Need Assistance?
  • US & Canada:
    +
  • UK: +

BV6

* Please kindly note that our products are not to be used for therapeutic purposes and cannot be sold to patients.

BV-6 is a selective inhibitor of apoptosis (IAP) family with IC50 value of 7.2 μM when tested with H460 cells. It acts by promoting glioblastoma cancer stem-like cell differentiation by activating NF-κB. BV-6 induces autoubiquitination and proteasomal degradation of IAP1 and XIAP in MDA-MB-231 cells at 5 μM. In BALB/c mice model with transplanted abdominal cavities from donor mouse uterine tissue, intraperitoneally with BV-6 repressed the advancement of endometriosis, cell proliferative activity via inhibiting the expression of IAPs. In hematological THP-1 cells, pre-treatment with BV-6 increased the CIK cells killing ability and the same results were achieved in solid malignancy RH30 cells.

Category
Peptide Inhibitors
Catalog number
BAT-010145
CAS number
1001600-56-1
Molecular Formula
C70H96N10O8
Molecular Weight
1205.57
BV6
IUPAC Name
(2S)-1-[(2S)-2-cyclohexyl-2-[[(2S)-2-(methylamino)propanoyl]amino]acetyl]-N-[(2S)-1-[6-[[(2S)-2-[[(2S)-1-[(2S)-2-cyclohexyl-2-[[(2S)-2-(methylamino)propanoyl]amino]acetyl]pyrrolidine-2-carbonyl]amino]-3,3-diphenylpropanoyl]amino]hexylamino]-1-oxo-3,3-diphenylpropan-2-yl]pyrrolidine-2-carboxamide
Appearance
Solid Powder
Purity
≥98%
Density
1.177±0.06 g/cm3(Predicted)
Boiling Point
1376.7±65.0°C(Predicted)
Storage
Store at -20°C
Solubility
Soluble to 100 mg/mL in DMSO, to 25 mg/mL in water.
InChI
InChI=1S/C70H96N10O8/c1-47(71-3)63(81)75-59(53-37-21-11-22-38-53)69(87)79-45-27-41-55(79)65(83)77-61(57(49-29-13-7-14-30-49)50-31-15-8-16-32-50)67(85)73-43-25-5-6-26-44-74-68(86)62(58(51-33-17-9-18-34-51)52-35-19-10-20-36-52)78-66(84)56-42-28-46-80(56)70(88)60(54-39-23-12-24-40-54)76-64(82)48(2)72-4/h7-10,13-20,29-36,47-48,53-62,71-72H,5-6,11-12,21-28,37-46H2,1-4H3,(H,73,85)(H,74,86)(H,75,81)(H,76,82)(H,77,83)(H,78,84)/t47-,48-,55-,56-,59-,60-,61-,62-/m0/s1
InChI Key
DPXJXGNXKOVBJV-YLOPQIBLSA-N
Canonical SMILES
O=C(N[C@@H](C(C1=CC=CC=C1)C2=CC=CC=C2)C(NCCCCCCNC([C@H](C(C3=CC=CC=C3)C4=CC=CC=C4)NC([C@@H]5CCCN5C([C@H](C6CCCCC6)NC([C@@H](NC)C)=O)=O)=O)=O)=O)[C@@H]7CCCN7C([C@H](C8CCCCC8)NC([C@@H](NC)C)=O)=O
1.Smac mimetic sensitizes renal cell carcinoma cells to interferon-α-induced apoptosis.
Reiter M1, Eckhardt I2, Haferkamp A3, Fulda S4. Cancer Lett. 2016 May 28;375(1):1-8. doi: 10.1016/j.canlet.2016.02.019. Epub 2016 Feb 18.
The prognosis of metastatic or relapsed renal cell carcinoma (RCC) is still very poor, highlighting the need for new treatment strategies. Here, we identify a cooperative antitumor activity of interferon-α (IFNα) together with the Smac mimetic BV6 that antagonizes antiapoptotic IAP proteins. BV6 and IFNα act together to reduce cell viability and to induce apoptosis in various RCC cell lines. Molecular studies revealed that BV6/IFNα co-treatment triggers apoptosis independently of autocrine/paracrine Tumor Necrosis Factor (TNF)α signaling, since the TNFα-blocking antibody Enbrel fails to rescue cell death. Importantly, knockdown of Receptor-Interacting Protein (RIP)1 significantly decreases BV6/IFNα-mediated apoptosis, whereas the RIP1 kinase inhibitor necrostatin-1 (Nec-1) provides no protection. This demonstrates that RIP1 protein is critically required for BV6/IFNα-induced apoptosis, while RIP1 kinase activity is dispensable, pointing to a scaffold function of RIP1.
2.Smac Mimetic-Induced Upregulation of CCL2/MCP-1 Triggers Migration and Invasion of Glioblastoma Cells and Influences the Tumor Microenvironment in a Paracrine Manner.
Lindemann C1, Marschall V1, Weigert A2, Klingebiel T3, Fulda S4. Neoplasia. 2015 Jun;17(6):481-9. doi: 10.1016/j.neo.2015.05.002.
