yingweiwo

Deferiprone

Alias: Ferriprox
Cat No.:V5309 Purity: ≥98%
Deferiprone (tradename Ferriprox) is novel, potent and orally bioactive iron-chelating drug used for transfusional iron overload clinically.
Deferiprone
Deferiprone Chemical Structure CAS No.: 30652-11-0
Product category: HCV
This product is for research use only, not for human use. We do not sell to patients.
Size Price Stock Qty
5g
10g
25g
50g
100g
200g
Other Sizes

Other Forms of Deferiprone:

  • Deferiprone-d3 (deferiprone d3)
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
Purity & Quality Control Documentation

Purity: ≥98%

Product Description

Deferiprone (tradename Ferriprox) is novel, potent and orally bioactive iron-chelating drug used for transfusional iron overload clinically. It was approved in 1994 for treating thalassaemia major in Europe and Asia. On October 14, 2011, it was approved for use in the US under the FDAÂ’s accelerated approval program. Deferiprone binds to iron and removes it from the blood stream. Deferiprone is used to treat iron overload caused by blood transfusions in people with certain hereditary red blood cell disorders (thalassemia syndrome).

Biological Activity I Assay Protocols (From Reference)
Targets
Free iron chelating agent
ln Vitro
Deferiprone (66-660 μM, 48-96 h) significantly inhibits the growth of 22rv1, Myc-CaP, and TRAMP-C2 cells[1]. Deferiprone (100 μM, for a maximum of 192 hours) prevents TRAMP-C2, Myc-CaP, and 22rv1 cells from migrating[1]. Deferiprone (100 μM, 24 h) decreases m-Acon expression and activity in Myc-CaP, 22rv1, and TRAMP-C2 cells[1].
Deferiprone lowers the free iron in thalassemic red blood cells by up to 1μM over 0.5–24 hours[2].
Deferiprone (10 min) has IC50 values of 0.24, 0.25, 3.36, and 3.73 mM, respectively, and inhibits human platelet aggregation stimulated by AA, ADP, epinephrine, and collagen[3]. With an IC50 value of 0.33 μM, deferiprone (0.1-3.2 μM, 5 mins) inhibits COX-1 activity[3]. The production of cAMP induced by ADP is inhibited by deferiprone (4 mM, 5 min)[3].
In aged fibroblasts, deferiprone (156.25 μg/mL, 24 h) improves survival rate, lowers LDH levels, and exhibits normal cell morphology[4].
Conventional antibiotics' antibacterial activity against S. epidermidis is amplified by deferiprone (25μM, 6 h)[5].
ln Vivo
In the tauopathy model of rTg(tauP301L)4510 mice, deferiprone (100 mg/kg/daily for e.g., 4 weeks) has a neuroprotective effect[6].
Enzyme Assay
Cancer growth and proliferation rely on intracellular iron availability. We studied the effects of Deferiprone (DFP), a chelator of intracellular iron, on three prostate cancer cell lines: murine, metastatic TRAMP-C2; murine, non-metastatic Myc-CaP; and human, non-metastatic 22rv1. The effects of DFP were evaluated at different cellular levels: cell culture proliferation and migration; metabolism of live cells (time-course multi-nuclear magnetic resonance spectroscopy cell perfusion studies, with 1-13 C-glucose, and extracellular flux analysis); and expression (Western blot) and activity of mitochondrial aconitase, an iron-dependent enzyme. The 50% and 90% inhibitory concentrations (IC50 and IC90 , respectively) of DFP for the three cell lines after 48 h of incubation were within the ranges 51-67 μM and 81-186 μM, respectively. Exposure to 100 μM DFP led to: (i) significant inhibition of cell migration after different exposure times, ranging from 12 h (TRAMP-C2) to 48 h (22rv1), in agreement with the respective cell doubling times; (ii) significantly decreased glucose consumption and glucose-driven tricarboxylic acid cycle activity in metastatic TRAMP-C2 cells, during the first 10 h of exposure, and impaired cellular bioenergetics and membrane phospholipid turnover after 23 h of exposure, consistent with a cytostatic effect of DFP. At this time point, all cell lines studied showed: (iii) significant decreases in mitochondrial functional parameters associated with the oxygen consumption rate, and (iv) significantly lower mitochondrial aconitase expression and activity. Our results indicate the potential of DFP to inhibit prostate cancer proliferation at clinically relevant doses and plasma concentrations.[1]
Cell Assay
Cell Line: TRAMP-C2, Myc-CaP, and 22rv1 cells
Concentration: 0, 16, 30, 66, 100, 160, 300, 660 μM
Incubation Time: 48 h, 72 h
Result: exhibited cytostatic activity in three cell lines, with IC50 and IC90 values of roughly 50 and 100 μM, in relation to each other.
Animal Protocol
Animal Model: The rTg(tauP301L)4510 mouse model of tauopathy[6].
Dosage: 100 mg/kg/daily, 4 weeks
Administration: Intragastric administration (i.g.)
Result: enhanced performance on the Y-maze and open field, as well as a 28% reduction in brain iron levels and a decrease in AT8-labeled p-tau in the hippocampus of transgenic tau mice.
References

