Size | Price | |
---|---|---|
500mg | ||
1g | ||
Other Sizes |
Purity: ≥98%
Pasireotide pamoate (formerly SOM-230; SOM230) is the pamoate salt of Pasireotide, which is a cyclohexapeptide somatostatin mimic and an orphan drug approved for the treatment of Cushing's disease in patients who fail or are ineligible for surgical therapy. It was developed by Novartis. Pasireotide is a somatostatin analogue with a 40-fold increased affinity to somatostatin receptor 5 compared to other somatostatin analogues.
Targets |
sst1 ( pKi = 8.2 ); sst2 ( pKi = 9.0 ); sst3 ( pKi = 9.1 ); sst4 ( pKi < 7.0 ); sst5 ( pKi = 9.9 )
|
---|---|
ln Vitro |
The human somatostatin receptor (subtypes sst1/2/3/4/5, pKi 8.2/9.0/9.1/<7.0/9.9, respectively) reacts uniquely and highly favorably to pasireotide pamoate[1]. With an effective inhibitory concentration (IC50 of 0.4 nM), pasireotide pamoate suppresses growth hormone-releasing hormone (GHRH)-induced growth hormone (GH) release in primary cultures of rat pituitary cells.
The strategy pursued in this research has been rewarded with the demonstrated superiority of Pasireotide/25 compared to 2. Pharmacological studies in vitro have clearly shown that Pasireotide/25 effectively inhibited the growth hormone releasing hormone (GHRH) induced growth hormone (GH) release in primary cultures of rat pituitary cells with an IC50 of 0.4 ± 0.1 nmol/L (n = 5) [1]. Pasireotide is a somatostatin analogue with high binding affinity to somatostatin receptor subtypes sst1,2,3 and sst5, as shown in competitive binding studies using CHO-K1 cells expressing human recombinant somatostatin receptors (Table 2) (Bruns et al., 2002, Schmid and Schoeffter, 2004). In CCL39 cells expressing human recombinant sst receptors, pasireotide and somatostatin (SRIF-14) inhibited forskolin-stimulated cAMP accumulation with approximately the same efficacy and potency. Compared with octreotide, the functional activity (based on EC50 values) of pasireotide on sst1, sst3 and sst5 was >30-, 11- and 158-fold higher, respectively, but 7-fold lower on sst2 (Schmid and Schoeffter, 2004). Based on the differences in binding affinity and functional activity of Pasireotide and octreotide, it can be speculated that in cells and tissues that express sst receptors other than the sst2 receptor subtype, pasireotide will have a stronger inhibitory effect on hormone secretion than octreotide [3]. |
ln Vivo |
Pasireotide pamoate (160 mg/kg/oral; subcutaneous injection for 4 months) dramatically lowered serum insulin, raised blood glucose, reduced tumor growth, and enhanced apoptosis in Pdx1-Cre [2]. Pasireotide pamoate (2-50 μg/kg; subcutaneously administered twice daily for 42 days) produces analgesic and anti-inflammatory effects through the SSTR2 receptor in a mouse model of immune-mediated arthritis [3 ].