Second mitochondria-derived activator of caspase (Smac) mimetics are considered as promising anticancer therapeutics that are currently under investigation in early clinical trials. They induce apoptosis by antagonizing inhibitor of apoptosis proteins, which are frequently overexpressed in cancer. We previously reported that Smac mimetics, such as BV6, additionally exert non-apoptotic functions in glioblastoma (GBM) cells by stimulating migration and invasion in a nuclear factor kappa B (NF-κB)-dependent manner. Because NF-κB target genes mediating these effects are largely unknown, we performed whole-genome expression analyses. Here, we identify chemokine (C-C motif) ligand 2 (CCL2) as the top-listed NF-κB-regulated gene being upregulated upon BV6 treatment in GBM cells. BV6-induced upregulation and secretion of CCL2 are required for migration and invasion of GBM cells because knockdown of CCL2 in GBM cells abolishes these effects. Co-culture experiments of GBM cells with non-malignant astroglial cells reveal that BV6-stimulated secretion of CCL2 by GBM cells into the supernatant triggers migration of astroglial cells toward GBM cells because CCL2 knockdown in BV6-treated GBM cells impedes BV6-stimulated migration of astroglial cells.
3.Intrinsic and chemo-sensitizing activity of SMAC-mimetics on high-risk childhood acute lymphoblastic leukemia.
Schirmer M1, Trentin L1, Queudeville M1, Seyfried F1, Demir S1, Tausch E2, Stilgenbauer S2, Eckhoff SM1, Meyer LH1, Debatin KM1. Cell Death Dis. 2016 Jan 14;7:e2052. doi: 10.1038/cddis.2015.382.
SMAC-mimetics represent a targeted therapy approach to overcome apoptosis resistance in many tumors. Here, we investigated the efficacy of the SMAC-mimetic BV6 in B-cell precursor acute lymphoblastic leukemia (BCP-ALL). In ALL cell lines, intrinsic apoptosis sensitivity was associated with rapid cIAP degradation, NF-κB activation, TNF-α secretion and induction of an autocrine TNF-α-dependent cell death loop. This pattern of responsiveness was also observed upon ex vivo analysis of 40 primograft BCP-ALL samples. Treatment with BV6 induced cell death in the majority of ALL primografts including leukemias with high-risk and poor-prognosis features. Inhibition of cell death by the TNF receptor fusion protein etanercept demonstrated that BV6 activity is dependent on TNF-α. In a preclinical NOD/SCID/huALL model of high-risk ALL, marked anti-leukemia effectivity and significantly prolonged survival were observed upon BV6 treatment. Interestingly, also in vivo, intrinsic SMAC-mimetic activity was mediated by TNF-α.
4.NF-κB functions as a molecular link between tumor cells and Th1/Tc1 T cells in the tumor microenvironment to exert radiation-mediated tumor suppression.
Simon PS1,2,3, Bardhan K1, Chen MR1, Paschall AV1,2,3, Lu C1,3, Bollag RJ2, Kong FC4,2, Jin J4,2, Kong FM4,2, Waller JL5, Pollock RE6, Liu K1,2,3. Oncotarget. 2016 Mar 21. doi: 10.18632/oncotarget.8246. [Epub ahead of print]
Radiation modulates both tumor cells and immune cells in the tumor microenvironment to exert its anti-tumor activity; however, the molecular connection between tumor cells and immune cells that mediates radiation-exerted tumor suppression activity in the tumor microenvironment is largely unknown. We report here that radiation induces rapid activation of the p65/p50 and p50/p50 NF-κB complexes in human soft tissue sarcoma (STS) cells. Radiation-activated p65/p50 and p50/p50 bind to the TNFα promoter to activate its transcription in STS cells. Radiation-induced TNFα induces tumor cell death in an autocrine manner. A sublethal dose of Smac mimetic BV6 induces cIAP1 and cIAP2 degradation to increase tumor cell sensitivity to radiation-induced cell death in vitro and to enhance radiation-mediated suppression of STS xenografts in vivo. Inhibition of caspases, RIP1, or RIP3 blocks radiation/TNFα-induced cell death, whereas inhibition of RIP1 blocks TNFα-induced caspase activation, suggesting that caspases and RIP1 act sequentially to mediate the non-compensatory cell death pathways.
Online Inquiry
Verification code
Inquiry Basket