Size | Price | Stock | Qty |
---|---|---|---|
10mg |
|
||
25mg |
|
||
50mg |
|
||
100mg |
|
||
250mg |
|
||
Other Sizes |
|
Purity: ≥98%
Lusutrombopag (formerly known as S888711; S-888711; trade name: Mulpleta) is a novel orally bioavailable, small molecule agonist of thrombopoietin (TPO) receptor. It can activate the TPO signal transduction pathways, thus increasing platelet levels. Lusutrombopag was developed by Shionogi for chronic liver disease (CLD) patients with thrombocytopenia prior to elective invasive surgery. On 7/31/2018, Lusutrombopag was approved by FDA to treat thrombocytopenia in adult patients with chronic liver disease who are scheduled to undergo a procedure. Lusutrombopag acts selectively on the human TPO receptor and activates signal transduction pathways that promote the proliferation and differentiation of bone marrow cells into megakaryocytes, thereby increasing platelet levels. In September 2015, lusutrombopag received its first global approval in Japan for the improvement of CLD-associated thrombocytopenia in patients scheduled to undergo elective invasive procedures.
Lusutrombopag (S-888711), an oral small-molecule thrombopoietin receptor (TPOR) agonist, has gained first approval as a drug to treat thrombocytopenia of chronic liver disease in patients undergoing elective invasive procedures in Japan. Preclinical studies were performed to evaluate its efficacy against megakaryopoiesis and thrombopoiesis.Targets |
TPO/thrombopoietin receptor
|
---|---|
ln Vitro |
By specifically acting on human TPO, lutetrombopag effectively receives and initiates signal transmission, thereby driving bone marrow cell growth and necrosis into megakaryocytes and elevating the warning level [1].
To investigate the proliferative activity and efficacy of megakaryocytic colony formation via human TPOR, lusutrombopag was applied to cultured human c-Mpl-expressing Ba/F3 (Ba/F3-hMpl) cells and human bone marrow-derived CD34-positive cells, respectively. Lusutrombopag caused a robust increase in Ba/F3-hMpl cells by activating pathways in a manner similar to that of thrombopoietin and induced colony-forming units-megakaryocyte and polyploid megakaryocytes in human CD34-positive cells. [2] Lusutrombopag exhibits agonist activity for human TPO receptor c-Mpl [2] Lusutrombopag promoted the proliferation of Ba/F3-hMpl cells. The 50% EC50 values of lusutrombopag and rhTPO in Ba/F3-hMpl cells were 84.0 and 0.08 nmol/L, respectively (Fig. 2A), whereas lusutrombopag exhibited no proliferative activity in Ba/F3-mMpl cells (Fig. 2B). These results indicate that lusutrombopag promotes the proliferation of Ba/F3-hMpl cells via human c-Mpl. To investigate the signal transduction pathway of lusutrombopag, we evaluated the phosphorylation of JAK2, STAT3, STAT5 and p44/42 MAPK in Ba/F3-hMpl cells. Lusutrombopag phosphorylated these molecules similarly to rhTPO (Fig. 2C). These results suggest that lusutrombopag activates the same signal transduction pathways activated by rhTPO. Differentiation of CD34-positive hematopoietic cells promoted by lusutrombopag [2] CFU-Mk activity of lusutrombopag was investigated using HuBM-CD34-positive cells. After incubation with lusutrombopag or rhTPO for 12 days, the CFU-Mk colonies were detected by anti-human CD41 antibody, and the CFU-Mk colonies were counted. The mean number of CFU-Mk colonies was 147.3 in the rhTPO (1.846 nmol/L)-treated group. The activity of rhTPO was defined as 100% activity, and the EC50 value of lusutrombopag was 0.31 µmol/L (Fig. 3A). CFU-Mk colonies of rhTPO and lusutrombopag were immunohistochemically detected using anti-human CD41 antibody. CFU-Mk colonies did not morphologically differ between rhTPO and lusutrombopag (Fig. 3B). For further investigation of megakaryocyte maturation, we examined megakaryocytic ploidization induced by lusutrombopag using a flow cytometer. HuBM-CD34-positive cells were cultured under serum-free conditions with lusutrombopag or rhTPO for 10 days. DNA ploidy of megakaryocytes in lusutrombopag had a distribution similar to that in rhTPO (Fig. 3C). These findings indicate that lusutrombopag is able to differentiate mature megakaryocytes from human hematopoietic stem cells. |
