Size | Price | Stock | Qty |
---|---|---|---|
10mg |
|
||
25mg |
|
||
50mg |
|
||
100mg |
|
||
250mg |
|
||
500mg |
|
||
1g |
|
||
Other Sizes |
|
Purity: ≥98%
Vortioxetine (Lu-AA21004; Lu-AA-21004; Trintellix and Brintellix) is an approved anti-depressant drug acting as an SSRI (selective serotonin reuptake inhibitor) and serotonin receptor modulator. It is a multi-modal serotonergic agent with IC50 values of 15 nM, 33 nM, 3.7 nM, 19 nM, and 1.6 nM, respectively, that inhibits the 5-HT1A, 5-HT1B, 5-HT3A, 5-HT7 receptor and SERT. The FDA authorized vortioxetine, an atypical antidepressant, in 2013 for the treatment of major depressive disorder (MDD) in adults. Vortioxetine is a so-called 'serotonin modulator and stimulator'.
Targets |
sPLA2 ( Ki = 15 nM ); 5-HT3A Receptor ( Ki = 3.7 nM ); Human 5-HT7 Receptor ( Ki = 19 nM ); SERT ( Ki = 1.6 nM )
|
||
---|---|---|---|
ln Vitro |
|
||
ln Vivo |
|
||
Enzyme Assay |
Vortioxetine (Compound 5m) is a multimodal serotonergic agent that inhibits SERT with values of 1.6 nM, 33 nM, 3.7 nM, 19 nM, and 5-HT1A, 5-HT1B, and 5-HT7 receptors, respectively. Vortioxetine exhibits strong suppression of SERT as well as antagonistic effects at 5-HT3A and 5-HT7 receptors, partial agonist effects at 5-HT1B receptors, and agonistic effects at 5-HT1A receptors.
Compound 5m (Lu AA21004) was the lead compound, displaying high affinity for recombinant human 5-HT(1A) (K(i) = 15 nM), 5-HT(1B) (K(i) = 33 nM), 5-HT(3A) (K(i) = 3.7 nM), 5-HT(7) (K(i) = 19 nM), and noradrenergic β(1) (K(i) = 46 nM) receptors, and SERT (K(i) = 1.6 nM). Compound 5m displayed antagonistic properties at 5-HT(3A) and 5-HT(7) receptors, partial agonist properties at 5-HT(1B) receptors, agonistic properties at 5-HT(1A) receptors, and potent inhibition of SERT.[1] Ex vivo SERT and 5-HT3 receptor occupancy assays[2] Brains from mice treated with vehicle, fluoxetine, or vortioxetine (1 h after acute administration or 24 h after the 14th or 21st injection) were flash frozen, sectioned coronally using a cryostat, and then mounted on slides and frozen until use. Slices were 20 μm thick, and began at approximately +1.2 mm anterior from bregma for SERT receptor occupancy or −2.7 mm posterior from bregma for 5-HT3 receptor occupancy determination (Franklin and Paxinos, 2008). Slides were stored for at least 24 h at −20 °C before use in autoradiography experiments. |
||
Cell Assay |
Vortioxetine is a partial h5-HT1B receptor agonist that, in a whole-cell cAMP-based assay, has an EC50 of 460 nM and an intrinsic activity of 22%. In vitro whole-cell cAMP assay, vortioxetine binds to the r5-HT7 receptor with a Kivalue of 200 nM and is a functional antagonist at the r5-HT7 receptor with an IC50 of 2 μM.
