Size | Price | Stock | Qty |
---|---|---|---|
1mg |
|
||
5mg |
|
||
10mg |
|
||
25mg |
|
||
50mg |
|
||
100mg |
|
||
Other Sizes |
|
Purity: ≥98%
BMS-8 is a novel small molecule inhibitor of the PD-1 (Programmed death-1)/PD-L1 (Programmed death-ligand 1) protein/protein interaction with potential anticancer activities. BMS-8 inhibits PD-1/PD-L1 interaction with an IC50 of 146 nM, and may augment therapeutic immune response to various histologically distinct tumors. Blockade of the PD-1/PD-Ll ligation using antibodies to PD-Ll has been shown to restore and augment T cell activation in many system. Structural basis for blocking the PD-1/PD-L1 interaction by small molecules is illustrated with the compound BMS-202 that binds to a hydrophobic cavity and induces dimerization of PD-L1.
Targets |
PD-1/PD-L1 PPI; PD-L1 protein
|
---|---|
ln Vitro |
One of the PD-L1 monomers tends to have a more stable binding mode with BMS-8 than the other, and the small-molecule inducing PD-L1 dimerization was further stabilized by the non-polar interaction of Ile54, Tyr56, Met115, Ala121, and Tyr123 on both monomers and the water bridges involved in ALys124[2].
|
ln Vivo |
In Vivo Antitumor Activity of NP19 [[an analog of BMS-8]] in an H22 Hepatoma Mouse Model[3]
Encouraged by the excellent in vivo antitumor efficacy of NP19 on the melanoma B16-F10 tumor model, and the fact that PD-1/PD-L1 inhibitors have broad spectrum of antitumor activities, we further evaluated the in vivo antitumor efficacy of compound NP19 using an H22 hepatoma tumor model in BALB/c mice. Each mouse was injected with 0.8 million H22 cells subcutaneously into the right flank. After tumors reached approximately 100 mm3 in volume, mice were randomized and treated by intraperitoneal (i.p.) injection of NP19 or a vehicle solution for 14 days. As shown in Figure 8, NP19 demonstrated significant in vivo antitumor efficacy with a TGI of 76.5% at the dose of 25 mg/kg (Figure 8A, 8B, 8C). In addition, NP19 did not cause an obvious body weight loss (Figure 8D), indicating that the compound was well tolerated. In Vivo Antitumor Activity of NP19 [an analog of BMS-8] in a B16-F10 Mouse Melanoma Model[3] To determine whether the in vitro anti-PD-1/PD-L1 activity of the newly synthesized compounds can be translated into in vivo efficacy, we tested the antitumor activity of compound NP19 on a mice melanoma B16–F10 tumor model. NP19 was chosen for the in vivo efficacy study due to the ease of synthesis and less cytotoxicity (Table 9) when compared to the more potent compound NP2 or equally potent compound NP12. We treated BALB/c mice bearing melanoma tumors with vehicle control and NP19 (25 mg/kg, 50 mg/kg, 100 mg/kg) administered via intragastric gavage once a day for 15 days. As shown in Figure 6, after 15 days of treatment, the growth of melanoma tumors was inhibited dramatically following NP19 treatment. In Vivo Pharmacokinetic Properties of NP19 [an analog of BMS-8][3] As compound NP19 showed high potency in vitro, the pharmacokinetic (PK) profiles were next evaluated in male Sprague–Dawley rats by intravenous and oral administration. The key p.o. and i.v. administration PK parameters are summarized in Table 8. After a single i.v. administration with 1 mg/kg compound NP19, the half time (t1/2), the clearance rate (CL), and the apparent distribution volume (Vss) of NP19 are 1.5 ± 0.5 h, 0.9 ± 0.2 L/h/kg, and 2.1 ± 0.5 L/kg, respectively. When NP19 was administrated by the oral route at 10 mg/kg, the oral absorption (Tmax = 0.6 ± 0.2 h), long half-life (t1/2 = 10.9 ± 7.7 h), and oral bioavailability (F = 5%) were observed. In addition, no apparent adverse effects were observed in rats. NP19 showed a much longer half-life (10.9 h) following oral gavage as compared to the i.v. half-life (1.5 h); this may be due to the high lipophilicity (logP = 7.9) or poor aqueous solubility of NP19. As a result, NP19 exhibited flip-flop pharmacokinetics. Such flip-flop pharmacokinetics can sometimes occur for poorly water-soluble compounds such as Rebamipide, which has a t1/2 (p.o.)