In vivo, Pasireotide/25 also potently suppressed GH secretion in rats. The ED50 values determined at 1 and 6 h after injection of 25 indicated its very long duration of action in vivo. In the rat, 25 strongly decreases IGF-1 plasma levels, with the efficacy being markedly enhanced compared with the effects elicited by 2 after 7 days of treatment. Furthermore, in rats, dogs, and rhesus monkeys, 25 potently and dose-dependently decreased IGF-1 levels for prolonged periods of time without desensitization as observed with SMS 201-995 (2).[1] Background: Pasireotide (SOM230), a long-acting somatostatin analogue (LAR), has improved agonist activity at somatostatin receptors. We tested the effect of SOM230 on insulin secretion, serum glucose concentrations, tumor growth, and survival using an MEN1 transgenic mouse model. Methods: Eight 12-month-old conditional Men1 knockout mice with insulinoma were assessed. The treatment (n = 4) and control groups (n = 4) received monthly subcutaneous injections of SOM230 or PBS. Serum insulin and glucose levels were determined by enzyme-linked immunosorbent assay and enzymatic colorimetric assay, respectively. Tumor activity, growth, and apoptosis were determined by microPET/CT scan and histologic analysis. Results: On day 7, there was a decrease in serum insulin levels from 1.06 ± 0.28 μg/L to 0.37 ± 0.17 μg/L (P = .0128) and a significant increase in serum glucose from 4.2 ± 0.45 mmol/L to 7.12 ± 1.06 mmol/L (P = .0075) in the treatment group but no change in the control group. Tumor size was less in the treatment group (2,098 ± 388 μm(2)) compared with the control group (7,067 ± 955 μm(2); P = .0024). Furthermore, apoptosis was increased in the treatment group (6.9 ± 1.23%) compared with the control group (0.29 ± 0.103%; P = .002). Conclusion: SOM230 demonstrates antisecretory, antiproliferative, and proapoptotic activity in our MEN1 model of insulinoma. Further studies of the effects of SOM230 in PNET patients with MEN1 mutations are warranted.[2] Objective: Clinical and preclinical evidence suggests that somatostatin exhibits potent antiinflammatory and antinociceptive properties. However, it is not known which of the 5 somatostatin receptor subtypes (SSTRs 1-5) is involved in these actions. The purpose of this study was to assess the effects of the stable somatostatin analogs octreotide and Pasireotide (SOM230) in a mouse model of antigen-induced arthritis (AIA). Methods: Studies were performed in SSTR2-deficient mice (SSTR2(-/-)) and their wild-type littermates (SSTR2(+/+)). The expression of SSTR1, SSTR2A, SSTR3, and SSTR5 in dorsal root ganglia was examined by immunohistochemistry. Results: Untreated SSTR2(-/-) mice with AIA displayed joint swelling and mechanical hyperalgesia similar to that seen in SSTR2(+/+) mice. In wild-type mice, both octreotide and Pasireotide significantly attenuated knee joint swelling and histopathologic manifestations of arthritis to an extent comparable to that of dexamethasone. In SSTR2(-/-) mice, the antiinflammatory effects of both octreotide and pasireotide were completely abrogated. Prolonged administration of Pasireotide also inhibited joint swelling and protected against joint destruction during AIA flare reactions. In addition, both octreotide and pasireotide reduced inflammatory hyperalgesia. The antinociceptive actions of octreotide were abolished in SSTR2(-/-) mice, but those of pasireotide were retained. In dorsal root ganglia of naive wild-type mice, only SSTR1 and SSTR2A, but not SSTR3 or SSTR5, were detected in a subset of small- and medium-diameter neurons. Conclusion: Our findings indicate that the antinociceptive and antiinflammatory actions of octreotide and Pasireotide are largely mediated via the SSTR2 receptor. In addition, we identified the SSTR1 receptor as a novel pharmacologic target for somatostatin-mediated peripheral analgesia in inflammatory pain [4]. |
Enzyme Assay |
Pharmacological Characterization. Radioligand Binding Assays. [1]
Radioligand binding assays were performed as described previously. Briefly, membranes from CHO and COS cells expressing the respective human SRIF receptor subtype were incubated with the SRIF receptor ligand Tyr11[125I]-SRIF in the presence or absence of various concentrations of SRIF receptor ligands. The incubation was stopped after 1 h by rapid filtration through Whatman GF/C filters. Inhibition curves were analyzed, and IC50 values were calculated. |
Cell Assay |
Apoptosis analysis [2]