Assessment of SERT occupancy[2] Slides were incubated at room temperature for 60 min in buffer (50 mM Tris–HCl, 150 mM NaCl, 5 mM KCl, pH = 7.4) containing 4.5 nM [3H]-escitalopram. Nonspecific binding was determined using 1 μM escitalopram. Slides were washed briefly in cold buffer, dried, and exposed in a Beta imager for 16 h. The region of interest (ROI) for the SERT assay included the lateral and medial septum, the nucleus accumbens and the olfactory tubercle. An example image of the ROI for the SERT assay can be found in Supplementary Fig. 2A. Assessment of 5-HT3 receptor occupancy[2] Slides were preincubated for 5 min in a buffer consisting of 50 mM Tris and 150 mM NaCl. Slides were dried under a stream of air for 30–45 min. Subsequently, slides were incubated at room temperature for 60 min in buffer (50 mM Tris–HCl, 150 mM NaCl, 5 mM KCl, pH = 7.4) containing 1 nM [3H]LY278584. Nonspecific binding was determined using 1 μM ondansetron. Slides were washed briefly in cold buffer, dried, and exposed in a Beta imager for 24 h. The ROI for the 5-HT3 receptor occupancy assay consisted of the hippocampus. An example image for the 5-HT3 receptor occupancy assay can be found in Supplementary Fig. 2B . |
||
Animal Protocol |
|
||
ADME/Pharmacokinetics |
Absorption, Distribution and Excretion
The maximal plasma vortioxetine concentration (Cmax) after dosing is reached within 7 to 11 hours postdose. Absolute bioavailability is 75%. No effect of food on the pharmacokinetics was observed. Following a single oral dose of [14C]labeled vortioxetine, approximately 59% and 26% of the administered radioactivity was recovered in the urine and feces, respectively as metabolites. Negligible amounts of unchanged vortioxetine were excreted in the urine up to 48 hours. The apparent volume of distribution of vortioxetine is approximately 2600 L, indicating extensive extravascular distribution. /MILK/ It is not known whether vortioxetine is present in human milk. Vortioxetine is present in the milk of lactating rats. Vortioxetine is a new multi-modal drug against major depressive disorder with high affinity for a range of different serotonergic targets in the CNS. We report the (11)C-labeling of vortioxetine with (11)C-MeI using a Suzuki-protocol that allows for the presence of an unprotected amine. Preliminary evaluation of (11)C-vortioxetine in a Danish Landrace pig showed rapid brain uptake and brain distribution in accordance with the pharmacological profile, all though an unexpected high binding in cerebellum was also observed. (11)C-vortioxetine displayed slow tracer kinetics with peak uptake after 60 min and with limited wash-out from the brain. Further studies are needed but this radioligand may prove to be a valuable tool in unraveling the clinical effects of vortioxetine. Vortioxetine and related material was mainly excreted by faeces in mice (84%), rats (69%) and dogs (59-65% in two separate studies), whereas humans showed prominent urinary excretion (59%) compared to faeces (26%). In excretion studies, the recovery of (14)C-Vortioxetine and related material was close to 100% in rodents. Dogs and humans exhibited a protracted excretion and the recovery was approximately 90% and 85% after 168 hours and 360 hours, respectively. The objective was to describe the pharmacokinetics of vortioxetine and evaluate the effect of intrinsic and extrinsic factors in the healthy population. Data from 26 clinical pharmacology studies were pooled. A total of 21,758 vortioxetine quantifiable plasma concentrations were collected from 887 subjects with corresponding demography. The doses ranged from 2.5 to 75 mg (single dose) and 2.5-60 mg (multiple QD doses). The pharmacokinetics of vortioxetine was best characterised by a two-compartment model with first-order absorption, lag-time and linear elimination, with interindividual error terms for absorption rate constant, oral clearance and central volume of distribution. The population mean was 32.7 L/hr for oral clearance and 1.97*10(3) L for the central volume of distribution. The average elimination half-life was 65.8 hr. CYP2D6 inferred metabolic status (ultra, extensive, intermediate or poor metabolisers) and age on oral clearance and height on central volume of distribution were identified as statistically significant covariate-parameter