/t1/2 (i.v.) ratio of 13.5 due to poor water solubility (7.6 μg/mL). Another example is the lipophilic compound IAT (an antitubulin agent with 19 μg/mL of water solubility) reported by Chien-ming Li et al., which has a t1/2 (p.o.)/t1/2 (i.v.) ratio of ∼5, similar to NP19 [t1/2 (p.o.)/t1/2 (i.v.) = 7.1]. Due to the low oral bioavailability of the compound NP19, we presumed that a high dosage is needed to offer sufficient drug concentration to exhibit antitumor efficacy. Therefore, we further studied the in vivo activity of compound NP19. |
Enzyme Assay |
All binding studies are performed in an HTRF assay buffer consisting of dPBS supplemented with 0.1% (with v) bovine serum albumin and 0.05% (v/v) Tween-20. For the PD-l-Ig/PD-Ll-His binding assay, inhibitors are pre-incubated with PD-Ll-His (10 nM final) for 15 m in 4 μL of assay buffer, followed by addition of PD-l-Ig (20 nM final) in 1 μL of assay buffer and further incubation for 15 m. PD-L1 from either human, cyno, or mouse are used. HTRF detection is achieved using europium crypate-labeled anti- Ig (1 nM final) and allophycocyanin (APC) labeled anti-His (20 nM final). Antibodies are diluted in HTRF detection buffer and 5 μL is dispensed on top of binding reaction. The reaction mixture is allowed to equilibrate for 30 minutes and signal (665 nm/620 nm ratio) is obtained using an En Vision fluorometer. Additional binding assays are established between PD-1-Ig/PD-L2-His (20, 5 nM, respectively), CD80-His/PD-Ll-Ig (100, 10 nM, respectively) and CD80-His/CTLA4-Ig (10, 5 nM, respectively).
|
Cell Assay |
Particularly in a tumor microenvironment where lysis of tumor cells is a concern, the interaction between programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) plays a dominant role in the suppression of T cell responses. With an IC50 value of 18 nM, PD-1/PD-L1 inhibitor 2 is said to stop PD-L1 from interacting with PD-1.
|
Animal Protocol |
Pharmacokinetic Study in Male Sprague–Dawley Rats[3]
Male Sprague–Dawley rats (200–220 g) were used to study the pharmacokinetics of compound NP19 [an analog of BMS-8]. Diet was prohibited for 12 h before the experiment, but water was freely available. Blood samples (0.3 mL) were collected from the tail vein into heparinized 1.5 mL polythene tubes at 0.0833, 0.25, 0.5, 1, 1.5, 2, 4, 6, 8, 12, and 24 h after oral (10 mg/kg) or intravenous (1 mg/kg) administration of compound NP19. The compound was dissolved in 5% DMSO and 95% PEG-300 for intravenous administration or suspended in 0.5% sodium carboxymethyl cellulose (CMC-Na) for oral administration. The samples were immediately centrifuged at 3000g for 10 min. The plasma as-obtained (100 μL) was stored at −20 °C until analysis. PK parameters were determined from individual animal data using noncompartmental analysis in DAS (Drug and statistics) software. Instruments and analytical conditions for PK studies: A UPLC-MS/MS system with ACQUITY I-Class UPLC and a XEVO TQD triple quadrupole mass spectrometer, equipped with an electrospray ionization (ESI) interface, was used to analyze the blood samples. The UPLC system was comprised of a Binary Solvent Manager (BSM) and a Sample Manager with Flow-Through Needle (SM-FTN). Masslynx 4.1 software was used for data acquisition and instrument control. Multiple reaction monitoring (MRM) modes of m/z 555.35 → 181.03 for NP19 and m/z 237 → 194.1 for carbamazepine were utilized to conduct quantitative analysis. In Vivo Efficacy Study in Mouse B16F10 Melanoma Model[3] BALB/c mice, aged 6–8 weeks old, were used to study the inhibition effect of NP19 [an analog of BMS-8]on subcutaneous transplanted model of melanoma cells. Murine B16F10 melanoma cells growing in a logarithmic growth phase were suspended in PBS at a density of 2 × 106 per mL. Each mouse was inoculated subcutaneously with 200 μL containing 4 × 105 