferris, R.; James, K.; Liu, M.; Thomas, S.D.; McKenzie, M.; Lefresne, S. Comparison of Rapid to Standard Volumetric Modulated Arc Therapy for Palliative Radiotherapy in Lung Cancer Patients. Cureus 2020, 12, e10055. [Google Scholar] [CrossRef]
  88. Eytan, D.F.; Snow, G.E.; Carlson, S.; Derakhshan, A.; Saleh, A.; Schiltz, S.; Cheng, H.; Mohan, S.; Cornelius, S.; Coupar, J.; et al. SMAC Mimetic Birinapant plus Radiation Eradicates Human Head and Neck Cancers with Genomic Amplifications of Cell Death Genes FADD and BIRC2. Cancer Res. 2016, 76, 5442–5454. [Google Scholar] [CrossRef] [Green Version]
  89. Brumatti, G.; Ma, C.; Lalaoui, N.; Nguyen, N.Y.; Navarro, M.; Tanzer, M.C.; Richmond, J.; Ghisi, M.; Salmon, J.M.; Silke, N.; et al. The caspase-8 inhibitor emricasan combines with the SMAC mimetic birinapant to induce necroptosis and treat acute myeloid leukemia. Sci. Transl. Med. 2016, 8, 339ra69. [Google Scholar] [CrossRef] [Green Version]
  90. Kearney, C.J.; Lalaoui, N.; Freeman, A.J.; Ramsbottom, K.M.; Silke, J.; Oliaro, J. PD-L1 and IAPs co-operate to protect tumors from cytotoxic lymphocyte-derived TNF. Cell Death Differ. 2017, 24, 1705–1716. [Google Scholar] [CrossRef] [Green Version]
  91. Zhao, L.; Yang, G.; Bai, H.; Zhang, M.; Mou, D. NCTD promotes Birinapant-mediated anticancer activity in breast cancer cells by downregulation of c-FLIP. Oncotarget 2017, 8, 26886–26895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Lalaoui, N.; Merino, D.; Giner, G.; Vaillant, F.; Chau, D.; Liu, L.; Kratina, T.; Pal, B.; Whittle, J.R.; Etemadi, N.; et al. Targeting triple-negative breast cancers with the Smac-mimetic birinapant. Cell Death Differ. 2020, 27, 2768–2780. [Google Scholar] [CrossRef] [PubMed]
  93. Orozco, C.A.; Martinez-Bosch, N.; Guerrero, P.E.; Vinaixa, J.; Dalotto-Moreno, T.; Iglesias, M.; Moreno, M.; Djurec, M.; Poirier, F.; Gabius, H.J.; et al. Targeting galectin-1 inhibits pancreatic cancer progression by modulating tumor-stroma crosstalk. Proc. Natl. Acad. Sci. USA 2018, 115, E3769–E3778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Brands, R.C.; Scheurer, M.J.J.; Hartmann, S.; Seher, A.; Kubler, A.C.; Muller-Richter, U.D.A. Apoptosis-sensitizing activity of birinapant in head and neck squamous cell carcinoma cell lines. Oncol. Lett. 2018, 15, 4010–4016. [Google Scholar] [CrossRef] [PubMed]
  95. Zhu, X.; Shen, X.; Qu, J.; Straubinger, R.M.; Jusko, W.J. Proteomic Analysis of Combined Gemcitabine and Birinapant in Pancreatic Cancer Cells. Front. Pharmacol. 2018, 9, 84. [Google Scholar] [CrossRef] [PubMed]
  96. Zhu, X.; Straubinger, R.M.; Jusko, W.J. Mechanism-based mathematical modeling of combined gemcitabine and birinapant in pancreatic cancer cells. J. Pharmacokinet. Pharmacodyn. 2015, 42, 477–496. [Google Scholar] [CrossRef]
  97. Cekay, M.J.; Roesler, S.; Frank, T.; Knuth, A.K.; Eckhardt, I.; Fulda, S. Smac mimetics and type II interferon synergistically induce necroptosis in various cancer cell lines. Cancer Lett. 2017, 410, 228–237. [Google Scholar] [CrossRef]
  98. Michie, J.; Beavis, P.A.; Freeman, A.J.; Vervoort, S.J.; Ramsbottom, K.M.; Narasimhan, V.; Lelliott, E.J.; Lalaoui, N.; Ramsay, R.G.; Johnstone, R.W.; et al. Antagonism of IAPs Enhances CAR T-cell Efficacy. Cancer Immunol. Res. 2019, 7, 183–192. [Google Scholar] [CrossRef]
  99. Amaravadi, R.K.; Schilder, R.J.; Martin, L.P.; Levin, M.; Graham, M.A.; Weng, D.E.; Adjei, A.A. A Phase I Study of the SMAC-Mimetic Birinapant in Adults with Refractory Solid Tumors or Lymphoma. Mol. Cancer Ther. 2015, 14, 2569–2575. [Google Scholar] [CrossRef] [Green Version]