ln Vivo |
Thrombopoietic activity of lusutrombopag in TPOR-Ki/Shi mice [2]
Lusutrombopag induced proliferative activity in human TPO receptor-expressing cells, but induced no proliferative activity in murine TPO receptor-expressing cells in vitro. Platelet production activity of lusutrombopag was evaluated in TPOR-Ki/Shi mice or wild-type mice. Lusutrombopag or vehicle (0.5% MC) was orally administered to TPOR-Ki/Shi mice or wild-type mice once a day for 14 days and the platelets were counted on days 7, 14, 21, and 28. Fig. 6 illustrates the development of platelet production after lusutrombopag administration. Platelet production was used to determine the platelet increase ratio (dividing platelet count on treatment by basal platelet count). Platelet increase ratios of TPOR-Ki/Shi mice treated with lusutrombopag were significantly higher than those of the vehicle-treated TPOR-Ki/Shi. The platelet number was increased 2.5 times by the treatment with lusutrombopa. After discontinuation of lusutrombopag, the platelets decreased by day 21 and returned to close to the basal level on day 28. In contrast, the platelets of wild-type mice treated with lusutrombopag did not increase during the experimental period. To clarify its minimum thrombocytopoietic effect, lusutrombopag at doses of 0.3, 1, 3, and 10 mg/kg/day or vehicle (0.5% MC) was orally administered to TPOR-Ki/Shi mice once daily for 21 consecutive days. Blood samples were collected from veins at group assignment (day 0), prior to dosing on days 8 and 15, and 24 hours after dosing on day 21 (day 22), and then the platelets in each sample were counted. Lusutrombopag significantly increased platelet count on day 8, in a dose-dependent manner, as compared with the vehicle control group. The platelet numbers continued to increase on days 15 and 22 (Fig. 7). The minimum effective dose of lusutrombopag was 0.3 mg/kg/day in the 21-day repeated oral administration study. Because lusutrombopag has high species specificity for human TPOR, there was no suitable experimental animal model for drug evaluation, except for immunodeficient mouse-based xenograft models. Therefore, a novel genetically modified knock-in mouse, TPOR-Ki/Shi, was developed by replacing mouse Mpl with human-mouse chimera Mpl. In TPOR-Ki/Shi mice, lusutrombopag significantly increased circulating platelets in a dose-dependent manner during 21-day repeated oral administration. Histopathological study of the TPOR-Ki/Shi mice on day 22 also revealed a significant increase in megakaryocytes in the bone marrow. These results indicate that lusutrombopag acts on human TPOR to upregulate differentiation and proliferation of megakaryocytic cells, leading to platelet production.[2] |
Cell Assay |
Proliferation assay [2]
Ba/F3-hMpl cells and Ba/F3-mMpl cells were engineered from the murine interleukin (IL)-3-dependent pro-B-cell line Ba/F3, as previously described. Cells were cultured at a density of 7.5 × 103 cells/200 µL in 96-well plates with lusutrombopag (4.88–5000 nmol/L) or recombinant human TPO (rhTPO: 4.88–5000 pmol/L). The plates were incubated for 3 days at 37°C in a humidified chamber with 5% CO2; 10 µL WST-8 reagent was added to each well during the last 2–8 hours of culture. The absorbance was measured at a wavelength of 450 nm using a 96-well microplate reader. Megakaryocytic colony formation and ploidy assay [2] Human bone marrow-derived CD34-positive (HuBM-CD34-positive) cells were obtained from human tissue with informed consent. This study was approved by the relevant