cells. After tumors reached approximately 100 mm3 in volume, mice were divided into four groups randomly (n = 10) and treated with NP19 (25, 50, 100 mg/kg) and vehicle, respectively. The drugs were administered via intragastric gavage once a day for 15 days. The vehicle group was administered with 0.5% sodium carboxymethyl cellulose (CMC-Na). Animal activity and body weight were monitored during the entire experiment period to assess acute toxicity. Mice were sacrificed 16 days after the initiation of the treatment, and the tumor tissue and major organ (liver, spleen, thymus, and kidney) samples were collected. The harvested tumor tissue and organs (liver, kidney) were fixed in 4% paraformaldehyde, processed into paraffin routinely, stained with hematoxylin and eosin (H&E), and captured by microscope. Tumor growth inhibition value (TGI) was calculated using the formula: TGI(%) = [1 – Wt/Wv] × 100%, where Wt and Wv are the mean tumor weight of treatment group and vehicle control. In Vivo Efficacy Study in Mouse H22 Hepatoma Tumor Model[3] 6–8 weeks old male BALB/c mice were used. A total of 8 × 105 H22 cells were inoculated into the right flank of each mouse according to protocols of tumor transplant research. NP19 [an analog of BMS-8]was dissolved in 5% DMSO, 40% PEG-200 and 55% saline solution to produce desired concentrations. Mice in control groups were injected intraperitoneally with 200 μL of vehicle solution only. Tumor volume was measured every 2 days with a traceable electronic digital caliper and calculated using the formula a × b2 × 0.5, where a and b represented the larger and smaller diameters, respectively. The mice were sacrificed after the treatments and tumors were excised and weighed. |
References |
|
Additional Infomation |
Recently, small-molecule compounds have been reported to block the PD-1/PD-L1 interaction by inducing the dimerization of PD-L1. All these inhibitors had a common scaffold and interacted with the cavity formed by two PD-L1 monomers. This special interactive mode provided clues for the structure-based drug design, however, also showed limitations for the discovery of small-molecule inhibitors with new scaffolds. In this study, we revealed the structure-activity relationship of the current small-molecule inhibitors targeting dimerization of PD-L1 by predicting their binding and unbinding mechanism via conventional molecular dynamics and metadynamics simulation. During the binding process, the representative inhibitors (BMS-8 and BMS-1166) tended to have a more stable binding mode with one PD-L1 monomer than the other and the small-molecule inducing PD-L1 dimerization was further stabilized by the non-polar interaction of Ile54, Tyr56, Met115, Ala121, and Tyr123 on both monomers and the water bridges involved in ALys124. The unbinding process prediction showed that the PD-L1 dimerization kept stable upon the dissociation of ligands. It's indicated that the formation and stability of the small-molecule inducing PD-L1 dimerization was the key factor for the inhibitory activities of these ligands. The contact analysis, R-group based quantitative structure-activity relationship (QSAR) analysis and molecular docking further suggested that each attachment point on the core scaffold of ligands had a specific preference for pharmacophore elements when improving the inhibitory activities by structural modifications. Taken together, the results in this study could guide the structural optimization and the further discovery of novel small-molecule inhibitors targeting PD-L1.[2]
