Home>>Signaling Pathways>> Apoptosis>> Other Apoptosis>>Birinapant (TL32711)

Birinapant (TL32711) Sale

(Synonyms: 比瑞那帕; TL32711) 目录号 : GC12426

An antagonist of cIAP1, cIAP2, and XIAP

Birinapant (TL32711) Chemical Structure

Cas No.:1260251-31-7

规格 价格 库存 购买数量
10mM (in 1mL DMSO)
¥945.00
现货
5mg
¥651.00
现货
10mg
¥1,040.00
现货
25mg
¥1,649.00
现货
50mg
¥2,373.00
现货

电话:400-920-5774 Email: sales@glpbio.cn

Customer Reviews

Based on customer reviews.

Sample solution is provided at 25 µL, 10mM.

产品文档

Quality Control & SDS

View current batch:

实验参考方法

IAP binding affinity of Birinapant (TL32711)[1]:

Preparation Method

A fluorescence polarization assay was used to determine the dissociation constants (K d) of Smac mimetics Birinapant (TL32711) and GT13402 for XIAP and cIAP1 by monitoring the decrease in fluorescence polarization signal due to competitive displacement of an FP peptide from the BIR3/FP peptide binary complex.

Applications

The binding constants (K d) of Birinapant (TL32711) for XIAP and cIAP1 were determined to be 45 and

Cell experiment [2]:

Cell lines

451Lu and WM1366 melanoma cells

Preparation Method

Cells were treated with Birinapant (TL32711) 1 μmol/L in combination with TNF-α 1 ng/mL. Cells were then incubated for 72 hours in the presence or absence of Necrostatin-1 a RIP1 kinase inhibitor. Proliferation was assessed using the MTS assay.

Reaction Conditions

1 μmol/L Birinapant (TL32711) for 72 hours

Applications

The majority of cell lines exhibited strong combination activity of Birinapant (TL32711) and TNF-α.

Animal experiment [3]:

Animal models

Male C57BL/6J mice aged 8 week old, weighing 21 ± 1.3g

Preparation Method

Before the LPS administration, mice were injected intraperitoneally with Birinapant (TL32711) (30 mg/kg body weight, Birinapant (TL32711) group) either vehicle control (vehicle group) for 24 h. Birinapant (TL32711) was dissolved in 12.5% Captisol in distilled water. Twenty-four mice in each group were euthanized and the samples of the liver and blood were harvested at 0, 6, 12 and 24 h after LPS challleage.

Dosage form

30 mg/kg Birinapant (TL32711) ip. for 24 h.

Applications

Birinapant (TL32711) pretreatment significantly improved mice survival.

References:

[1]. Allensworth JL, Sauer SJ, et,al. Smac mimetic Birinapant induces apoptosis and enhances TRAIL potency in inflammatory breast cancer cells in an IAP-dependent and TNF-α-independent mechanism. Breast Cancer Res Treat. 2013 Jan;137(2):359-71. doi: 10.1007/s10549-012-2352-6. Epub 2012 Dec 7. PMID: 23225169.

[2]. Krepler C, Chunduru SK, et,al. The novel SMAC mimetic birinapant exhibits potent activity against human melanoma cells. Clin Cancer Res. 2013 Apr 1;19(7):1784-94. doi: 10.1158/1078-0432.CCR-12-2518. Epub 2013 Feb 12. PMID: 23403634; PMCID: PMC3618495.

[3]. Liu H, Liao R, et,al. 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. 2017 May;185:79-83. doi: 10.1016/j.imlet.2017.02.016. Epub 2017 Mar 9. PMID: 28286228.

产品描述

Birinapant (TL32711) is a SMAC mimetic designed to specifically target cIAP1 and cIAP2 for degradation[1]. the binding constants (K d) of Birinapant (TL32711) for XIAP and cIAP1 were determined to be 45 and[3].

When Birinapant (TL32711) was combined with TNF-α, strong combination activity, that is, neither compound was effective individually but the combination was highly effective, was observed in 12 of 18 cell lines.Birinapant (TL32711) combined with TNF-α inhibited the growth of a melanoma cell line with acquired resistance to BRAF inhibition to the same extent as in the parental cell line[1]. When investigated the role of Birinapant (TL32711) in radiosensitization of glioblastoma cells, Birinapant (TL32711) can enhance the radiosensitivity of glioblastoma cell lines in cell-based assays and tumor models via radiation-induced TNF-α[4]. Combination of Birinapant (TL32711) and TNFα induced sub-G0 DNA fragmentation in sensitive lines and Birinapant (TL32711) alone also induced significant G2-M cell-cycle arrest and cell death in UM-SCC-46 cells[6]. Birinapant (TL32711) induced death receptor-/caspase-8-mediated apoptosis in AML cells, including in AML stem/progenitor cells, but not in normal CD34(+) cells[7].