institutional ethics committee. A colony-forming unit-megakaryocyte (CFU-Mk) assay was performed using the MegaCult-C kit. HuBM-CD34-positive cells were cultured on chamber slides with lusutrombopag or rhTPO for 12 days, the slides were stained with anti-human CD41 antibody, and the CFU-Mk colonies were counted. The 50% effective concentration (EC50) of lusutrombopag was calculated using the sigmoid maximum pharmacological effect (Emax) model. To investigate megakaryocyte maturation, HuBM-CD34 positive cells were cultured with lusutrombopag or hTPO for 10 days, and megakaryocytic ploidization was detected using a flow cytometer. More details of methods can be found in the Supplementary Methods (online only, available at www.exphem.org). Western blotting [2] Ba/F3-hMpl cells were cultured in RPMI containing 0.5% bovine serum albumin (BSA, Wako, Osaka, Japan) for 5 hours and stimulated with 3 µM lusutrombopag or 1 nM rhTPO at 37°C for 15 minutes. After stimulation with lusutrombopag or rhTPO, the cells were washed with ice-cold PBS and lysed at 4°C for 30 minutes in lysis buffer containing protease inhibitor cocktail and phosphatase inhibitor cocktail. The protein samples were added to equal volume of 2 × Laemmli's sample buffer / 10% 2-mercaptoethanol, and boiled at 95°C for 5 min. The protein samples were separated by SDS-PAGE and transferred onto Immun-Blot PVDF Membrane. The blots were blocked by 5% BSA in TBS-T (20 mM Tris/HCl, pH 7.5, 150 mM NaCl, 0.05% Tween20) for 2 hours, and then incubated overnight at 4°C with anti-phospho JAK2 (Tyr1007/1008), anti-phospho STAT3 (Tyr705), anti-phospho STAT5 and anti-phospho p44/42 MAP kinase (Thr202/204) antibodies. The blots were stripped with Re-blot plus mild solution and re-probed with anti-JAK2, anti-STAT3, anti-STAT5 and anti-p44/42 MAP kinase antibodies. The blots were incubated with ECL Western blotting detection reagent and analysis system and exposed to Amersham Hyperfilm ECL. CFU-MK assay [2] HuBM-CD34 positive cells were cultured with lusutrombopag (0.0923–9.23 µM) or rhTPO (1.8 nM) and after 12 days of incubation, the slides were stained with anti-human CD41 antibody and the CFU-Mk colonies were counted. The maximum CFU-Mk activity of rhTPO was defined as 100%, and 50% effective concentration (EC50) of lusutrombopag was calculated using the sigmoid maximum pharmacological effect (Emax) model. Megakaryocytes in liquid culture and ploidy analysis [2] HuBM-CD34 positive cells were cultured at a density of 7.5 × 104 cells/mL in 24-well plates. As a serum-free medium, we used Iscove's modified Dulbecco's medium supplemented with 20% BIT9500 and 40 µg/mL human low density lipoprotein (LDL). Cells were treated with 3 µM lusutrombopag or 1 nM rhTPO in triplicate for 10 days at 37°C in a humidified chamber with 5% CO2. The cells were resuspended in Hank's Balanced Salt Solutions (HBSS) containing 13.6 mM sodium citrate, 2 µM prostaglandin E-1, 1 mM theophylline, 3% BSA, 11 mM glucose (Mk medium). Cells were stained with fluorescein isothiocyanate (FITC)-labeled anti-human CD41 antibody and propidium iodide as described previously. The cells were analyzed using an EPICS-XL flow cytometer. Data was obtained by electronically gating PI-stained CD41 positive cells, and at least 10,000 cells were analyzed for each sample. The ploidy distribution was determined by setting markers at the nadirs between the peaks. |
Animal Protocol |
Effect on platelet production in human TPOR-expressing mice [2]
