yingweiwo

Leriglitazone

Alias: Leriglitazonum; Leriglitazona; MIN-102; MIN102; Hydroxypioglitazone; 146062-44-4; Leriglitazone; Hydroxy Pioglitazone (M-IV); MIN-102; Leriglitazone [USAN]; K824X25AYA; 2,4-Thiazolidinedione, 5-[[4-[2-[5-(1-hydroxyethyl)-2-pyridinyl]ethoxy]phenyl]methyl]-; Leriglitazone1-Hydroxypioglitazone; MIN 102
Cat No.:V23919 Purity: ≥98%
Leriglitazone (MIN-102; Hydroxypioglitazone), a metabolite of pioglitazone.
Leriglitazone
Leriglitazone Chemical Structure CAS No.: 146062-44-4
Product category: New1
This product is for research use only, not for human use. We do not sell to patients.
Size Price Stock Qty
1mg
5mg
10mg
Other Sizes

Other Forms of Leriglitazone:

  • Leriglitazone HCl
  • Leriglitazone-d4
Official Supplier of:
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Top Publications Citing lnvivochem Products
Product Description
Leriglitazone (MIN-102; Hydroxypioglitazone), a metabolite of pioglitazone. Leriglitazone is a PPARγ agonist that provides high transcriptional potency by stabilizing the PPARγ (AF-2) coactivator binding surface and enhancing coactivator binding capacity. Leriglitazone binds to PPARγC (LBD) with Ki of 1.2 μM and induces PPARγ (LBD) transcription efficiency with EC50 of 680 nM.
Biological Activity I Assay Protocols (From Reference)
Targets
PPARγ LBD (Ki = 1.2 μM); PPARγ LBD (EC50 = 680 nM (PPARγ LBD)
ln Vitro
Pioglitazone (Pio) is an FDA-approved drug for type 2 diabetes that binds and activates the nuclear receptor PPARγ. yet it remains unclear how in vivo Pio metabolites affect PPARγ structure and function. In this study, researchers present a structure-function comparison of Pio and its most abundant in vivo metabolite, Leriglitazone (1-hydroxypioglitazone; PioOH). PioOH displayed a lower binding affinity and reduced potency in coregulator recruitment assays. X-ray crystallography and molecular docking analysis of PioOH-bound PPARγ ligand-binding domain (LBD) revealed an altered hydrogen bonding network, including formation of water-mediated bonds that could underlie its altered biochemical phenotype. NMR spectroscopy and hydrogen/deuterium exchange mass spectrometry (HDX-MS) analysis coupled to activity assays revealed that PioOH better stabilizes the PPARγ activation function-2 (AF-2) coactivator binding surface and better enhances coactivator binding, affording slightly better transcriptional efficacy. These results indicating Pio hydroxylation affects its potency and efficacy as a PPARγ agonist contribute to our understanding of PPARγ-drug metabolite interactions[1].
ln Vivo
In this study, researchers found that leriglitazone improved motor function deficit in YG8sR mice, a FRDA mouse model[https://pubmed.ncbi.nlm.nih.gov/33171227/].
Enzyme Assay
Circular dichroism (CD) spectroscopy[1]
CD wavelength scans and thermal denaturation experiments monitored at 222 nm were performed in CD buffer (10 mM potassium phosphate (pH 7.4) and 50 mM potassium fluoride) to determine the folding and stability of PPARγ LBD (10 µM) in the presence of one molar equivalent of Pio or Leriglitazone (1-hydroxypioglitazone; PioOH) on a Jasco J-815 spectropolarimeter. Thermal denaturation was monitored at 1°C intervals along a temperature gradient from 20°C to 80°C. Raw ellipticity was plotted using GraphPad Prism and fit to a set of equations based on the Gibbs-Helmholtz equation, as previously reported.
TR-FRET competitive ligand displacement and coregulator interaction assays[1]