  100. Senzer, N.N.; LoRusso, P.; Martin, L.P.; Schilder, R.J.; Amaravadi, R.K.; Papadopoulos, K.P.; Segota, Z.E.; Weng, D.E.; Graham, M.; Adjei, A.A. Phase II clinical activity and tolerability of the SMAC-mimetic birinapant (TL32711) plus irinotecan in irinotecan-relapsed/refractory metastatic colorectal cancer. J. Clin. Oncol. 2013, 31 (Suppl. 15), 3621. [Google Scholar] [CrossRef]
  101. Hu, R.; Li, J.; Liu, Z.; Miao, M.; Yao, K. GDC-0152 induces apoptosis through down-regulation of IAPs in human leukemia cells and inhibition of PI3K/Akt signaling pathway. Tumour Biol. 2015, 36, 577–584. [Google Scholar] [CrossRef] [PubMed]
  102. Yang, L.; Shu, T.; Liang, Y.; Gu, W.; Wang, C.; Song, X.; Fan, C.; Wang, W. GDC-0152 attenuates the malignant progression of osteosarcoma promoted by ANGPTL2 via PI3K/AKT but not p38MAPK signaling pathway. Int. J. Oncol. 2015, 46, 1651–1658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Shekhar, T.M.; Burvenich, I.J.G.; Harris, M.A.; Rigopoulos, A.; Zanker, D.; Spurling, A.; Parker, B.S.; Walkley, C.R.; Scott, A.M.; Hawkins, C.J. Smac mimetics LCL161 and GDC-0152 inhibit osteosarcoma growth and metastasis in mice. BMC Cancer 2019, 19, 924. [Google Scholar] [CrossRef] [PubMed]
  104. Tchoghandjian, A.; Souberan, A.; Tabouret, E.; Colin, C.; Denicolai, E.; Jiguet-Jiglaire, C.; El-Battari, A.; Villard, C.; Baeza-Kallee, N.; Figarella-Branger, D. Inhibitor of apoptosis protein expression in glioblastomas and their in vitro and in vivo targeting by SMAC mimetic GDC-0152. Cell Death Dis. 2016, 7, e2325. [Google Scholar] [CrossRef] [Green Version]
  105. Mantik, P.; Xie, M.; Wong, H.; La, H.; Steigerwalt, R.W.; Devanaboyina, U.; Ganem, G.; Shih, D.; Flygare, J.A.; Fairbrother, W.J.; et al. Cyclodextrin Reduces Intravenous Toxicity of a Model Compound. J. Pharm. Sci. 2019, 108, 1934–1943. [Google Scholar] [CrossRef] [PubMed]
  106. Shin, Y.G.; Jones, S.A.; Murakami, S.C.; Budha, N.; Ware, J.; Wong, H.; Buonarati, M.H.; Dean, B.; Hop, C.E. Validation and application of a liquid chromatography-tandem mass spectrometric method for the determination of GDC-0152 in human plasma using solid-phase extraction. Biomed. Chromatogr. 2013, 27, 102–110. [Google Scholar] [CrossRef]
  107. Nikkhoo, A.; Rostami, N.; Farhadi, S.; Esmaily, M.; Moghadaszadeh Ardebili, S.; Atyabi, F.; Baghaei, M.; Haghnavaz, N.; Yousefi, M.; Aliparasti, M.R.; et al. Codelivery of STAT3 siRNA and BV6 by carboxymethyl dextran trimethyl chitosan nanoparticles suppresses cancer cell progression. Int. J. Pharm. 2020, 581, 119236. [Google Scholar] [CrossRef]
Figure 1. Smac/DIABLO inhibits IAPs in cancer cells. The precursor of Smac (yellow) is transported to the intermembrane space of mitochondria by the import signal (blue box). Then, mature Smac is released from the mitochondria into the cytosol and subsequently inhibits the cellular functions of IAPs. AVPI stands for “Ala-Val-Pro-Ile”. BIR stands for “baculoviral IAP repeat”. Ub stands for “ubiquitin”.
Figure 1. Smac/DIABLO inhibits IAPs in cancer cells. The precursor of Smac (yellow) is transported to the intermembrane space of mitochondria by the import signal (blue box). Then, mature Smac is released from the mitochondria into the cytosol and subsequently inhibits the cellular functions of IAPs. AVPI stands for “Ala-Val-Pro-Ile”. BIR stands for “baculoviral IAP repeat”. Ub stands for “ubiquitin”.