To evaluate the thrombocytopoietic effect of lusutrombopag in vivo, the genetically modified mouse TPOR-Ki/Shi was developed using knocked-in technology that enabled replacement of the mouse TM domain of TPOR with a human–mouse chimera TM domain. TPOR-Ki/Shi mice were used in this study. Development of the genetically modified mouse is discussed in the Supplementary Methods. To clarify the effectiveness of TPOR-Ki/Shi mice, we investigated the effect on platelet production of oral administration of lusutrombopag to TPOR-Ki/Shi mice or C57BL/6 mice, the wild type of TPOR-Ki/Shi mice. Eight female TPOR-Ki/Shi mice and C57BL/6 mice in each group were administered 10 mg/kg/day lusutrombopag or vehicle (0.5% methylcellulose [MC] aqueous solution) daily for 14 days. Blood was collected from veins under anesthesia 1 day before the start of administration (day 0) and on days 7, 14, 21 and 28, and the platelets were counted using a K-4500 multi-automatic hemocytometer. Dose escalation of platelet production of lusutrombopag and megakaryocytopoiesis in bone marrow was investigated using TPOR-Ki/Shi mice. Thrombopoietic effect and morphometric analysis of megakaryocytes in TPOR-Ki/Shi mice [2] The thrombocytopoietic effect of lusutrombopag (0.3, 1, 3 and 10 mg/kg) and an increase of megakaryocytopoiesis in the bone marrow and hematological changes of lusutrombopag (0.3 and 10 mg/kg/day) were examined in TPOR-Ki/Shi mice (female, eight per group, 11–12 weeks old). Lusutrombopag or vehicle (0.5% MC) was orally administered once daily for 21 consecutive days. Small amounts of blood samples were collected from the vein under anesthesia and the platelet number was then counted using a K-4500 multi automatic hemocytometer. For histological and hematological study, on the day following the final administration (Day 22), mice were anesthetized with pentobarbital and blood was collected from the posterior vena cava and whole blood treated with EDTA-2K was subjected to analysis using the ADVIA 120 Hematology System. The following tissues were fixed in 10% neutral buffered formalin and embedded in paraffin. Paraffin sections were stained with hematoxylin and eosin (H&E), and then five fields of bone marrow were randomly selected and photographed by computer-digitizing imaging system consisting of a light microscope with a camera. The megakaryocyte number was counted in the fields and average data were expressed as mean ± standard deviation (SD). |
ADME/Pharmacokinetics |
Absorption, Distribution and Excretion
Lusutrombopag is rapidly absorbed following oral administration. It exhibited a dose‐proportional pharmacokinetic profile over the single dose range of 1 mg to 50 mg, which was similar in both healthy subjects and those with chronic liver disease. A geometric mean (%CV) maximal concentration (Cmax) and area under the curve (AUC) in healthy subjects receiving 3 mg of lusutrombopag were 111 (20.4) ng/mL and 2931 (23.4) ng.hr/mL. The accumulation ratios of Cmax and AUC were approximately 2 with once‐daily multiple‐dose administration, and steady‐state plasma lusutrombopag concentrations were achieved after Day 5. The time to reach peak plasma concentrations (Tmax) were approximately 6 to 8 hours after oral administration in patients with chronic liver disease. Food consumption is not reported to affect the absorption and bioavailability of lusutrombopag. About 1% of the administered dose of lusutrombopag undergoes urinary excretion. Fecal excretion accounted for 83% of the total dose, where 16% of the dose was excreted as unchanged parent compound. The mean (%CV) lusutrombopag apparent volume of distribution in healthy adult subjects was 39.5 (23.5) L. The approximate mean (%CV) clearance of lusutrombopag in patients with chronic liver disease is estimated to be 1.1 (36.1) L/hr. Metabolism / Metabolites CYP4 enzymes predominantly contribute to the metabolism of lusutrombopag, especially CYP4A11. Lusutrombopag is reported to mainly undergo ω- and β-oxidation, as well as glucuronidation. Biological Half-Life In healthy adult subjects, the terminal elimination half‐life (t1/2) was approximately 27 hours. |
Toxicity/Toxicokinetics |
Effects During Pregnancy and Lactation