Cancer immunotherapy has been revolutionized by the development of monoclonal antibodies (mAbs) that inhibit interactions between immune checkpoint molecules, such as programmed cell-death 1 (PD-1), and its ligand PD-L1. However, mAb-based drugs have some drawbacks, including poor tumor penetration and high production costs, which could potentially be overcome by small molecule drugs. BMS-8, one of the potent small molecule drugs, induces homodimerization of PD-L1, thereby inhibiting its binding to PD-1. Our assay system revealed that BMS-8 inhibited the PD-1/PD-L1 interaction with IC50 of 7.2 μM. To improve the IC50 value, we designed and synthesized a small molecule based on the molecular structure of BMS-8 by in silico simulation. As a result, we successfully prepared a biphenyl-conjugated bromotyrosine (X) with IC50 of 1.5 μM, which was about five times improved from BMS-8. We further prepared amino acid conjugates of X (amino-X), to elucidate a correlation between the docking modes of the amino-Xs and IC50 values. The results suggested that the displacement of amino-Xs from the BMS-8 in the pocket of PD-L1 homodimer correlated with IC50 values. This observation provides us a further insight how to derivatize X for better inhibitory effect.[1] This work describes the modification of a gold electrode with the BMS-8 compound that interacts with the Programmed Death-Ligand 1 (PD-L1), an immune checkpoint protein. The results show that we can confirm the presence of the sPD-L1 in the concentration range of 10-18 to 10-8 M using electrochemical impedance spectroscopy (EIS) with a limit of detection (LOD) of 1.87 × 10-14 M for PD-L1 (S/N = 3.3) and at a concentration of 10-14 M via cyclic voltammetry (CV). Additionally, high-resolution X-ray photoelectron spectroscopy (XPS), contact angle, and surface free energy measurements were applied to confirm the functionalization of the electrode. We investigated the selectivity of the electrode for other proteins: Programmed Death-1 (PD-1), cluster of differentiation 160 (CD160), and B- and T-lymphocyte attenuator (BTLA) at concentrations of 10-8 M. Differentiation between PD-L1 and PD-1 was achieved based on the analysis of the capacitance effect frequency dispersion at the surface of the modified Au electrode with BMS-8 after incubation at various concentrations of PD-L1 and PD-1 proteins in the range of 10-18 to 10-8 M. Significant differences were observed in the heterogeneity of PD-L1 and PD-1. The results of the quasi-capacitance studies demonstrate that BMS-8 strongly and specifically interacts with the PD-L1 protein. https://pubmed.ncbi.nlm.nih.gov/33517203/ |
Molecular Formula |
C27H28BRNO3
|
|
---|---|---|
Molecular Weight |
494.43
|
|
Exact Mass |
493.125
|
|
Elemental Analysis |
C, 65.59; H, 5.71; Br, 16.16; N, 2.83; O, 9.71
|
|
CAS # |
1675201-90-7
|
|
Related CAS # |
|
|
PubChem CID |
117941742
|
|
Appearance |
White to off-white solid powder
|
|
Density |
1.3±0.1 g/cm3
|
|
Boiling Point |
616.9±55.0 °C at 760 mmHg
|
|
Flash Point |
326.9±31.5 °C
|
|
Vapour Pressure |
0.0±1.9 mmHg at 25°C
|
|
Index of Refraction |
1.620
|
|
LogP |
6.28
|
|
Hydrogen Bond Donor Count |
1
|
|
Hydrogen Bond Acceptor Count |
4
|
|
Rotatable Bond Count |
7
|
|
Heavy Atom Count |
32
|
|
Complexity |
596
|
|
Defined Atom Stereocenter Count |
0
|
|
SMILES |
BrC1=C(C=CC(=C1)CN1CCCCC1C(=O)O)OCC1C=CC=C(C2C=CC=CC=2)C=1C
|
|
InChi Key |
QRXBPPWUGITQLE-UHFFFAOYSA-N
|
|
InChi Code |
InChI=1S/C27H28BrNO3/c1-19-22(10-7-11-23(19)21-8-3-2-4-9-21)18-32-26-14-13-20(16-24(26)28)17-29-15-6-5-12-25(29)27(30)31/h2-4,7-11,13-14,16,25H,5-6,12,15,17-18H2,1H3,(H,30,31)
|
|
Chemical Name |
1-[[3-bromo-4-[(2-methyl-3-phenylphenyl)methoxy]phenyl]methyl]piperidine-2-carboxylic acid
|
|
Synonyms |
|
|
HS Tariff Code |
2934.99.9001
|
|
Storage |
Powder -20°C 3 years 4°C 2 years In solvent -80°C 6 months -20°C 1 month Note: This product requires protection from light (avoid light exposure) during transportation and storage. |
|
Shipping Condition |
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
|
Solubility (In Vitro) |
|
|||
---|---|---|---|---|
Solubility (In Vivo) |
Note: Listed below are some common formulations that may be used to formulate products with low water solubility (e.g. < 1 mg/mL), you may test these formulations using a minute amount of products to avoid loss of samples.