[1]. Rui V. Simões, Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed 2017 Jun;30(6):10.1002/nbm.3712.

[2]. Deferiprone (L1) Chelates Pathologic Iron Deposits From Membranes of Intact Thalassemic and Sickle Red Blood Cells Both In Vitro and In Vivo. Blood. 1995 Sep 1;86(5):2008-13.

[3]. Antiplatelet activity of deferiprone through cyclooxygenase-1 inhibition. Platelets 2020 May 18;31(4):505-512.

[4]. Deferiprone Stimulates Aged Dermal Fibroblasts via HIF-1α Modulation.Pathog Dis. 2018 Jul 1;76(5).

[5]. Iron chelation destabilizes bacterial biofilms and potentiates the antimicrobial activity of antibiotics against coagulase-negative Staphylococci. Pathogens and Disease, Volume 76, Issue 5, July 2018, fty052

[6]. Deferiprone Treatment in Aged Transgenic Tau Mice Improves Y-Maze Performance and Alters Tau Pathology. Neurotherapeutics. 2021 Apr;18(2):1081-1094.

Additional Infomation
Red blood cell (RBC) membranes from patients with the thalassemic and sickle hemoglobinopathies carry abnormal deposits of iron presumed to mediate a variety of oxidative-induced membrane dysfunctions. We hypothesized that the oral iron chelator deferiprone (L1), which has an enhanced capacity to permeate cell membranes, might be useful in chelating these pathologic iron deposits from intact RBCs. We tested this hypothesis in vitro by incubating L1 with RBCs from 15 patients with thalassemia intermedia and 6 patients with sickle cell anemia. We found that removal of RBC membrane free iron by L1 increased both as a function of time of incubation and L1 concentration. Thus, increasing the time of incubation of thalassemic RBCs with 0.5 mmol/L L1 from 0.5 to 6 hours, enhanced removal of their membrane free iron from 18% +/- 9% to 96% +/- 4%. Dose-response studies showed that incubating thalassemic RBC for 2 hours with L1 concentrations ranging from 0.125 to 0.5 mmol/L resulted in removal of membrane free iron from 28% +/- 15% to 68% +/- 11%. Parallel studies with sickle RBCs showed a similar pattern in time and dose responses. Deferoxamine (DFO), on the other hand, was ineffective in chelating membrane free iron from either thalassemic or sickle RBCs regardless of dose (maximum, 0.333 mmol/L) or time of incubation (maximum, 24 hours). In vivo efficacy of L1 was shown in six thalassemic patients whose RBC membrane free iron decreased by 50% +/- 29% following a 2-week course of L1 at a daily dose of 25 mg/kg. As the dose of L1 was increased to 50 mg/kg/d (n = 5), and then to 75 mg/kg/d (n = 4), 67% +/- 14% and 79% +/- 11%, respectively, of their RBC membrane free iron was removed. L1 therapy--both in vitro and in vivo--also significantly attenuated the malondialdehyde response of thalassemic RBC membranes to in vitro stimulation with peroxide. Remarkably, the heme content of RBC membranes from L1-treated thalassemic patients decreased by 28% +/- 10% during the 3-month study period. These results indicate that L1 can remove pathologic deposits of chelatable iron from thalassemic and sickle RBC membranes, a therapeutic potential not shared by DFO. Furthermore, membrane defects possibly mediated by catalytic iron, such as lipid peroxidation and hemichrome formation, may also be alleviated, at least in part, by L1.[2]
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
C7H9NO2
Molecular Weight
139.15186
Exact Mass
139.06
Elemental Analysis
C, 60.42; H, 6.52; N, 10.07; O, 23.00
CAS #
30652-11-0
Related CAS #
Deferiprone-d3;1346601-82-8
PubChem CID
2972
Appearance
White to off-white solid powder
Density
1.2±0.1 g/cm3
Boiling Point
232.7±40.0 °C at 760 mmHg
Melting Point
272-275 °C
Flash Point
94.5±27.3 °C
Vapour Pressure
0.0±1.0 mmHg at 25°C
Index of Refraction
1.565
LogP
-0.22
tPSA
42.23
SMILES
O=C1C(O)=C(C)N(C)C=C1
InChi Key
TZXKOCQBRNJULO-UHFFFAOYSA-N
InChi Code
InChI=1S/C7H9NO2/c1-5-7(10)6(9)3-4-8(5)2/h3-4,10H,1-2H3
Chemical Name
3-hydroxy-1,2-dimethylpyridin-4(1H)-one
Synonyms
Ferriprox
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)
Water : 3.33~27 mg/mL(~23.93 mM)
DMSO : ~7.14 mg/mL (~51.31 mM)
Solubility (In Vivo)
Solubility in Formulation 1: ≥ 0.71 mg/mL (5.10 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 7.1 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: ≥ 0.71 mg/mL (5.10 mM) (saturation unknown) in 10% DMSO + 90% (20% SBE-β-CD in 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 7.1 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: ≥ 0.71 mg/mL (5.10 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (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 7.1 mg/mL clear DMSO stock solution to 900 μL of corn oil and mix evenly.


Solubility in Formulation 4: 10% DMSO+40% PEG300+5% Tween-80+45% Saline: ≥ 0.71 mg/mL (5.10 mM)

Solubility in Formulation 5: 10 mg/mL (71.86 mM) in PBS (add these co-solvents sequentially from left to right, and one by one), clear solution; with ultrasonication.

 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 7.1865 mL 35.9324 mL 71.8649 mL
5 mM 1.4373 mL 7.1865 mL 14.3730 mL
10 mM 0.7186 mL 3.5932 mL 7.1865 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
NCT Number Recruitment interventions Conditions Sponsor/Collaborators Start Date Phases
NCT02442310 COMPLETEDWITH RESULTS Drug: Deferiprone delayed release tablet formulation
Drug: Deferiprone oral solution
Healthy ApoPharma 2015-05 Phase 1
NCT01835496 COMPLETEDWITH RESULTS Drug: single 1500 mg dose of Ferriprox Sickle Cell Disease ApoPharma 2013-05 Phase 1
NCT01770652 COMPLETEDWITH RESULTS Drug: Deferiprone Renal Impairment ApoPharma 2013-01 Phase 4
NCT01767103 COMPLETEDWITH RESULTS Drug: Ferriprox® Hepatic Impairment ApoPharma 2013-01 Phase 4
NCT02189941 COMPLETEDWITH RESULTS ApoPharma Healthy 公司 2014-05 Phase 1
Biological Data
  • Inhibition of prostate cancer cell proliferation after incubation with different concentrations of DFP. [1].Rui V. Simões, Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed.
  • Inhibition of cell migration due to DFP (100 μM) exposure.[1].Rui V. Simões, Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed.
  • Time-course effect of DFP (100 μM) on live TRAMP-C2 cell metabolism during MR perfusion experiments, as detected by 31P MRS.[1].Rui V. Simões, Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed.
  • Time-course effect of DFP (100 μM) on live TRAMP-C2 cell metabolism during perfusion experiments, as detected by 13C MRS. [1].Rui V. Simões, Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed.
  • Extracellular flux analysis in TRAMP-C2, Myc-CaP and 22rv1 cells incubated with 100 μM DFP for 24 h. [1].Rui V. Simões, Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed.
  • Effect of DFP (100 μM) on m-Acon expression in TRAMP-C2, Myc-CaP and 22rv1 cells after a 24 h incubation period.[1].Rui V. Simões, Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed.
Contact Us