◉ Summary of Use during Lactation No information is available on the use of lusutrombopag during breastfeeding. The manufacturer recommends avoiding breastfeeding during the use of lusutrombopag and for at least 28 days after the last dose. ◉ Effects in Breastfed Infants Relevant published information was not found as of the revision date. ◉ Effects on Lactation and Breastmilk Relevant published information was not found as of the revision date. Protein Binding The plasma protein binding of lusutrombopag is more than 99.9%. |
References | |
Additional Infomation |
Pharmacodynamics
The AUC of lusutrombopag was found to correlate the increased platelet counts. Following administration of 3 mg daily dose in patients with chronic liver disease and thrombocytopenia, the mean (standard deviation) maximum platelet count in patients (N=74) without platelet transfusion was 86.9 (27.2) × 10^9/L, and the median time to reach the maximum platelet count was 12.0 (5 to 35) days. Lusutrombopag was not shown to induce any clinically significant QTc prolongation at a dose 8 times the recommended dosage. Lusutrombopag is a member of cinnamic acids. Lusutrombopag is an orally bioavailable thrombopoietin receptor (TPOR) agonist developed by Shionogi & Company (Osaka, Japan). TPOR is a regulatory target site for endogenous thrombopoietin, which acts as a primary cytokine to promote megakaryocyte proliferation and differentiation, and affect other hematopoietic lineages as well, including erythroid, granulocytic and lymphoid lineages. Thrombocytopenia, which indicates abnormally low levels of platelets, is a common complication related to chronic liver disease. This hematological abnormality, especially in cases of severe thrombocytopenia (platelet count <50,000/μL), creates challenges to patients requiring invasive medical procedures where there is a significant risk for spontaneous bleeding. Lusutrombopag binds to the transmembrane domain of TPOR expressed on megakaryocytes, and causes the proliferation and differentiation of megakaryocytic progenitor cells from hematopoietic stem cells. In September 2015, lusutrombopag received its first global approval in Japan to reduce the need for platelet transfusion in adults with chronic liver disease and thrombocytopenia who are schedule to undergo an invasive medical procedure. Lusutrombopag was approved by the FDA on July 31st, 2018 for the same therapeutic indication under the market name Mulpleta. In two randomized, double-blind, placebo-controlled trials, patients with chronic liver disease and severe thrombocytopenia who were undergoing an invasive procedure with a platelet count less than 50 x 10^9/L were administered lusutrombopag orally. Higher percentages (65-78%) of the patients receiving lusutrombopag required no platelet transfusion prior to the primary invasive procedure compared to those receiving placebo. Lusutrombopag is currently in phase III development in various European countries including Austria, Belgium, Germany, and the UK. Lusutrombopag is an orally available thrombopoietin (TPO) receptor (TPOR; MPL) agonist, with potential megakaryopoiesis stimulating activity. Upon administration, lusutrombopag binds to and interacts with the transmembrane domain of human TPO receptor expressed on megakaryocytes, which leads to the proliferation and differentiation of megakaryocytic progenitor cells from hematopoietic stem cells. This increases the production of platelets and may prevent or treat thrombocytopenia in patients with chronic liver disease. TPOR is a cytokine receptor and member of the hematopoietic receptor superfamily. LUSUTROMBOPAG is a small molecule drug with a maximum clinical trial phase of IV (across all indications) that was first approved in 2018 and is indicated for thrombocytopenia and hemorrhage and has 1 investigational indication. Lusutrombopag (Mulpleta®) is an orally bioavailable, small molecule