Birinapant (TL32711) significantly improved the survival rate of endotoxemic mice and attenuated LPS-induced liver pathologic damage and inflammatory response. IL-1 and TNF-α levels in the serum were markedly decreased in Birinapant (TL32711) pretreatment mice compared with control mice .The cellular inhibitor of apoptosis protein 1 (cIAP1) expression in liver resident macrophage (Kupffer cells, KCs) was significantly decreased in the Birinapant (TL32711) group compared to the Vehicle group[2].

References:
[1]. Krepler C, Chunduru SK, et,al. The novel SMAC mimetic birinapant exhibits potent activity against human melanoma cells. Clin Cancer Res. 2013 Apr 1;19(7):1784-94. doi: 10.1158/1078-0432.CCR-12-2518. Epub 2013 Feb 12. PMID: 23403634; PMCID: PMC3618495.
[2]. Liu H, Liao R, et,al. 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. 2017 May;185:79-83. doi: 10.1016/j.imlet.2017.02.016. Epub 2017 Mar 9. PMID: 28286228.
[3]. Allensworth JL, Sauer SJ, et,al. Smac mimetic Birinapant induces apoptosis and enhances TRAIL potency in inflammatory breast cancer cells in an IAP-dependent and TNF-α-independent mechanism. Breast Cancer Res Treat. 2013 Jan;137(2):359-71. doi: 10.1007/s10549-012-2352-6. Epub 2012 Dec 7. PMID: 23225169.
[4]. Cerna D, Lim B, A et,al.SMAC Mimetic/IAP Inhibitor Birinapant Enhances Radiosensitivity of Glioblastoma Multiforme. Radiat Res. 2021 Jun 1;195(6):549-560. doi: 10.1667/RADE-20-00171.1. PMID: 33826739.
[5]. Zhu DL, Shuai LY. Effects of Birinapant on Proliferation and Invasion of MGC-803 Gastric Cancer Cells and Mechanism Underlying These Effects. Bull Exp Biol Med. 2021 May;171(1):56-61. doi: 10.1007/s10517-021-05172-w. Epub 2021 May 29. PMID: 34050412.
[6]. Eytan DF, Snow GE, 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 Sep 15;76(18):5442-5454. doi: 10.1158/0008-5472.CAN-15-3317. Epub 2016 Jul 28. PMID: 27469115; PMCID: PMC5026594.
[7]. Carter BZ, Mak PY, et,al. Synergistic targeting of AML stem/progenitor cells with IAP antagonist birinapant and demethylating agents. J Natl Cancer Inst. 2014 Feb;106(2):djt440. doi: 10.1093/jnci/djt440. PMID: 24526787; PMCID: PMC3952202.

Birinapant (TL32711) 是一种 SMAC 模拟物,旨在专门针对 cIAP1 和 cIAP2 进行降解[1]。 Birinapant (TL32711) 对 XIAP 和 cIAP1 的结合常数 (K d) 被确定为 45 和[3]

当 Birinapant (TL32711) 与 TNF-α 联合使用时,在 18 种细胞系中的 12 种细胞系中观察到很强的联合活性,即两种化合物单独使用均无效,但联合使用非常有效。Birinapant (TL32711) 与 TNF 联合使用-α 对 BRAF 抑制具有获得性抗性的黑色素瘤细胞系的生长的抑制程度与亲本细胞系相同[1]。当研究 Birinapant (TL32711) 在胶质母细胞瘤细胞放射增敏中的作用时,Birinapant (TL32711) 可以通过辐射诱导的 TNF-α 增强胶质母细胞瘤细胞系在基于细胞的检测和肿瘤模型中的放射敏感性[4]。 Birinapant (TL32711) 和 TNFα 的组合在敏感细胞系中诱导亚 G0 DNA 片段化,单独使用 Birinapant (TL32711) 也在 UM-SCC-46 细胞中诱导显着的 G2-M 细胞周期停滞和细胞死亡 [6]。 Birinapant (TL32711) 在 AML 细胞(包括 AML 干细胞/祖细胞)中诱导死亡受体/caspase-8 介导的细胞凋亡,但在正常 CD34(+) 细胞中没有这种作用[7]

\n

Birinapant (TL32711) 显着提高了内毒素血症小鼠的存活率,并减轻了 LPS 诱导的肝脏病理损伤和炎症反应。与对照小鼠相比,Birinapant (TL32711) 预处理小鼠血清中的 IL-1 和 TNF-α 水平显着降低。肝驻留巨噬细胞(Kupffer 细胞,KCs)中细胞凋亡蛋白 1 (cIAP1) 的表达显着降低Birinapant (TL32711) 组与载体组的比较[2]