relationships. For CYP2D6 poor metabolizers, CL/F was approximately 50% to that seen in CYP2D6 extensive metabolizers. The impact of height on V2/F and age on CL/F was low and not considered to be clinically relevant. The final model was found to be reliable, stable and predictive. A reliable, stable and predictive pharmacokinetic model was developed to characterize pharmacokinetics of vortioxetine in the healthy population. For more Absorption, Distribution and Excretion (Complete) data for VORTIOXETINE (10 total), please visit the HSDB record page. Metabolism / Metabolites Vortioxetine is extensively metabolized primarily through oxidation via cytochrome P450 isozymes CYP2D6, CYP3A4/5, CYP2C19, CYP2C9, CYP2A6, CYP2C8 and CYP2B6 and subsequent glucuronic acid conjugation. CYP2D6 is the primary enzyme catalyzing the metabolism of vortioxetine to its major, pharmacologically inactive, carboxylic acid metabolite, and poor metabolizers of CYP2D6 have approximately twice the vortioxetine plasma concentration of extensive metabolizers. Vortioxetine is extensively metabolized primarily through oxidation via cytochrome P450 isozymes CYP2D6, CYP3A4/5, CYP2C19, CYP2C9, CYP2A6, CYP2C8 and CYP2B6 and subsequent glucuronic acid conjugation. CYP2D6 is the primary enzyme catalyzing the metabolism of vortioxetine to its major, pharmacologically inactive, carboxylic acid metabolite, and poor metabolizers of CYP2D6 have approximately twice the vortioxetine plasma concentration of extensive metabolizers. All metabolites detected in human hepatocytes were also present in dogs, mice and rats (plasma and/or urine) in vivo, except for a glucuronide conjugate of monohydroxy-Vortioxetine which was not found in mice or rats. Among all species tested, rabbit hepatocytes appeared to have the metabolite profile closer to human hepatocyte metabolite profile. Biological Half-Life Mean terminal halflife is approximately 66 hours The oral absolute bioavailability was approximately 10% in the rat, 48% in the dog and 75% in patients, with terminal elimination half-life values of 3.0, 7.9 and 66 hours, respectively. ... Data from 26 clinical pharmacology studies were pooled. A total of 21,758 vortioxetine quantifiable plasma concentrations were collected from 887 subjects with corresponding demography. The doses ranged from 2.5 to 75 mg (single dose) and 2.5-60 mg (multiple QD doses). ... The average elimination half-life was 65.8 hr. ... The mean elimination half-life and oral clearance are 66 hours and 33 L/hr, respectively. |
||
Toxicity/Toxicokinetics |
Toxicity Summary
IDENTIFICATION AND USE: Vortioxetine is a white to very slightly beige powder formulated into film-coated tablets. It is used for the management of major depressive disorders in adults. HUMAN EXPOSURE AND TOXICITY: There is limited clinical trial experience regarding human overdosage with vortioxetine. In pre-marketing clinical studies, cases of overdose were limited to patients who accidentally or intentionally consumed up to a maximum dose of 40 mg of vortioxetine. The maximum single dose tested was 75 mg in men. Ingestion of vortioxetine in the dose range of 40 to 75 mg was associated with increased rates of nausea, dizziness, diarrhea, abdominal discomfort, generalized pruritus, somnolence, and flushing. Toxicity may also occur at therapeutic dosage levels of vortioxetine. Potentially life-threatening serotonin syndrome has been reported with serotonergic antidepressants, including vortioxetine, when used alone, but particularly with concurrent use of other serotonergic drugs (including serotonin (5-hydroxytryptamine; 5-HT) type 1 receptor agonists ("triptans"), tricyclic antidepressants, buspirone, fentanyl, lithium, tramadol, tryptophan, and St. John's wort (Hypericum perforatum)) and with drugs that impair the metabolism of serotonin (particularly monoamine oxidase (MAO) inhibitors, both those used to treat psychiatric disorders and others, such as linezolid and methylene blue). Manifestations of serotonin syndrome may include mental status changes (e.g., agitation, hallucinations, delirium, and coma), autonomic instability (e.g., tachycardia, labile blood pressure, dizziness, diaphoresis, flushing, and hyperthermia), neuromuscular symptoms (e.g., tremor, rigidity, myoclonus, hyperreflexia, and incoordination), seizures, and/or GI