thrombopoietin (TPO) receptor agonist being developed by Shionogi for chronic liver disease (CLD) patients with thrombocytopenia prior to elective invasive surgery. Lusutrombopag acts selectively on the human TPO receptor and activates signal transduction pathways that promote the proliferation and differentiation of bone marrow cells into megakaryocytes, thereby increasing platelet levels. In September 2015, lusutrombopag received its first global approval in Japan for the improvement of CLD-associated thrombocytopenia in patients scheduled to undergo elective invasive procedures. This article summarizes the milestones in the development of lusutrombopag leading to this first approval.[1] The TPO-Ki/Shi mouse could be a suitable animal model for evaluation of the systemic effects of synthetic TPOR agonists that require the histidine residue in the TM region of c-Mpl for their activity. The results indicate that lusutrombopag acts on TPOR to upregulate differentiation and proliferation of megakaryocytic cells leading to platelet production. [2] |
Molecular Formula |
C29H32CL2N2O5S
|
---|---|
Molecular Weight |
591.54
|
Exact Mass |
590.14
|
Elemental Analysis |
C, 58.88; H, 5.45; Cl, 11.99; N, 4.74; O, 13.52; S, 5.42
|
CAS # |
1110766-97-6
|
Related CAS # |
Lusutrombopag-d13
|
PubChem CID |
49843517
|
Appearance |
White to off-white solid powder
|
Density |
1.3±0.1 g/cm3
|
Index of Refraction |
1.618
|
LogP |
8.64
|
Hydrogen Bond Donor Count |
2
|
Hydrogen Bond Acceptor Count |
7
|
Rotatable Bond Count |
13
|
Heavy Atom Count |
39
|
Complexity |
822
|
Defined Atom Stereocenter Count |
1
|
SMILES |
CCCCCCO[C@@H](C)C1=CC=CC(=C1OC)C2=CSC(=N2)NC(=O)C3=CC(=C(C(=C3)Cl)/C=C(\C)/C(=O)O)Cl
|
InChi Key |
NOZIJMHMKORZBA-KJCUYJGMSA-N
|
InChi Code |
InChI=1S/C29H32Cl2N2O5S/c1-5-6-7-8-12-38-18(3)20-10-9-11-21(26(20)37-4)25-16-39-29(32-25)33-27(34)19-14-23(30)22(24(31)15-19)13-17(2)28(35)36/h9-11,13-16,18H,5-8,12H2,1-4H3,(H,35,36)(H,32,33,34)/b17-13+/t18-/m0/s1
|
Chemical Name |
(2E)-3-(2,6-Dichloro-4-((4-(3-((1S)-1-(hexyloxy)ethyl)-2-methoxyphenyl)-1,3-thiazol-2-yl)carbamoyl)phenyl)-2-methylprop-2-enoic acid
|
Synonyms |
S-888711; S888711; LUSUTROMBOPAG; 1110766-97-6; mulpleta; (S,E)-3-(2,6-Dichloro-4-((4-(3-(1-(hexyloxy)ethyl)-2-methoxyphenyl)thiazol-2-yl)carbamoyl)phenyl)-2-methylacrylic acid; UNII-6LL5JFU42F; 6LL5JFU42F; RSC888711; S 888711; Trade name: Mulpleta
|
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 |
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) |
Solubility in Formulation 1: ≥ 2.5 mg/mL (4.23 mM) (saturation unknown) in 10% DMSO + 40% PEG300 + 5% Tween80 + 45% Saline (add these co-solvents sequentially from left to right, and one by one), clear solution.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 400 μL PEG300 and mix evenly; then add 50 μL Tween-80 to the above solution and mix evenly; then add 450 μL normal saline to adjust the volume to 1 mL. Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH₂ O to obtain a clear solution. Solubility in Formulation 2: 2.5 mg/mL (4.23 mM) in 10% DMSO + 90% (20% SBE-β-CD in Saline) (add these co-solvents sequentially from left to right, and one by one), suspension solution; with ultrasonication. For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 900 μL of 20% SBE-β-CD physiological saline solution and mix evenly. Preparation of 20% SBE-β-CD in Saline (4°C,1 week): Dissolve 2 g SBE-β-CD in 10 mL saline to obtain a clear solution. View More
Solubility in Formulation 3: ≥ 2.5 mg/mL (4.23 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (add these co-solvents sequentially from left to right, and one by one), clear solution. |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 1.6905 mL | 8.4525 mL | 16.9050 mL | |
5 mM | 0.3381 mL | 1.6905 mL | 3.3810 mL | |
10 mM | 0.1691 mL | 0.8453 mL | 1.6905 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.