Apoptotic status in endocrine tumor tissues was measured in control and treated mice by Terminal deoxynucleotidyl transferase dUTP Nick End-Labeling (TUNEL) assay. For quantification of apoptosis, the TUNEL assay was performed according to the manufacturer on paraffin-embedded sections with an In Situ Cell Death Detection Kit. The tissue sections were deparaffinized and treated with proteinase K (10 μg/ml) for 20 min. The sections were then washed twice with PBS, labeled and stained with the TUNEL reaction mixture (label plus enzyme solutions) for 60 min at 37°C, and washed twice with PBS in the dark. The slides were mounted in Vectashield mounting medium with DAPI. The apoptotic fluorescent cells were counted under a fluorescent microscope and the numbers were expressed as the percentage of total cells ± standard deviation (SD). A negative control without enzyme treatment and a positive control with DNase I treatment were also performed. SSTR detection [2] The detection of sstr1-5 in pancreatic endocrine tumor tissue was measured in our mouse model. Immunofluorescent (IF) staining for sstr1-5 on sections was performed using rabbit or goat anti-sstr1, sstr2, sstr3, sstr4, and sstr5 antibodies (Abs) and were incubated at a 1:50 dilution overnight at 4° C, respectively. Pig anti-insulin Ab was also used for identification of β-cells. Sections were then washed with PBS and were incubated with a 1:200 dilution of anti-rabbit or anti-goat Alexa Fluor 488 and anti-pig Alexa Fluor 647 secondary antibodies for 45 min in the dark, respectively. A negative control without primary antibody was also performed. |
Animal Protocol |
Animal/Disease Models: 12-month-old conditional Men1 gene knockout insulinoma mice [2]
Doses: 160 mg/kg/oral Route of Administration: monthly subcutaneous injection for 4 months Experimental Results: Serum insulin diminished from 1.060 μg/L to 0.3653 μg/L, and increased blood sugar from 4.246 mM to 7.122 mM. Dramatically diminished tumor size and increased apoptosis. SOM230/Pasireotide and PBS Administration [2] Mice were anesthetized using halothane and then shaved on their flank for subcutaneous injection of either phosphate buffered saline (PBS) buffer or Pasireotide/SOM230 at a concentration of 160mg/Kg/month (64mg/ml) every month for 4 months. Treatment protocol and drugs. [4] Mice were randomly allocated to the following groups (8–10 animals per experimental condition): 0.9% saline; 2, 20, or 50 μg/kg of octreotide; or 2, 20, or 50 μg/kg of Pasireotide. These doses have been shown to elicit long-lasting therapeutic effects on pituitary hormone secretion in rodents and humans. Octreotide and Pasireotide were a kind gift from Novartis and were administered subcutaneously in a volume of 0.1 ml/kg of body weight. Treatment was started 12 hours before the induction of AIA and was continued for 3, 21, or 42 days, with administration every 12 hours for the indicated time periods. Flare reactions were provoked by injecting the right knee joint cavity with 100 μg of mBSA dissolved in 20 μl of PBS on days 21 and 35 of AIA. An additional group received 0.6 mg/kg of dexamethasone palmitate by intraperitoneal injection. Dexamethasone treatment was carried out for 5 days, followed by a 2-day pause starting 12 hours before AIA induction. |
ADME/Pharmacokinetics |
Absorption, Distribution and Excretion
The peak plasma concentration of pasireotide occurs in 0.25-0.5 hours. After administration of single and multiple doses, there is dose-proportionoal increases in Cmax and AUC. Pasireotide is eliminated mostly by hepatic clearance (biliary excretion)(about 48%) with some minor renal clearance (about 7.63%). Pasireotide is widely distributed and has a volume of distribution of >100L. The clearance in healthy patient is ~7.6 L/h and in Cushing’s disease patients is ~3.8 L/h. Metabolism / Metabolites Metabolism is minimal. Biological Half-Life The half-life is 12 hours. |
Toxicity/Toxicokinetics |
Effects During Pregnancy and Lactation
◉ Summary of Use during Lactation The excretion of pasireotide into breastmilk has not been studied. However, because it has a high molecular weight of 1047 daltons it is likely to be poorly excreted into breastmilk and it is a peptide that is likely digested in the infant's gastrointestinal tract. It is unlikely to reach the clinically important levels in infant serum. However, the manufacturer states that nursing mothers should not use pasireotide. An alternate drug is preferred. ◉ 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. |
References |
|
Additional Infomation |
Pasireotide is a six-membered homodetic cyclic peptide composed from L-phenylglycyl, D-tryptophyl, L-lysyl, O-benzyl-L-tyrosyl, L-phenylalanyl and modified L-hydroxyproline residues joined in sequence. A somatostatin analogue with pharmacologic properties mimicking those of the natural hormone somatostatin; used (as its diaspartate salt) for treatment of Cushing's disease. It has a role as an antineoplastic agent. It is a homodetic cyclic peptide and a peptide hormone. It is a conjugate base of a pasireotide(2+).