Time-resolved fluorescence resonance energy transfer (TR-FRET) assays were performed in black low-volume 384-well plate using a buffer containing 20 mM potassium phosphate (pH 7.4), 50 mM potassium chloride, 0.5 mM EDTA, and 5 mM TCEP, and 0.01% Tween-20. For the ligand displacement assay, each well (22.5 μL per well) contained 1 nM 6xHis-PPARγ LBD protein, 1 nM LanthaScreen Elite Tb-anti-His Antibody, and 5 nM Fluormone Pan-PPAR Green tracer ligand in TR-FRET buffer. For the TR-FRET coregulator interaction assay, each well contained 400 nM FITC-labeled TRAP220 or NCoR1 peptides, 4 nM 6xHis-PPARγ LBD protein, 1 nM LanthaScreen Elite Tb-anti-His Antibody, and 400 nM peptide in TR-FRET buffer in 22.5 μL total volume per well. Ligand stocks were prepared via serial dilution in DMSO, added to wells in triplicate to a final DMSO concentration of 1%, and the plates were incubated at 25 °C for ~1 h and read using BioTek Synergy Neo multimode plate reader. The Tb donor was excited at 340 nm; its fluorescence emission was monitored at 495 nm, and the acceptor FITC emission was measured at 520 nm. The TR-FRET ratio was calculated as the signal at 520 nm divided by the signal at 495 nm. Data were plotted using GraphPad Prism and fit to the appropriate equation: for the ligand displacement assay, data were fit to a competitive one site fit Ki equation using the known binding affinity of Fluormone™ Pan-PPAR Green tracer ligand (2.8 nM); and for the coregulator interaction assay data were fit to a sigmoidal dose response equation. For the competitive binding assay, significance was determined by F-test analysis of the Pio vs. Leriglitazone (1-hydroxypioglitazone; PioOH) fit Ki value. For the TR-FRET coregulator recruitment assays, significance was determined by unpaired t-test of EC/IC50 values from n=2 individual experiments.
Fluorescence polarization coregulator interaction assays[1]
Fluorescence polarization assays were performed in black low-volume 384-well plates (Greiner) using a buffer containing 20 mM potassium phosphate (pH 7.4), 50 mM potassium chloride, 0.5 mM EDTA, 5 mM TCEP, and 0.01% Tween-20. Each well contained 100 nM FITC-labeled TRAP220 coactivator peptide, a serial dilution of PPARγ LBD (1.5 nM–90 µM), with a fixed concentration of vehicle control (1% DMSO) or ligand equal to the highest protein concentration (90 µM Pio or Leriglitazone (1-hydroxypioglitazone; PioOH)) in triplicate. Plates were incubated 2 hrs at 4°C and read using BioTek Synergy Neo multimode plate reader. Data were plotted in GraphPad Prism and fit to a sigmoidal dose response equation. Peptide affinity and polarization window from n=2 individual experiments was analyzed by unpaired t-test.
Isothermal titration calorimetry[1]
A peptide derived from the TRAP220 coactivator (residues 638–656; NTKNHPMLMNLLKDNPAQD) was synthesized by Lifetein and resuspended at 500 μM in buffer containing 20 mM potassium phosphate (pH 7.4), 50 mM potassium chloride, 0.5 mM EDTA, and 5 mM TCEP. PPARγ LBD was prepared at 50 μM in identical buffer. 100 μM Pio or Leriglitazone (1-hydroxypioglitazone; PioOH) were added to PPARγ LBD and TRAP220 and incubated on ice 30 minutes before each experiment. TRAP220 peptide (syringe) was titrated into PPARγ LBD (sample cell). 20 total injections were made per experiment (0.4 μL for the first injection, 2.0 μL for subsequent injections), using a mixing speed of 1200 rpm, a reference power of 5 μcal/second, and a cell temperature of 25ºC. Two runs were performed for each ligand-bound condition. Experiments were performed using a MicroCal iTC200 (Malvern). Data were processed in NITPIC 45 to determine binding stoichiometry and further analyzed by unbiased global fitting of both replicate runs per ligand-bound condition in SEDPHAT 46, followed by export to GUSSI for publication-quality figure preparation. The SEDPHAT fitting model used was A + B to AB heteroassociation and the fit parameters were enthalpy (ΔH) and affinity (Kd).
NMR spectroscopy[1]
Two dimensional [1H,15N] -transverse relaxation optimized spectroscopy (TROSY)-heteronuclear single quantum correlation (HSQC) data were collected at 298K using a Bruker 700 Mhz NMR instrument equipped with a QCI cryoprobe. Samples contained 200 µM 15N-labeled PPARγ LBD in a buffer (NMR buffer) containing 20 mM potassium phosphate (pH 7.4), 50 mM potassium chloride, 0.5 mM EDTA, 5 mM TCEP, and 10% D2O in the absence or presence of two molar equivalents of Pio or Leriglitazone (1-hydroxypioglitazone; PioOH). Data were processed and analyzed using Topspin 3.0 (Bruker) and NMRViewJ. NMR analysis was performed using previously described rosiglitazone-bound NMR chemical shift assignments (BMRB entry 17975) for well resolved residues with consistent NMR peak positions via the minimum chemical shift procedure
Cell Assay
Cell-based transactivation assays[1]
HEK293T cells cultured in DMEM media supplemented with 10% fetal bovine serum (FBS) and 50 units mL–1 of penicillin, streptomycin, and glutamine were grown to 90% confluency in a T-75 flask before seeding 4 million cells per well in 10-cm dishes. Seeded cells were transfected using transfection reagent containing 27 µL X-treme Gene 9 in serum-free Opti-mem reduced serum media (Gibco) with either 4.5 µg pCMV6-XL4 plasmid containing full-length human PPARγ2 and 4.5 µg 3X multimerized PPRE-luciferase reporter or 4.5 µg Gal4-PPARγ LBD and 4.5 µg 5X Upstream Activation Sequence (UAS) luciferase reporter. After 18 hrs incubation at 37 °C in a 5% CO2 incubator, the transfected cells were plated in quadruplicate in white 384-well plates at a density of 10,000 cells per well (20 µL volume) and incubated 4 hrs then treated with 20 µL of vehicle control (1% DMSO in DMEM media) or 1:2 serial dilution of each compound from 56 pM–10 µM (1% final DMSO concentration). After 18 hrs, luciferase activity was measured by addition of 20 µL Britelite Plus and luminescence was read using a BioTek Synergy Neo multimode plate reader. Data were plotted in GraphPad Prism as fold change in luminescence of compound-treated cells over DMSO-treated control cells vs ligand concentration and fit to a sigmoidal dose response equation. EC50 values from n=4 individual experiments for full length PPARγ and n=2 individual experiments for PPARγ-Gal4 were analyzed by unpaired t-test; the response window from individual experiments was analyzed by paired t-test.
Animal Protocol
In this study, researchers assess whether MIN-102 (INN: Leriglitazone (1-hydroxypioglitazone; PioOH)), a novel brain penetrant and orally bioavailable PPARγ agonist with an improved profile for central nervous system (CNS) diseases, rescues phenotypic features in cellular and animal models of FRDA. In frataxin-deficient dorsal root ganglia (DRG) neurons, leriglitazone increased frataxin protein levels, reduced neurite degeneration and α-fodrin cleavage mediated by calpain and caspase 3, and increased survival. Leriglitazone also restored mitochondrial membrane potential and partially reversed decreased levels of mitochondrial Na+/Ca2+ exchanger (NCLX), resulting in an improvement of mitochondrial functions and calcium homeostasis. In frataxin-deficient primary neonatal cardiomyocytes, leriglitazone prevented lipid droplet accumulation without increases in frataxin levels. Furthermore, leriglitazone improved motor function deficit in YG8sR mice, a FRDA mouse model. In agreement with the role of PPARγ in mitochondrial biogenesis, leriglitazone significantly increased markers of mitochondrial biogenesis in FRDA patient cells. Overall, these results suggest that targeting the PPARγ pathway by leriglitazone may provide an efficacious therapy for FRDA increasing the mitochondrial function and biogenesis that could increase frataxin levels in compromised frataxin-deficient DRG neurons. Alternately, leriglitazone improved the energy metabolism by increasing the fatty acid β-oxidation in frataxin-deficient cardiomyocytes without elevation of frataxin levels. This could be linked to a lack of significant mitochondrial biogenesis and cardiac hypertrophy. The results reinforced the different tissue requirement in FRDA and the pleiotropic effects of leriglitazone that could be a promising therapy for FRDA.https://pubmed.ncbi.nlm.nih.gov/33171227/
ADME/Pharmacokinetics
Metabolism / Metabolites
5-[(4-{2-[5-(1-hydroxyethyl)pyridin-2-yl]ethoxy}phenyl)methyl]-1,3-thiazolidine-2,4-dione is a known human metabolite of Pioglitazone.
References

[1]. Structural Basis of Altered Potency and Efficacy Displayed by a Major in Vivo Metabolite of the Antidiabetic PPARγ Drug Pioglitazone. J Med Chem. 2019 Feb 28;62(4):2008-2023.

Additional Infomation
Hydroxypioglitazone is a member of the class of thiazolidenediones that is the hydroxy derivative of pioglitazone. It has a role as a human xenobiotic metabolite. It is a member of thiazolidinediones, a member of pyridines and an aromatic ether. It is functionally related to a pioglitazone.
Leriglitazone is under investigation in clinical trial NCT03917225 (A Clinical Study to Evaluate the Effect of MIN-102 on the Progression of Friedreich's Ataxia in Male and Female Patients).
Leriglitazone is an orally bioavailable, blood-brain-barrier (BBB) penetrable, selective peroxisome proliferator-activated receptor (PPAR) subtype gamma agonist, with potential neuroprotective activity that could be used for certain central nervous system (CNS) diseases, such as adrenomyeloneuropathy, cerebral adrenoleukodystrophy (cALD), Friedreich's ataxia, and certain other CNS diseases. Upon oral administration, leriglitazone selectively targets, binds to and activates PPARgamma, thereby regulating the expression of genes involved in mitochondrial biogenesis. This modulates pathways leading to the restoration of mitochondrial function in which dysfunction is caused by the accumulation of very long-chain fatty acids (VLCFAs), and increases energy production, decreases oxidative stress, decreases nuclear factor kappa B (NF-kB) levels, inhibits neuroinflammation, protects the BBB integrity, prevents demyelination and axonal degeneration, increases neuronal survival, increases myelination and oligodendrocyte survival and improves motor function. Mutations in the ABCD1 gene, which encodes the peroxisomal membrane adrenoleukodystrophy protein, cause a defective function of the ABCD1 transporter leading to an accumulation of VLCFA. VLCFA accumulation contributes to membrane destabilization of the myelin sheath, mitochondrial dysfunction, oxidative stress, neuroinflammation and compromised BBB integrity.
Drug Indication
Treatment of adrenoleukodystrophy
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
C19H20N2O4S
Molecular Weight
372.439
Exact Mass
372.114
Elemental Analysis
C, 61.27; H, 5.41; N, 7.52; O, 17.18; S, 8.61
CAS #
146062-44-4
Related CAS #
Leriglitazone hydrochloride;146062-46-6;Leriglitazone-d4;1188263-49-1; 146062-44-4
PubChem CID
4147757
Appearance
White to light yellow solid powder
Density
1.3±0.1 g/cm3
Boiling Point
627.6±55.0 °C at 760 mmHg
Melting Point
157-158ºC
Flash Point
333.4±31.5 °C
Vapour Pressure
0.0±1.9 mmHg at 25°C
Index of Refraction
1.628
LogP
1.11
Hydrogen Bond Donor Count
2
Hydrogen Bond Acceptor Count
6
Rotatable Bond Count
7
Heavy Atom Count
26
Complexity
496
Defined Atom Stereocenter Count
0
InChi Key
OXVFDZYQLGRLCD-RGUGMKFQSA-N
InChi Code
InChI=1S/C19H20N2O4S/c1-12(22)14-4-5-15(20-11-14)8-9-25-16-6-2-13(3-7-16)10-17-18(23)21-19(24)26-17/h2-7,11-12,17,22H,8-10H2,1H3,(H,21,23,24)/t12?,17-/m1/s1
Chemical Name
rac-(5R)-5-{[4-(2-{5-[(1RS)-1-hydroxyethyl]pyridin-2- yl}ethoxy)phenyl]methyl}-1,3-thiazolidine-2,4-dione
Synonyms
Leriglitazonum; Leriglitazona; MIN-102; MIN102; Hydroxypioglitazone; 146062-44-4; Leriglitazone; Hydroxy Pioglitazone (M-IV); MIN-102; Leriglitazone [USAN]; K824X25AYA; 2,4-Thiazolidinedione, 5-[[4-[2-[5-(1-hydroxyethyl)-2-pyridinyl]ethoxy]phenyl]methyl]-; Leriglitazone1-Hydroxypioglitazone; MIN 102
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 Data
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
(e.g. IP/IV/IM/SC)
Injection Formulation 1: DMSO : Tween 80: Saline = 10 : 5 : 85 (i.e. 100 μL DMSO stock solution 50 μL Tween 80 850 μL Saline)
*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 : PEG300Tween 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)]
*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.
Injection Formulation 5: 2-Hydroxypropyl-β-cyclodextrin : Saline = 50 : 50 (i.e. 500 μL 2-Hydroxypropyl-β-cyclodextrin 500 μL Saline)
Injection Formulation 6: DMSO : PEG300 : castor oil : Saline = 5 : 10 : 20 : 65 (i.e. 50 μL DMSO 100 μLPEG300 200 μL castor oil 650 μL Saline)
Injection Formulation 7: Ethanol : Cremophor : Saline = 10: 10 : 80 (i.e. 100 μL Ethanol 100 μL Cremophor 800 μL Saline)
Injection Formulation 8: Dissolve in Cremophor/Ethanol (50 : 50), then diluted by Saline
Injection Formulation 9: EtOH : Corn oil = 10 : 90 (i.e. 100 μL EtOH 900 μL Corn oil)
Injection Formulation 10: EtOH : PEG300Tween 80 : Saline = 10 : 40 : 5 : 45 (i.e. 100 μL EtOH 400 μLPEG300 50 μL Tween 80 450 μL 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
Oral Formulation 4: Suspend in 0.2% Carboxymethyl cellulose
Oral Formulation 5: Dissolve in 0.25% Tween 80 and 0.5% Carboxymethyl cellulose
Oral Formulation 6: Mixing with food powders


Note: Please be aware that the above formulations are for reference only. InvivoChem strongly recommends customers to read literature methods/protocols carefully before determining which formulation you should use for in vivo studies, as different compounds have different solubility properties and have to be formulated differently.

 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 2.6850 mL 13.4250 mL 26.8500 mL
5 mM 0.5370 mL 2.6850 mL 5.3700 mL
10 mM 0.2685 mL 1.3425 mL 2.6850 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.

Calculator

Molarity Calculator allows you to calculate the mass, volume, and/or concentration required for a solution, as detailed below:

  • Calculate the Mass of a compound required to prepare a solution of known volume and concentration
  • Calculate the Volume of solution required to dissolve a compound of known mass to a desired concentration
  • Calculate the Concentration of a solution resulting from a known mass of compound in a specific volume
An example of molarity calculation using the molarity calculator is shown below:
What is the mass of compound required to make a 10 mM stock solution in 5 ml of DMSO given that the molecular weight of the compound is 350.26 g/mol?
  • Enter 350.26 in the Molecular Weight (MW) box
  • Enter 10 in the Concentration box and choose the correct unit (mM)
  • Enter 5 in the Volume box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 17.513 mg appears in the Mass box. In a similar way, you may calculate the volume and concentration.

Dilution Calculator allows you to calculate how to dilute a stock solution of known concentrations. For example, you may Enter C1, C2 & V2 to calculate V1, as detailed below:

What volume of a given 10 mM stock solution is required to make 25 ml of a 25 μM solution?
Using the equation C1V1 = C2V2, where C1=10 mM, C2=25 μM, V2=25 ml and V1 is the unknown:
  • Enter 10 into the Concentration (Start) box and choose the correct unit (mM)
  • Enter 25 into the Concentration (End) box and select the correct unit (mM)
  • Enter 25 into the Volume (End) box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 62.5 μL (0.1 ml) appears in the Volume (Start) box
g/mol

Molecular Weight Calculator allows you to calculate the molar mass and elemental composition of a compound, as detailed below:

Note: Chemical formula is case sensitive: C12H18N3O4  c12h18n3o4
Instructions to calculate molar mass (molecular weight) of a chemical compound:
  • To calculate molar mass of a chemical compound, please enter the chemical/molecular formula and click the “Calculate’ button.
Definitions of molecular mass, molecular weight, molar mass and molar weight:
  • Molecular mass (or molecular weight) is the mass of one molecule of a substance and is expressed in the unified atomic mass units (u). (1 u is equal to 1/12 the mass of one atom of carbon-12)
  • Molar mass (molar weight) is the mass of one mole of a substance and is expressed in g/mol.
/

Reconstitution Calculator allows you to calculate the volume of solvent required to reconstitute your vial.

  • Enter the mass of the reagent and the desired reconstitution concentration as well as the correct units
  • Click the “Calculate” button
  • The answer appears in the Volume (to add to vial) box
In vivo Formulation Calculator (Clear solution)
Step 1: Enter information below (Recommended: An additional animal to make allowance for loss during the experiment)
Step 2: Enter in vivo formulation (This is only a calculator, not the exact formulation for a specific product. Please contact us first if there is no in vivo formulation in the solubility section.)
+
+
+

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.

Clinical Trial Information
A Clinical Study to Assess the Efficacy and Safety of Leriglitazone in Adult Male Subjects With Cerebral Adrenoleukodystrophy
CTID: NCT05819866
Phase: Phase 3
Status: Recruiting
Date: 2024-03-13
A Clinical Study to Evaluate the Efficacy and Safety of MIN-102 (IMP) in Male AMN Patients.
CTID: NCT03231878
Phase: Phase 2/Phase 3
Status: Active, not recruiting
Date: 2023-11-29
A Clinical Study in Male Pediatric Patients With Cerebral X-linked Adrenoleukodystrophy (Cald) to Assess the Effects of MIN-102 Treatment on Disease Progression Prior to Human Stem Cell Transplant (HSCT)
CTID: NCT04528706
Phase: Phase 2
Status: Active, not recruiting
Date: 2023-07-18
A Clinical Study to Evaluate the Effect of MIN-102 on the Progression of Friedreich's Ataxia in Male and Female Patients
CTID: NCT03917225
Phase: Phase 2
Status: Completed
Date: 2022-10-13
An exploratory, open-label, multicenter study in male pediatric patients with cerebral X-linked Adrenoleukodystrophie (cALD) to assess the effect of MIN-102 treatment on the progression of cerebral lesions.
EudraCT: 2019-000654-59
Phase: Phase 2
Status: Ongoing, Trial now transitioned
Date: 2019-06-25
A Double-Blind, Placebo-Controlled Study on the Effects of MIN-102 on Biochemical, Imaging, Neurophysiological, and Clinical Markers in Patients with Friedreich’s Ataxia
EudraCT: 2018-004405-64
Phase: Phase 2
Status: Completed
Date: 2019-03-07
A RANDOMIZED, DOUBLE-BLIND, PLACEBO-CONTROLLED, MULTINATIONAL, MULTICENTER STUDY WITH OPEN-LABEL TREATMENT EXTENSION TO ASSESS THE EFFECT OF MIN-102 ON THE PROGRESSION OF ADRENOMYELONEUROPATHY IN MALE PATIENTS WITH X-LINKED ADRENOLEUKODYSTROPHY
EudraCT: 2017-000748-16
Phase: Phase 2, Phase 3
Status: Ongoing, Prematurely Ended, Completed, GB - no longer in EU/EEA
Date: 2017-09-14
Contact Us