symptoms (e.g., nausea, vomiting, and diarrhea). Concurrent or recent (i.e., within 2 weeks) therapy with MAO inhibitors intended to treat psychiatric disorders is contraindicated. Use of an MAO inhibitor intended to treat psychiatric disorders within 3 weeks of vortioxetine discontinuance also is contraindicated. Vortioxetine also should not be initiated in patients who are being treated with other MAO inhibitors such as linezolid or IV methylene blue. If concurrent therapy with vortioxetine and other serotonergic drugs is clinically warranted, the patient should be made aware of the potential increased risk for serotonin syndrome, particularly during initiation of therapy or when dosage is increased. Antidepressants increased the risk of suicidal thoughts and behavior in children, adolescents, and young adults in short-term studies. These studies did not show an increase in the risk of suicidal thoughts and behavior with antidepressant use in patients over age 24; there was a trend toward reduced risk with antidepressant use in patients aged 65 and older. Vortioxetine was not genotoxic in an in vitro chromosome aberration assay in cultured human lymphocytes. ANIMAL STUDIES: The acute oral single dose toxicity of vortioxetine is relatively low with a maximum tolerated dose (MTD) in mice and rats of 300 and 500 mg/kg, respectively. Clinical signs consisted of marked sensitivity to touch and disturbance, rapid breathing, and brown perinasal staining in rats administered 500 mg/kg. In mice, tremors, sensitivity to touch, eyes partly closed, and hypoactivity were seen after 200 and 300 mg/kg, as well as rapid, noisy and/or labored breathing, incoordination, unsteady gait, leaning, salivation, and hyperactivity after 400 and 500 mg/kg. When administered as two vortioxetine doses given an hour apart (200 mg/kg), clinical signs included convulsions, and resulted in death. Carcinogenicity studies were conducted in which mice and rats were given oral doses of vortioxetine up to 50 and 100 mg/kg/day for male and female mice, respectively, and 40 and 80 mg/kg/day for male and female rats, respectively, for 2 years. In rats, the incidence of benign polypoid adenomas of the rectum was statistically significantly increased in females. These were considered related to inflammation and hyperplasia and possibly caused by an interaction with a vehicle component of the formulation used for the study. The finding did not occur in male rats. In mice, vortioxetine was not carcinogenic in males or females. Vortioxetine caused developmental delays when administered during pregnancy to rats and rabbits. Developmental delays were also seen after birth in rats treated with vortioxetine during pregnancy and through lactation. There were no teratogenic effects in rats or rabbits treated with the drug during organogenesis. Treatment of rats with vortioxetine at doses up to 120 mg/kg/day had no effect on male or female fertility. Vortioxetine was not genotoxic in the in vitro bacterial reverse mutation assay (Ames test) and in the in vivo rat bone marrow micronucleus assay. Hepatotoxicity Liver test abnormalities occur in a small proportion of patients ( Likelihood score: E* (unproven but suspected rare cause of clinically apparent liver injury). Effects During Pregnancy and Lactation ◉ Summary of Use during Lactation Amounts of vortioxetine in milk appear to be low. If vortioxetine is required by the mother, it is not a reason to discontinue breastfeeding. However, until more data are available, vortioxetine should be used with careful infant monitoring during breastfeeding. ◉ Effects in Breastfed Infants Three lactating mothers were taking vortioxetine for depression, two were taking 10 mg once daily and one was taking 20 mg once daily. All mothers were exclusively breastfeeding their infants aged 1, 2 and 6 months of age. No mothers reported any unusual behavior in their infants. A woman who was taking a vortioxetine dose of 76.1 mcg/kg daily partially breastfed her infant. She did not observe any adverse effects in her infant. A postpartum Japanese woman with depression was taking vortioxetine 20 mg zolpidem 10 mg, duloxetine 20 mg, rebamipide 100 mg and the Asian herbal medicine Kami-kihi-tou 2.5 grams daily. She partially (over 50%) breastfed her infant for 3 months. The infant had no detectable