Chemical Properties

Cas No. 1260251-31-7 SDF
别名 比瑞那帕; TL32711
化学名 (2S,2'S)-N,N'-((2S,2'S)-((3S,3'S,5R,5'R)-5,5'-((6,6'-difluoro-1H,1'H-[2,2'-biindole]-3,3'-diyl)bis(methylene))bis(3-hydroxypyrrolidine-5,1-diyl))bis(1-oxobutane-2,1-diyl))bis(2-(methylamino)propanamide)
Canonical SMILES CCC(C(=O)N1CC(CC1CC2=C(NC3=C2C=CC(=C3)F)C4=C(C5=C(N4)C=C(C=C5)F)CC6CC(CN6C(=O)C(CC)NC(=O)C(C)NC)O)O)NC(=O)C(C)NC
分子式 C42H56F2N8O6 分子量 806.94
溶解度 ≥ 40.35 mg/mL in DMSO, ≥ 46.9 mg/mL in EtOH 储存条件 Store at -20°C
General tips 请根据产品在不同溶剂中的溶解度选择合适的溶剂配制储备液;一旦配成溶液,请分装保存,避免反复冻融造成的产品失效。
储备液的保存方式和期限:-80°C 储存时,请在 6 个月内使用,-20°C 储存时,请在 1 个月内使用。
为了提高溶解度,请将管子加热至37℃,然后在超声波浴中震荡一段时间。
Shipping Condition 评估样品解决方案:配备蓝冰进行发货。所有其他可用尺寸:配备RT,或根据请求配备蓝冰。

溶解性数据

制备储备液
1 mg 5 mg 10 mg
1 mM 1.2392 mL 6.1962 mL 12.3925 mL
5 mM 0.2478 mL 1.2392 mL 2.4785 mL
10 mM 0.1239 mL 0.6196 mL 1.2392 mL
  • 摩尔浓度计算器

  • 稀释计算器

  • 分子量计算器

质量
=
浓度
x
体积
x
分子量
 
 
 
*在配置溶液时,请务必参考产品标签上、MSDS / COA(可在Glpbio的产品页面获得)批次特异的分子量使用本工具。

计算

动物体内配方计算器 (澄清溶液)

第一步:请输入基本实验信息(考虑到实验过程中的损耗,建议多配一只动物的药量)
给药剂量 mg/kg 动物平均体重 g 每只动物给药体积 ul 动物数量
第二步:请输入动物体内配方组成(配方适用于不溶于水的药物;不同批次药物配方比例不同,请联系GLPBIO为您提供正确的澄清溶液配方)
% DMSO % % Tween 80 % saline
计算重置

Research Update

Therapeutically Increasing MHC-I Expression Potentiates Immune Checkpoint Blockade

Immune checkpoint blockade (ICB) therapy revolutionized cancer treatment, but many patients with impaired MHC-I expression remain refractory. Here, we combined FACS-based genome-wide CRISPR screens with a data-mining approach to identify drugs that can upregulate MHC-I without inducing PD-L1. CRISPR screening identified TRAF3, a suppressor of the NFκB pathway, as a negative regulator of MHC-I but not PD-L1. The Traf3-knockout gene expression signature is associated with better survival in ICB-na?ve patients with cancer and better ICB response. We then screened for drugs with similar transcriptional effects as this signature and identified Second Mitochondria-derived Activator of Caspase (SMAC) mimetics. We experimentally validated that the SMAC mimetic birinapant upregulates MHC-I, sensitizes cancer cells to T cell-dependent killing, and adds to ICB efficacy. Our findings provide preclinical rationale for treating tumors expressing low MHC-I expression with SMAC mimetics to enhance sensitivity to immunotherapy. The approach used in this study can be generalized to identify other drugs that enhance immunotherapy efficacy. SIGNIFICANCE: MHC-I loss or downregulation in cancer cells is a major mechanism of resistance to T cell-based immunotherapies. Our study reveals that birinapant may be used for patients with low baseline MHC-I to enhance ICB response. This represents promising immunotherapy opportunities given the biosafety profile of birinapant from multiple clinical trials.This article is highlighted in the In This Issue feature, p. 1307.

Birinapant Enhances Gemcitabine's Antitumor Efficacy in Triple-Negative Breast Cancer by Inducing Intrinsic Pathway-Dependent Apoptosis