Injection Formulations
Injection Formulation 1: DMSO : Tween 80: Saline = 10 : 5 : 85 (i.e. 100 μL DMSO stock solution → 50 μL Tween 80 → 850 μL Saline)(e.g. IP/IV/IM/SC) *Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH ₂ O to obtain a clear solution. Injection Formulation 2: DMSO : PEG300 :Tween 80 : Saline = 10 : 40 : 5 : 45 (i.e. 100 μL DMSO → 400 μLPEG300 → 50 μL Tween 80 → 450 μL Saline) Injection Formulation 3: DMSO : Corn oil = 10 : 90 (i.e. 100 μL DMSO → 900 μL Corn oil) Example: Take the Injection Formulation 3 (DMSO : Corn oil = 10 : 90) as an example, if 1 mL of 2.5 mg/mL working solution is to be prepared, you can take 100 μL 25 mg/mL DMSO stock solution and add to 900 μL corn oil, mix well to obtain a clear or suspension solution (2.5 mg/mL, ready for use in animals). View More
Injection Formulation 4: DMSO : 20% SBE-β-CD in saline = 10 : 90 [i.e. 100 μL DMSO → 900 μL (20% SBE-β-CD in saline)] Oral Formulations
Oral Formulation 1: Suspend in 0.5% CMC Na (carboxymethylcellulose sodium) Oral Formulation 2: Suspend in 0.5% Carboxymethyl cellulose Example: Take the Oral Formulation 1 (Suspend in 0.5% CMC Na) as an example, if 100 mL of 2.5 mg/mL working solution is to be prepared, you can first prepare 0.5% CMC Na solution by measuring 0.5 g CMC Na and dissolve it in 100 mL ddH2O to obtain a clear solution; then add 250 mg of the product to 100 mL 0.5% CMC Na solution, to make the suspension solution (2.5 mg/mL, ready for use in animals). View More
Oral Formulation 3: Dissolved in PEG400  (Please use freshly prepared in vivo formulations for optimal results.) |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 2.0225 mL | 10.1127 mL | 20.2253 mL | |
5 mM | 0.4045 mL | 2.0225 mL | 4.0451 mL | |
10 mM | 0.2023 mL | 1.0113 mL | 2.0225 mL |
*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.
Calculation results
Working concentration: mg/mL;
Method for preparing DMSO stock solution: mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.
Method for preparing in vivo formulation::Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.
(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
(2) Be sure to add the solvent(s) in order.
Structural Biology of the Immune Checkpoint Receptor PD-1 and Its Ligands PD-L1/PD-L2.Structure.2017 Aug 1;25(8):1163-1174. th> |
---|
New Directions in Designing the Therapeutics Targeting the PD-1/PD-L1 Interaction.Structure.2017 Aug 1;25(8):1163-1174. td> |
Structural Basis of the PD-1/PD-L1 (PD-L2) Interaction.Structure.2017 Aug 1;25(8):1163-1174. td> |
J Med Chem.2017Jul 13;60(13):5857-5867. th> |
---|
J Med Chem.2017Jul 13;60(13):5857-5867. |
J Med Chem.2017Jul 13;60(13):5857-5867. |