drug-related adverse effects on routine follow-up at 1, 3, 5, 7 and 9-months postpartum. ◉ Effects on Lactation and Breastmilk Vortioxetine has caused hyperprolactinemia and galactorrhea in some patients. An observational study looked at outcomes of 2859 women who took an antidepressant during the 2 years prior to pregnancy. Compared to women who did not take an antidepressant during pregnancy, mothers who took an antidepressant during all 3 trimesters of pregnancy were 37% less likely to be breastfeeding upon hospital discharge. Mothers who took an antidepressant only during the third trimester were 75% less likely to be breastfeeding at discharge. Those who took an antidepressant only during the first and second trimesters did not have a reduced likelihood of breastfeeding at discharge. The antidepressants used by the mothers were not specified. A retrospective cohort study of hospital electronic medical records from 2001 to 2008 compared women who had been dispensed an antidepressant during late gestation (n = 575) to those who had a psychiatric illness but did not receive an antidepressant (n = 1552) and mothers who did not have a psychiatric diagnosis (n = 30,535). Women who received an antidepressant were 37% less likely to be breastfeeding at discharge than women without a psychiatric diagnosis, but no less likely to be breastfeeding than untreated mothers with a psychiatric diagnosis. None of the mothers were taking vortioxetine. In a study of 80,882 Norwegian mother-infant pairs from 1999 to 2008, new postpartum antidepressant use was reported by 392 women and 201 reported that they continued antidepressants from pregnancy. Compared with the unexposed comparison group, late pregnancy antidepressant use was associated with a 7% reduced likelihood of breastfeeding initiation, but with no effect on breastfeeding duration or exclusivity. Compared with the unexposed comparison group, new or restarted antidepressant use was associated with a 63% reduced likelihood of predominant, and a 51% reduced likelihood of any breastfeeding at 6 months, as well as a 2.6-fold increased risk of abrupt breastfeeding discontinuation. Specific antidepressants were not mentioned. Protein Binding The plasma protein binding of vortioxetine in humans is 98%, independent of plasma concentrations. No apparent difference in the plasma protein binding between healthy subjects and subjects with hepatic (mild, moderate) or renal (mild, moderate, severe, ESRD) impairment is observed. Interactions Potentially serious, sometimes fatal adverse reactions may occur in patients who are receiving or have recently received a monoamine oxidase (MAO) inhibitor and then initiate therapy with serotonergic antidepressants or in those who received SSRI or SNRI therapy shortly before initiation of an MAO inhibitor. Concomitant use of MAO inhibitors intended to treat psychiatric disorders with vortioxetine is contraindicated. In addition, at least 2 weeks should elapse between discontinuance of an MAO inhibitor intended to treat psychiatric disorders and initiation of vortioxetine and at least 3 weeks should elapse between discontinuance of vortioxetine and initiation of an MAO inhibitor intended to treat psychiatric disorders. Concomitant use of vortioxetine and a selective serotonin-reuptake inhibitor (SSRI) or selective serotonin- and norepinephrine-reuptake inhibitor (SNRI) is associated with a risk of serious, sometimes fatal, serotonin syndrome. If concomitant use of vortioxetine and an SSRI or SNRI is clinically warranted, patients should be advised of the increased risk for serotonin syndrome, particularly during treatment initiation and dosage increases. If serotonin syndrome manifestations occur, treatment with vortioxetine and the concurrently administered SSRI or SNRI should be discontinued immediately and supportive and symptomatic treatment should be initiated. Concomitant use of vortioxetine and diuretics may increase the risk of hyponatremia. When vortioxetine is administered concurrently with carbamazepine (a potent CYP inducer) for longer than 14 days, increasing the dosage of vortioxetine should be considered. The manufacturer recommends that the maximum dosage of vortioxetine not exceed 3 times the original dosage. Following discontinuance of carbamazepine, the original vortioxetine dosage should be resumed within 14 days. For more Interactions (Complete) data for VORTIOXETINE (26 total), please visit the HSDB record page. |
||
References |
|
||
Additional Infomation |
Therapeutic Uses
Anti-Anxiety Agents; Serotonin 5-HT1 Receptor Agonists; Serotonin 5-HT1 Receptor Antagonists; Serotonin 5-HT3 Receptor Antagonists; Serotonin Uptake Inhibitors /CLINICAL TRIALS/ ClinicalTrials.gov is a registry and results database of publicly and privately supported clinical studies of human participants conducted around the world. The Web site is maintained by the National Library of Medicine (NLM) and the National Institutes of Health(NIH). Each ClinicalTrials.gov record presents summary information about a study protocol and includes the following: Disease or condition; Intervention (for example, the medical product, behavior, or procedure being studied); Title, description, and design of the study; Requirements for participation (eligibility criteria); Locations where the study is being conducted; Contact information for the study locations; and Links to relevant information on other health Web sites, such as NLM's MedlinePlus for patient health information and PubMed for citations and abstracts for scholarly articles in the field of medicine. Vortioxetine is included in the database. Brintellix is indicated for the treatment of major depressive disorder (MDD). The efficacy of Brintellix was established in six 6 to 8 week studies (including one study in the elderly) and one maintenance study in adults. /Included in US product label/ Drug Warnings /BOX WARNING/ WARNING: SUICIDAL THOUGHTS AND BEHAVIORS. Antidepressants increased the risk of suicidal thoughts and behavior in children, adolescents, and young adults in short-term studies. These studies did not show an increase in the risk of suicidal thoughts and behavior with antidepressant use in patients over age 24; there was a trend toward reduced risk with antidepressant use in patients aged 65 and older. In patients of all ages who are started on antidepressant therapy, monitor closely for worsening, and for emergence of suicidal thoughts and behaviors. Advise families and caregivers of the need for close observation and communication with the prescriber. Brintellix has not been evaluated for use in pediatric patients. Potentially life-threatening serotonin syndrome has been reported with serotonergic antidepressants, including vortioxetine, when used alone, but particularly with concurrent use of other serotonergic drugs (including serotonin (5-hydroxytryptamine; 5-HT) type 1 receptor agonists ("triptans"), tricyclic antidepressants, buspirone, fentanyl, lithium, tramadol, tryptophan, and St. John's wort (Hypericum perforatum)) and with drugs that impair the metabolism of serotonin (particularly monoamine oxidase (MAO) inhibitors, both those used to treat psychiatric disorders and others, such as linezolid and methylene blue). Manifestations of serotonin syndrome may include mental status changes (e.g., agitation, hallucinations, delirium, coma), autonomic instability (e.g., tachycardia, labile blood pressure, dizziness, diaphoresis, flushing, hyperthermia), neuromuscular symptoms (e.g., tremor, rigidity, myoclonus, hyperreflexia, incoordination), seizures, and/or GI symptoms (e.g., nausea, vomiting, diarrhea). Patients receiving vortioxetine should be monitored for the development of serotonin syndrome. Concurrent or recent (i.e., within 2 weeks) therapy with MAO inhibitors intended to treat psychiatric disorders is contraindicated. Use of an MAO inhibitor intended to treat psychiatric disorders within 3 weeks of vortioxetine discontinuance also is contraindicated. Vortioxetine also should not be initiated in patients who are being treated with other MAO inhibitors such as linezolid or IV methylene blue. If concurrent therapy with vortioxetine and other serotonergic drugs is clinically warranted, the patient should be made aware of the potential increased risk for serotonin syndrome, particularly during initiation of therapy or when dosage is increased. If manifestations of serotonin syndrome occur, treatment with vortioxetine and any concurrently administered serotonergic agents should be immediately discontinued and supportive and symptomatic treatment initiated. Serotonergic antidepressants, including vortioxetine, may increase the risk of bleeding events. Concomitant use of aspirin, nonsteroidal anti-inflammatory agents (NSAIAs), warfarin, and other anticoagulants may add to this risk. Case reports and epidemiologic studies have demonstrated an association between use of drugs that interfere with serotonin reuptake and the occurrence of GI bleeding. Bleeding events related to drugs that inhibit serotonin reuptake have ranged from ecchymoses, hematomas, epistaxis, and petechiae to life-threatening hemorrhages. The manufacturer recommends that patients be advised of the increased risk of bleeding associated with concomitant use of vortioxetine and aspirin or other NSAIAs, warfarin, or other drugs that affect coagulation or bleeding. Treatment with serotonergic drugs, including vortioxetine, may result in hyponatremia. In many cases, hyponatremia appears to be due to the syndrome of inappropriate antidiuretic hormone secretion (SIADH). One case of hyponatremia with a serum sodium concentration lower than 110 mmol/L has been reported with vortioxetine in a premarketing study. Geriatric individuals and patients receiving diuretics or who are otherwise volume depleted may be at greater risk of developing hyponatremia with serotonergic antidepressants. Signs and symptoms of hyponatremia include headache, difficulty concentrating, memory impairment, confusion, weakness, and unsteadiness, which may lead to falls; more severe and/or acute cases have been associated with hallucinations, syncope, seizures, coma, respiratory arrest, and death. Vortioxetine should be discontinued and appropriate medical intervention should be instituted in patients with symptomatic hyponatremia. For more Drug Warnings (Complete) data for VORTIOXETINE (18 total), please visit the HSDB record page. Pharmacodynamics Vortioxetine binds with high affinity to the human serotonin transporter (Ki=1.6 nM), but not to the norepinephrine (Ki=113 nM) or dopamine (Ki>1000 nM) transporters. Vortioxetine potently and selectively inhibits reuptake of serotonin by inhibition of the serotonin transporter (IC50=5.4 nM). Specifically, vortioxetine binds to 5HT3 (Ki=3.7 nM), 5HT1A (Ki=15 nM), 5HT7 (Ki=19 nM), 5HT1D (Ki=54 nM), and 5HT1B (Ki=33 nM), receptors and is a 5HT3, 5HT1D, and 5HT7 receptor antagonist, 5HT1B receptor partial agonist, and 5HT1A receptor agonist. |
Molecular Formula |
C18H22N2S
|
|
---|---|---|
Molecular Weight |
298.45
|
|
Exact Mass |
298.15
|
|
Elemental Analysis |
C, 72.44; H, 7.43; N, 9.39; S, 10.74
|
|
CAS # |
508233-74-7
|
|
Related CAS # |
Vortioxetine hydrobromide; 960203-27-4; Vortioxetine-d8; 2140316-62-5; 1253056-29-9 (lactate)
|
|
PubChem CID |
9966051
|
|
Appearance |
Light yellow to yellow solid powder
|
|
Density |
1.2±0.1 g/cm3
|
|
Boiling Point |
424.8±45.0 °C at 760 mmHg
|
|
Flash Point |
210.7±28.7 °C
|
|
Vapour Pressure |
0.0±1.0 mmHg at 25°C
|
|
Index of Refraction |
1.643
|
|
LogP |
4.26
|
|
Hydrogen Bond Donor Count |
1
|
|
Hydrogen Bond Acceptor Count |
3
|
|
Rotatable Bond Count |
3
|
|
Heavy Atom Count |
21
|
|
Complexity |
316
|
|
Defined Atom Stereocenter Count |
0
|
|
SMILES |
S(C1C([H])=C([H])C(C([H])([H])[H])=C([H])C=1C([H])([H])[H])C1=C([H])C([H])=C([H])C([H])=C1N1C([H])([H])C([H])([H])N([H])C([H])([H])C1([H])[H]
|
|
InChi Key |
YQNWZWMKLDQSAC-UHFFFAOYSA-N
|
|
InChi Code |
InChI=1S/C18H22N2S/c1-14-7-8-17(15(2)13-14)21-18-6-4-3-5-16(18)20-11-9-19-10-12-20/h3-8,13,19H,9-12H2,1-2H3
|
|
Chemical Name |
1-[2-(2,4-dimethylphenyl)sulfanylphenyl]piperazine
|
|
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 |
|
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 (8.38 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 (8.38 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 (8.38 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (add these co-solvents sequentially from left to right, and one by one), clear solution. Solubility in Formulation 4: 15% Captisol, pH 9: 10 mg/mL |
Preparing Stock Solutions | 1 mg | 5 mg | 10 mg | |
1 mM | 3.3506 mL | 16.7532 mL | 33.5064 mL | |
5 mM | 0.6701 mL | 3.3506 mL | 6.7013 mL | |
10 mM | 0.3351 mL | 1.6753 mL | 3.3506 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.
Vortioxetine for the Treatment of Hoarding Disorder
CTID: NCT04035850
Phase: Phase 3   Status: Withdrawn
Date: 2024-02-02
![]() |
---|
![]() |
![]() |