Triple-negative breast cancer (TNBC) is the most aggressive subgroup of breast cancer, and patients with TNBC have few therapeutic options. Apoptosis resistance is a hallmark of human cancer, and apoptosis regulators have been targeted for drug development for cancer treatment. One class of apoptosis regulators is the inhibitors of apoptosis proteins (IAPs). Dysregulated IAP expression has been reported in many cancers, including breast cancer, and has been shown to be responsible for resistance to chemotherapy. Therefore, IAPs have become attractive molecular targets for cancer treatment. Here, we first investigated the antitumor efficacy of birinapant (TL32711), a biindole-based bivalent mimetic of second mitochondria-derived activator of caspases (SMACs), in TNBC. We found that birinapant as a single agent has differential antiproliferation effects in TNBC cells. We next assessed whether birinapant has a synergistic effect with commonly used anticancer drugs, including entinostat (class I histone deacetylase inhibitor), cisplatin, paclitaxel, voxtalisib (PI3K inhibitor), dasatinib (Src inhibitor), erlotinib (EGFR inhibitor), and gemcitabine, in TNBC. Among these tested drugs, gemcitabine showed a strong synergistic effect with birinapant. Birinapant significantly enhanced the antitumor activity of gemcitabine in TNBC both in vitro and in xenograft mouse models through activation of the intrinsic apoptosis pathway via degradation of cIAP2 and XIAP, leading to apoptotic cell death. Our findings demonstrate the therapeutic potential of birinapant to enhance the antitumor efficacy of gemcitabine in TNBC by targeting the IAP family of proteins.

Retraction Note: Birinapant sensitizes platinum-resistant carcinomas with high levels of cIAP to carboplatin therapy

The IAP antagonist birinapant potentiates bortezomib anti-myeloma activity in vitro and in vivo

Background: Mechanisms by which Smac mimetics (SMs) interact with proteasome inhibitors (e.g., bortezomib) are largely unknown, particularly in multiple myeloma (MM), a disease in which bortezomib represents a mainstay of therapy.
Methods: Interactions between the clinically relevant IAP (inhibitor of apoptosis protein) antagonist birinapant (TL32711) and the proteasome inhibitor bortezomib were investigated in multiple myeloma (MM) cell lines and primary cells, as well as in vivo models. Induction of apoptosis and changes in gene and protein expression were monitored using MM cell lines and confirmed in primary MM cell populations. Genetically modified cells (e.g., exhibiting shRNA knockdown or ectopic expression) were employed to evaluate the functional significance of birinapant/bortezomib-induced changes in protein levels. A MM xenograft model was used to evaluate the in vivo activity of the birinapant/bortezomib regimen.
Results: Birinapant and bortezomib synergistically induced apoptosis in diverse cell lines, including bortezomib-resistant cells (PS-R). The regimen robustly downregulated cIAP1/2 but not the canonical NF-κB pathway, reflected by p65 phosphorylation and nuclear accumulation. In contrast, the bortezomib/birinapant regimen upregulated TRAF3, downregulated TRAF2, and diminished p52 processing and BCL-XL expression, consistent with disruption of the non-canonical NF-κB pathway. TRAF3 knockdown, ectopic TRAF2, or BCL-XL expression significantly diminished birinapant/bortezomib toxicity. The regimen sharply increased extrinsic apoptotic pathway activation, and cells expressing dominant-negative FADD or caspase-8 displayed markedly reduced birinapant/bortezomib sensitivity. Primary CD138+ (n = 43) and primitive MM populations (CD138-/19+/20+/27+; n = 31) but not normal CD34+ cells exhibited significantly enhanced toxicity with combined treatment (P < 0.0001). The regimen was also fully active in the presence of HS-5 stromal cells or growth factors (e.g., IL-6 and VEGF). Finally, the regimen was well tolerated and significantly increased survival (P < 0.05 and P < 0.001) compared to single agents in a MM xenograft model. Combined treatment also downregulated cIAP1/2 and p52 while increasing PARP cleavage in MM cells in vivo.
Conclusions: Our data suggest that birinapant and bortezomib interact synergistically in MM cells, including those resistant to bortezomib, through inactivation of the non-canonical NF-κB and activation of the extrinsic apoptotic pathway both in vitro and in vivo. They also argue that a strategy combining cIAP antagonists and proteasome inhibitors warrants attention in MM.

Birinapant (TL32711) Improves Responses to GEM/AZD7762 Combination Therapy in Triple-negative Breast Cancer Cell Lines

Background: Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer currently lacking targeted therapies. Our previous work demonstrated a therapeutic synergism with gemcitabine (GEM) and the CHK1 inhibitor (AZD7762) combination treatment in a TNBC cell line. We hypothesized that the response to this combination therapy would differ among heterogeneous TNBC patients and that addition of a SMAC mimetic (TL32711) could improve efficacy.
Materials and methods: Therapeutic responses to GEM, GEM/AZD7762, and GEM/AZD7762/TL32711 combinations were investigated by XTT assays and western blotting of cell cycle and apoptosis-related proteins in ten TNBC cell lines.
Results: TNBC cell lines harboring low levels of endogenous CHK1, cIAP1 and cIAP2 were responsive to GEM alone, whereas cell lines demonstrating a minimal increase in phospho-S345 CHK1 after treatment were responsive to GEM/AZD7762 or GEM/AZD7762/TL32711 combination.
Conclusion: The response of TNBC cells to particular therapies varies and will require development of predictive biomarkers.