Pasireotide is a synthetic long-acting cyclic hexapeptide with somatostatin-like activity. It is marketed as a diaspartate salt called Signifor, which is used in the treatment of Cushing's disease. Pasireotide is a Somatostatin Analog. The mechanism of action of pasireotide is as a Somatostatin Receptor Agonist. Pasireotide is a synthetic polypeptide analogue of somatostatin that resembles the native hormone in its ability to suppress levels and activity of growth hormone, insulin, glucagon and many other gastrointestinal peptides. Because its half-life is longer than somatostatin, pasireotide can be used clinically to treat neuroendocrine pituitary tumors that secrete excessive amounts of growth hormone causing acromegaly, or adrenocorticotropic hormone (ACTH) causing Cushing disease. Pasireotide has many side effects including suppression of gall bladder contractility and bile production, and maintenance therapy can cause cholelithiasis and accompanying elevations in serum enzymes and bilirubin. Pasireotide is a synthetic long-acting cyclic peptide with somatostatin-like activity. Pasireotide activates a broad spectrum of somatostatin receptors, exhibiting a much higher binding affinity for somatostatin receptors 1, 3, and 5 than octreotide in vitro, as well as a comparable binding affinity for somatostatin receptor 2. This agent is more potent than somatostatin in inhibiting the release of human growth hormone (HGH), glucagon, and insulin. See also: Pasireotide Diaspartate (is active moiety of); Pasireotide Pamoate (is active moiety of). Drug Indication For the treatment of Cushing’s disease, specifically for those patients whom pituitary surgery has not been curative or is not an option. FDA Label Signifor is indicated for the treatment of adult patients with Cushing's disease for whom surgery is not an option or for whom surgery has failed. Signifor is indicated for the treatment of adult patients with acromegaly for whom surgery is not an option or has not been curative and who are inadequately controlled on treatment with another somatostatin analogue. Treatment of acromegaly and pituitary gigantism Overproduction of pituitary ACTH, Pituitary dependant Cushing's disease, Pituitary dependant hyperadrenocorticism Mechanism of Action Pasireotide activates a broad spectrum of somatostatin receptors, exhbiting a much higher binding affinity for somatostatin receptors 1, 3, and 5 than octreotide in vitro, as well as a comparable binding affinity for somatostatin receptor 2. The binding and activation of the somatostatin receptors causes inhibition of ACTH secretion and results in reduced cortisol secretion in Cushing's disease patients. Also this agent is more potent than somatostatin in inhibiting the release of human growth hormone (HGH), glucagon, and insulin. Pharmacodynamics Signifor® is an analogue of somatostatin that promotes reduced levels of cortisol secretion in Cushing's disease patients. A rational drug design approach, capitalizing on structure-activity relationships and involving transposition of functional groups from somatotropin release inhibitory factor (SRIF) into a reduced size cyclohexapeptide template, has led to the discovery of SOM230 (25), a novel, stable cyclohexapeptide somatostatin mimic that exhibits unique high-affinity binding to human somatostatin receptors (subtypes sst1-sst5). SOM230 has potent, long-lasting inhibitory effects on growth hormone and insulin-like growth factor-1 release and is a promising development candidate currently under evaluation in phase I clinical trials. [1] Pasireotide (SOM230) is a multi-receptor ligand somatostatin analogue with high binding affinity for somatostatin receptor subtypes sst(1,2,3) and sst(5). Pasireotide potently suppresses GH, IGF-I and ACTH secretion, indicating potential efficacy in acromegaly and Cushing's disease. The prolonged inhibition of hormone secretion by pasireotide in animal models and expression of multiple sst receptors in carcinoid tumors suggests that pasireotide may have clinical advantages over octreotide in patients with carcinoid tumors. Direct and indirect antitumor activity has been observed in vitro with pasireotide, including sst receptor-mediated apoptosis and antiangiogenesis, suggesting a possible role for pasireotide in antineoplastic therapy. In summary, preclinical evidence, as well as preliminary results from clinical studies suggests that pasireotide is a promising new treatment for patients with symptoms of metastatic carcinoid tumors refractory or resistant to octreotide, de novo or persistent acromegaly, and that pasireotide has the potential to be the first directed medical therapy for Cushing's disease.[3] In summary, this study is the first of its kind to demonstrate the antisecretory, antiproliferative and proapoptotic actions of the novel long acting release somatostatin analogue SOM230 in a mouse model of insulinomas with improved overall survival. The enhanced spectrum of activity of SOM230 is a result of its enhanced activity at 4 of the 5 sstr subtypes: sstr5, sstr2, sstr3, and sstr1. This is of particular clinical importance in unresectable or metastatic insulinomas that are relatively unresponsive to traditional therapy with octreotide and/or diazoxide. Treatment with SOM230 may facilitate symptomatic relief and result in tumor regression. We believe that this novel strategy of targeted therapy with SOM230 will be of benefit to patients with PNETs. While these data strongly support the effects of pasireotide in this model, we acknowledge that further studies with larger sample sizes are warranted to advance this novel approach toward clinical trials. [2] We also studied the progression of histopathologic changes of chronic inflammation after prolonged administration of pasireotide. These changes become particularly evident upon repeated injections of the antigen into the joint. The present data clearly show that in this model of immune-mediated arthritis, not only the acute inflammatory reaction, but also the chronic inflammatory/destructive process, can be dampened by SSTR agonists. Due to their antiinflammatory actions (equivalent to dexamethasone) and their antinociceptive actions, SSTR agonists are of interest for clinical therapy. In fact, in a pilot study in RA patients, significant clinical improvement was noted after treatment with octreotide. The present observations suggest that pasireotide and octreotide would exhibit similar antiinflammatory effects; however, pasireotide would be expected to provide better pain control than octreotide in RA. As an important aspect, treatment with somatostatin analogs is considered to be relatively safe and well tolerated. Moreover, for prolonged use, long-acting versions of both octreotide and pasireotide are available; these need to be administered only once a month, thus eliminating the need for daily subcutaneous injections. In conclusion, we provide evidence of potent antiinflammatory and antinociceptive effects of the somatostatin receptor agonists octreotide and pasireotide. We identify SSTR2 as an important target involved in the antiinflammatory effects of somatostatin. Both SSTR2 and SSTR1 mediate antinociception. Concerning the clinical use of SSTR agonists, it should be considered that pan-SSTR agonists may be superior to selective SSTR2 agonists.[4] |
Molecular Weight |
1435.57580041885
|
---|---|
Exact Mass |
1434.596
|
Elemental Analysis |
C, 67.77; H, 5.76; N, 9.76; O, 16.72
|
CAS # |
396091-79-5
|
Related CAS # |
Pasireotide acetate;396091-76-2;Pasireotide ditrifluoroacetate;Pasireotide L-aspartate salt;396091-77-3;Pasireotide;396091-73-9;Pasireotide (diaspartate);1421446-02-7
|
PubChem CID |
11982961
|
Appearance |
Typically exists as solid at room temperature
|
Hydrogen Bond Donor Count |
13
|
Hydrogen Bond Acceptor Count |
17
|
Rotatable Bond Count |
22
|
Heavy Atom Count |
106
|
Complexity |
2510
|
Defined Atom Stereocenter Count |
7
|
SMILES |
O(C(NCCN)=O)[C@H]1CN2C([C@H](CC3C=CC=CC=3)NC([C@H](CC3C=CC(=CC=3)OCC3C=CC=CC=3)NC([C@H](CCCCN)NC([C@@H](CC3=CNC4C=CC=CC3=4)NC([C@H](C3C=CC=CC=3)NC([C@@H]2C1)=O)=O)=O)=O)=O)=O.OC1C(C(=O)O)=CC2C=CC=CC=2C=1CC1=C(C(C(=O)O)=CC2C=CC=CC1=2)O
|
InChi Key |
HSXBEUMRBMAVDP-QKXVGOHISA-N
|
InChi Code |
InChI=1S/C58H66N10O9.C23H16O6/c59-27-13-12-22-46-52(69)64-47(30-38-23-25-42(26-24-38)76-36-39-16-6-2-7-17-39)53(70)66-49(31-37-14-4-1-5-15-37)57(74)68-35-43(77-58(75)61-29-28-60)33-50(68)55(72)67-51(40-18-8-3-9-19-40)56(73)65-48(54(71)63-46)32-41-34-62-45-21-11-10-20-44(41)45;24-20-16(14-7-3-1-5-12(14)9-18(20)22(26)27)11-17-15-8-4-2-6-13(15)10-19(21(17)25)23(28)29/h1-11,14-21,23-26,34,43,46-51,62H,12-13,22,27-33,35-36,59-60H2,(H,61,75)(H,63,71)(H,64,69)(H,65,73)(H,66,70)(H,67,72);1-10,24-25H,11H2,(H,26,27)(H,28,29)/t43-,46+,47+,48-,49+,50+,51+;/m1./s1
|
Chemical Name |
(3S,6R,9S,12S,15S,19R,20aS)-6-((1H-indol-3-yl)methyl)-9-(4-aminobutyl)-15-benzyl-12-(4-(benzyloxy)benzyl)-1,4,7,10,13,16-hexaoxo-3-phenylicosahydropyrrolo[1,2-a][1,4,7,10,13,16]hexaazacyclooctadecin-19-yl (2-aminoethyl)carbamate 4,4'-methylenebis(3-hydroxy-2-naphthalenecarboxylate)
|
Synonyms |
Pasireotide pamoate; SOM 230; Pasireotide pamoate; Pasireotide embonate; SOM230 pamoate; SOM-230 pamoate; 396091-79-5; UNII-04F55A7UZ3; 04F55A7UZ3; Cyclo((2S)-2-phenylglycyl-D-tryptophyl-L-lysyl-O-(phenylmethyl)-L-tyrosyl-L-phenylalanyl-(4R)-4-((((2-aminoethyl)amino)carbonyl)oxy)-L-prolyl), 4,4'-methylenebis(3-hydroxy-2-naphthalenecarboxylate) (1:1); SOM230; SOM-230; trade name: Signifor; Signifor LAR.
|
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) |
May dissolve in DMSO (in most cases), if not, try other solvents such as H2O, Ethanol, or DMF with a minute amount of products to avoid loss of samples
|
---|---|
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 | 0.6966 mL | 3.4829 mL | 6.9658 mL | |
5 mM | 0.1393 mL | 0.6966 mL | 1.3932 mL | |
10 mM | 0.0697 mL | 0.3483 mL | 0.6966 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.
Reduction by Pasireotide of the Effluent Volume in High-output Enterostomy in Patients Refractory to Usual Medical Treatment
CTID: NCT02713776
Phase: Phase 2   Status: Completed
Date: 2022-04-04