Applsci 11 00335 g001
Figure 2. Chemical structure of Smac mimetics for cancer treatment. BV6 was modified with trifluoroacetic acid (xCF3COOH) to improve the solubility of BV6 to DMSO and water.
Figure 2. Chemical structure of Smac mimetics for cancer treatment. BV6 was modified with trifluoroacetic acid (xCF3COOH) to improve the solubility of BV6 to DMSO and water.
Applsci 11 00335 g002
Figure 3. Anti-cancer effects and the molecular mechanisms of action of LCL161 (black) and birinapant (blue) in cancer cells.
Figure 3. Anti-cancer effects and the molecular mechanisms of action of LCL161 (black) and birinapant (blue) in cancer cells.
Applsci 11 00335 g003
Table 1. Clinical status of LCL161 for cancer treatment.
Table 1. Clinical status of LCL161 for cancer treatment.
ClinicalTrials.Gov IdentifierPhaseCondition or Disease (in Patients)Combination Therapy
NCT01968915Phase 1
(completed)
Neoplasms-
NCT01955434Phase 2
(completed)
Recurrent plasma cell myeloma
Refractory plasma cell myeloma
Cyclophophamide
NCT01934634Phase 1Metastatic pancreatic cancerGemcitabine
Nab-Paclitaxel
NCT02098161Phase 2Polycythemia vera, post-polycythemic myelofibrosis phase
Primary myelofibrosis
Secondary myelofibrosis
-
NCT02649673Phase 1/2Small cell lung cancer
Ovarian cancer
Topotecan
Pegylated granulocyte colony stimulating factor
NCT01617668Phase 2
(completed)
Breast cancerPaclitaxel
NCT01240655Phase 1
(completed)
Solid tumorsPaclitaxel
NCT01098838Phase 1
(completed)
Advanced solid tumors (lung, skin, colon, pancreas, and others)-
NCT03111992Phase 1
(completed)
Multiple myelomaPDR001
CJM112
NCT02890069Phase 1Colorectal cancer
Non-small cell lung carcinoma (Adenocarcinoma)
Triple-negative breast cancer
Renal cell carcinoma
PDR001
Evrolimus
Panobinostat
QBM076
HDM201
Table 2. Clinical status of birinapant (TL32711) for cancer treatment.
Table 2. Clinical status of birinapant (TL32711) for cancer treatment.
ClinicalTrials.Gov IdentifierPhaseCondition or Disease (in Patients)Combination Therapy
NCT03803774Phase 1Head and neck squamous cell carcinomaRadiation therapy
NCT02587962Phase 1/2
(terminated)
Solid tumorPembrolizumab
NCT02147873Phase 2
(terminated)
Myelodysplastic syndrome
Chronic myelomonocytic leukemia
Azacitidine
Placebo
NCT01940172Phase 1
(completed)
Relapsed epithelial ovarian cancer
Relapsed primary peritoneal cancer
Relapsed fallopian tube cancer
Conatumumab
NCT01188499Phase 1/2
(completed)
CancerCarboplatin
Paclitaxel
Irinotecan
Docetaxel
Gemcitabine
Liposomal Doxorubicin
NCT01681368Phase 2
(terminated)
Epithelial ovarian cancer
Peritoneal neoplasms
Fallopian tube neoplasms
-
NCT00993239Phase 1
(completed)
Cancer-
NCT01573780Phase 1
(terminated)
Adult solid tumorGemcitabine
NCT01486784Phase 1/2
(terminated)
Acute myelogenous leukemia-
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chang, Y.-C.; Cheung, C.H.A. An Updated Review of Smac Mimetics, LCL161, Birinapant, and GDC-0152 in Cancer Treatment. Appl. Sci. 2021, 11, 335. https://doi.org/10.3390/app11010335

AMA Style

Chang Y-C, Cheung CHA. An Updated Review of Smac Mimetics, LCL161, Birinapant, and GDC-0152 in Cancer Treatment. Applied Sciences. 2021; 11(1):335. https://doi.org/10.3390/app11010335

Chicago/Turabian Style

Chang, Yung-Chieh, and Chun Hei Antonio Cheung. 2021. "An Updated Review of Smac Mimetics, LCL161, Birinapant, and GDC-0152 in Cancer Treatment" Applied Sciences 11, no. 1: 335. https://doi.org/10.3390/app11010335

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop