yingweiwo

Hydroxyurea (Hydroxycarbamide)

Alias: NCI-c04831; NSC-32065; NCI c04831; NSC 32065; NCIc04831; NSC32065; Biosupressin; Carbamoyl oxime; Droxia; Hydroxycarbamide
Cat No.:V1464 Purity: ≥98%
Hydroxyurea (formerly known as NCI-c04831, NSC-32065; NCIc04831, NSC32065; Biosupressin; Carbamoyl oxime; Droxia; Hydroxycarbamide) is an approved antineoplastic drug that has been used for treating various cancers such as melanoma, sickle-cell disease and resistant chronic myelocytic leukemia.
Hydroxyurea (Hydroxycarbamide)
Hydroxyurea (Hydroxycarbamide) Chemical Structure CAS No.: 127-07-1
Product category: DNA(RNA) Synthesis
This product is for research use only, not for human use. We do not sell to patients.
Size Price Stock Qty
2g
5g
10g
50g
100g
Other Sizes
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

 

  • Business Relationship with 5000+ Clients Globally
  • Major Universities, Research Institutions, Biotech & Pharma
  • Citations by Top Journals: Nature, Cell, Science, etc.
Top Publications Citing lnvivochem Products
Purity & Quality Control Documentation

Purity: ≥98%

Product Description

Hydroxyurea (formerly known as NCI-c04831, NSC-32065; NCIc04831, NSC32065; Biosupressin; Carbamoyl oxime; Droxia; Hydroxycarbamide) is an approved antineoplastic drug that has been used for treating various cancers such as melanoma, sickle-cell disease and resistant chronic myelocytic leukemia. Through the inhibition of ribonucleoside diphosphate reductase, it inhibits DNA synthesis and induces cell death. A hydroxyurea may prevent HIV-1 from replicating. The 90% inhibitory concentration (IC90) of hydroxyurea for laboratory strains of HIV-1 in activated PBMC has been demonstrated by in vitro experiments to be 0.4 mM.

Biological Activity I Assay Protocols (From Reference)
Targets
HIV-1
ln Vitro

Hydroxyurea can inhibit HIV-1 replication. The 90% inhibitory concentration (IC90) of hydroxyurea for laboratory strains of HIV-1 in activated PBMC has been demonstrated by in vitro experiments to be 0.4 mM. In activated PBMC, hydroxyurea was also found to inhibit HIV-1 replication and work in concert with didanosine, a nucleoside reverse transcriptase inhibitor; this inhibition may be brought about by a decrease in the amounts of deoxynucleoside triphosphate pools. Mutants resistant to didanosine have been shown to become sensitized to hydroxyurea[1][2]. In sickle cell anemia patients, hydroxyurea has been shown to be effective in reducing hemolysis by boosting the fetal hemoglobin production. Ribonucleotide reductase is the rate-limiting enzyme that converts ribonucleotides into deoxyribonucleotides, which are necessary for DNA synthesis. Hydroxyurea inhibits this enzyme to produce its cytostatic effect. Consequently, the S phase of cellular division is halted[1].

ln Vivo
Hydroxyurea treatment consistently lowers WBC and ANC without anemia improvement during a 17-week period. At 50 mg/kg, hydroxyurea creates a decreased white blood cell count, zero neutrophil count, and no improvement in anemia when compared to sickle cell mice receiving a vehicle treatment.
Cell Assay
The same patients' erythroid cells from their peripheral blood are treated with hydroxyurea in vitro one year after they stopped taking it orally for two years, starting at a dose of 5 mg/kg/day for five days a week and increasing to a maximum of 10 mg/kg/day. Thirteen β-Thal/HbE patients receive this treatment.Cells are treated in primary culture for 96 hours with 30 μM hydroxyurea.
Animal Protocol
Mice: SCD mice are given hydroxyurea at doses of 25 mg/kg, 50 mg/kg, and 100 mg/kg, or vehicle, five days a week, to investigate whether hydroxyurea would improve anemia and/or prevent or lessen the development of organ damage in the absence of HbF induction.
ADME/Pharmacokinetics
Absorption, Distribution and Excretion
After oral administration hydroxyurea is readily absorbed from the gastrointestinal tract. Peak plasma concentrations are reached within 2 hours and by 24 hours the serum concentrations are virtually zero. Bioavailability is complete or nearly complete in cancer patients. After oral administration of 20 mg/kg of hydroxyurea, a rapid absorption is observed with peak plasma levels of about 30 mg/L occurring after 0.75 and 1.2 h in children and adult patients with sickle cell syndrome, respectively. The total exposure up to 24 h post-dose is 124 mg.h/L in children and adolescents and 135 mg.h/L in adult patients. The oral bioavailability of hydroxyurea is almost complete as assessed in indications other than sickle cell syndrome. In a comparative bioavailability study in healthy adult volunteers (n=28), 500 mg of hydroxyurea oral solution was demonstrated to be bioequivalent to the reference 500 mg capsule, with respect to both the peak concentration and area under the curve. There was a statistically significant reduction in time to peak concentration with hydroxyurea oral solution compared to the reference 500 mg capsule (0.5 versus 0.75 hours, p = 0.0467), indicating a faster rate of absorption.[L47137 In a study of children with Sickle Cell Disease, liquid and capsule formulations resulted in similar area under the curve, peak concentrations, and half-life. The largest difference in the pharmacokinetic profile was a trend towards a shorter time to peak concentration following ingestion of the liquid compared with the capsule, but that difference did not reach statistical significance (0.74 versus 0.97 hours, p = 0.14).
A significant fraction of hydroxycarbamide is eliminated by nonrenal (mainly hepatic) mechanisms. In adults, the urinary recovery of unchanged drug is reported to be approximately 37% of the oral dose when renal function is normal. In children, the fraction of hydroxyurea excreted unchanged into the urine comprised about 50%.
Hydroxyurea distributes rapidly throughout the human body, enters the cerebrospinal fluid, appears in peritoneal fluid and ascites, and concentrates in leukocytes and erythrocytes. The estimated volume of distribution of hydroxycarbamide approximates total body water. The volume of distribution following oral dosing of hydroxycarbamide is approximately equal to total body water: adult values of 0.48 – 0.90 L/kg have been reported, whilst in children a population estimate of 0.7 L/kg has been reported.
The total body clearance of hydroxyurea in adult patients with Sickle Cell Disease is 0.17 L/h/kg. The respective value in children was similar, 0.22 L/h/kg.
Hydroxyurea is readily absorbed from the GI tract. Peak serum concentrations are attained within 1-4 hours following oral administration. Blood concentrations decline rapidly and there is no cumulative effect with repeated administration. For this reason, higher blood concentrations are attained if the regular dosage is given in a large, single oral dose than if it is administered in divided doses. Disproportionate increases in peak plasma concentrations and areas under the concentration-time curve (AUCs) result when drug dosage is increased. The effect of food on the absorption of hydroxyurea has not been determined.
Hydroxyurea distributes rapidly throughout the body and concentrates in leukocytes and erythrocytes. The estimated volume of distribution of the drug approximates total body water. Hydroxyurea crosses the blood-brain barrier; peak hydroxyurea CSF concentrations are attained within 3 hours following oral administration. The drug distributes into ascites fluid, resulting in drug concentrations in ascites fluid of 2-7.5 times less than plasma drug concentrations.
Studies using(14)C-labeled hydroxyurea indicate that about one-half an orally administered dose is degraded in the liver and is excreted as respiratory carbon dioxide and in urine as urea. The remaining portion of the drug is excreted intact in urine.
About 30-60% of an orally administered dose of hydroxyurea is excreted unchanged by the kidneys, although about 35% is generally excreted.
For more Absorption, Distribution and Excretion (Complete) data for HYDROXYUREA (7 total), please visit the HSDB record page.
Metabolism / Metabolites
Up to 60% of an oral dose undergoes conversion through saturable hepatic metabolism and a minor pathway of degradation to acetohydroxamic acid by urease found in intestinal bacteria.
Studies indicate that up to 50% of an orally administered dose of hydroxyurea is metabolized in the liver; however, the precise metabolic pathways have not been determined. A minor metabolic pathway may involve degradation of the drug by urease, an enzyme produced by intestinal bacteria. Acetohydroxamic acid, possibly resulting from the breakdown of hydroxyurea by urease, was detected in the serum of 3 patients with leukemia treated with hydroxyurea.
Hepatic.
Route of Elimination: Renal excretion is a pathway of elimination.
Half Life: 3-4 hours
Biological Half-Life
In adult cancer patients, hydroxyurea was eliminated with a half-life of approximately 2-3 hours. In a single-dose study in children with Sickle Cell Disease, the mean half-life was reported to be 1.7 hours.
The half-time of hydroxyurea is short, with an initial half-time of 0.63 hr after intravenous administration and 1.78 hr after oral administration and a terminal half-time of 3.32 hr after oral administration and 3.39 hr after intravenous administration /to humans/.
The half-time of hydroxyurea in rats given 137 mg/kg bw per day intraperitoneally on days 9-12 of gestation was 15 min in the dams and 85 min in the embryos. In rhesus monkeys given 100 mg/kg bw per day intravenously on days 23-32 of gestation, the half-time was 120 min after the last injection in the mothers and 265 min in their fetuses.
Toxicity/Toxicokinetics
Toxicity Summary
IDENTIFICATION AND USE: Hydroxyurea is a white, crystalline powder. It is a drug indicated for the following uses: the treatment of resistant chronic myeloid leukemia; treatment of locally advanced squamous cell carcinomas of the head and neck (excluding the lip) in combination with chemoradiation; the palliative treatment of sickle cell anemia generally in patients with recurrent moderate to severe painful crises occurring on at least 3 occasions during the preceding 12 months (designated an orphan drug by the US Food and Drug Administration for this use). HUMAN EXPOSURE AND TOXICITY: During a 2-year period, 26 patients taking hydroxyurea for more than 6 months who had consultations at the dermatology department were systematically examined regarding cutaneous side effects. All but one had cutaneous side-effects, including dryness, moderate alopecia, increased skin pigmentation, melanonychia, cutaneous atrophy, leg ulcers, plantar keratoderma, pseudodermatomyositis, lichen planus-like eruption on the dorsum of the hands actinic keratosis, squamous cell carcinomas, and mouth ulcerations. Hepatotoxicity and hepatic failure resulting in death have been reported during postmarketing surveillance in patients with HIV infection treated with hydroxyurea and other antiretroviral drugs. Fatal hepatic failure were reported most often in patients treated with the combination of hydroxyurea, didanosine, and stavudine (note: hydroxyurea is not indicated for the treatment of HIV infection). In one case of an 85-year-old man taking hydroxyurea for chronic myelomonocytic leukemia, severe interstitial pneumonitis was induced by the drug. Gangrene of the toes and digits is a rare but very severe complication of long-term hydroxyurea therapy. A retrospective review of four adult men who had semen analysis during hydroxyurea therapy and in three cases after its cessation, suggests that the drug generally reduces sperm counts and motility and results in abnormal morphology. Hydroxyurea induced DNA hypermethylation in normal human embryonic lung fibroblasts and their simian virus 40-transformed counterparts. While cancer has been observed in some patients receiving long-term treatment with hydroxyurea, the drug is not yet classifiable as to its carcinogenicity to humans. ANIMAL STUDIES: Symptoms of exposure in dogs have included vomiting, ataxia, methemoglobinemia, tachycardia, lethargy, and hypothermia. Groups of 50 mice of each sex were treated intraperitoneally with hydroxyurea starting at two days of age and then at weekly intervals for one year. The incidences of pulmonary tumors were 30/50 (60%) in control and 16/35 (46%) in treated mice. Hydroxyurea is a swiftly acting developmental toxicant that inhibits DNA synthesis and is teratogenic in all mammals studied. Data from a study on mice suggest that hydroxyurea compromises folliculogenesis and the ability of generated embryos to develop. A group of 27 pregnant golden hamsters received an intravenous injection of 50 mg/kg bw hydroxyurea on day 8 of pregnancy. A high rate of fetal death and malformations, especially of the central nervous system, was observed. Hydroxyurea caused cell transformation in mass cultures of embryonic cells from mice, but not in cultures derived from two other strains of mice, nor in BALB/c 3T3 cells. Hydroxyurea treatment led to hypermethylation of DNA in hamster fibrosarcoma cells. Hydroxyurea was inactive as either a frameshift or base-pair substitution mutagen in Salmonella typhimurium strains TA1537, TA1535, TA98 and TA100, and addition of an exogenous metabolic activation system did not affect these results. Hydroxyurea induced SOS repair in Escherichia coli K12 cells. In various Saccharomyces cerevisiae strains, hydroxyurea induced mitotic crossing over, mitotic gene conversion, intrachromosomal recombination and aneuploidy, but not petite mutations. It also increased the frequency of ultraviolet-induced mitotic gene conversion and induced recombination in dividing but not G1 or G2 arrested cells of the RS112 strain of yeast. In meiotic yeast cells, hydroxyurea increased the frequency of meiotic recombination. Hydroxyurea induced micronuclei in the bone marrow of non-tumor-bearing male NMRI mice but did not induce micronucleated cells in female C57BL/6 C3H/He hybrid mice, although it produced sperm abnormalities in male mice of this strain.
Hydroxyurea is converted to a free radical nitroxide (NO) in vivo, and transported by diffusion into cells where it quenches the tyrosyl free radical at the active site of the M2 protein subunit of ribonucleotide reductase, inactivating the enzyme. The entire replicase complex, including ribonucleotide reductase, is inactivated and DNA synthesis is selectively inhibited, producing cell death in S phase and synchronization of the fraction of cells that survive. Repair of DNA damaged by chemicals or irradiation is also inhibited by hydroxyurea, offering potential synergy between hydroxyurea and radiation or alkylating agents. Hydroxyurea also increases the level of fetal hemoglobin, leading to a reduction in the incidence of vasoocclusive crises in sickle cell anemia. Levels of fetal hemoglobin increase in response to activation of soluble guanylyl cyclase (sGC) by hydroxyurea-derived NO.
Toxicity Data
Oral, mouse: LD50 = 7330 mg/kg; Oral, rat: LD50 = 5760 mg/kg
Interactions
The purpose of this analysis is to investigate if the combination of didanosine and stauvudine, with or without hydroxyurea, has a higher incidence of neuropathy than a single drug regimen. Data were obtained from patients followed longitudinally by the Johns Hopkins AIDS Services. Incidence rates of development of neuropathy were calculated for each of five regimens: didanosine (+/- hydroxyurea), didanosine + stauvudine (+/- hydroxyurea), and stauvudine. Cox proportional hazard regression was used to compare the relative risk of neuropathy for each regimen adjusting for CD4 cell count, other drugs received, and time on therapy. A total of 1116 patients received at least one of the five regimens. There were 117 cases of neuropathy. The crude incidence rate of neuropathy ranged from 6.8 cases per 100 person-years for didanosine to 28.6 cases per 100 person-years for didanosine + stauvudine + hydroxyurea. Compared with didanosine alone, and adjusting for CD4 cell counts and other variables, the relative risk of neuropathy was 1.39 [95% confidence interval (CI): 0.84-2.32] for stauvudine alone, 2.35 (95% CI: 0.69-8.07) for didanosine + hydroxyurea, 3.50 (95% CI: 1.81-6.77) for didanosine + stauvudine, and 7.80 (95% CI: 3.92-15.5) for didanosine + stauvudine + hydroxyurea. Based on the data, the risk of neuropathy is additive or even synergistic for didanosine + stauvudine + hydroxyurea compared with didanosine or stauvudine alone. The combination of didanosine + stauvudine also increases the risk of neuropathy but less than when hydroxyurea is included.
Concomitant therapy with hydroxyurea and other myelosuppressive agents or radiation therapy may increase the likelihood of bone marrow depression or other adverse effects, and dosage adjustment may be required.
Because hydroxyurea therapy may cause increased serum uric acid concentrations, dosage adjustment of uricosuric medication may be required.
The antitumor activity of hydroxyurea can be potentiated by the addition of iron-chelating agents and prevented by the addition of Fe2+ to the medium.
For more Interactions (Complete) data for HYDROXYUREA (7 total), please visit the HSDB record page.
References

[1]. Clin Infect Dis . 2000 Jun:30 Suppl 2:S193-7.

[2]. Clin Infect Dis . 2000 Jun:30 Suppl 2:S143-50.

[3]. Exp Hematol . 2005 Dec;33(12):1486-92.

[4]. Gene Ther . 2002 Aug;9(15):1023-30.

Additional Infomation
Therapeutic Uses
Antineoplastic Agents; Antisickling Agents; Enzyme Inhibitors; Nucleic Acid Synthesis 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. Hydroxyurea is included in the database.
Hydroxyurea Capsules USP are indicated for the treatment of: Resistant chronic myeloid leukemia. Locally advanced squamous cell carcinomas of the head and neck (excluding the lip) in combination with chemoradiation. /Included in US product label/
Hydroxyurea has been used in the treatment of psoriasis and is reportedly beneficial in the treatment of hypereosinophilic syndrome that does not respond to corticosteroid therapy. /NOT included in US product label/
For more Therapeutic Uses (Complete) data for HYDROXYUREA (12 total), please visit the HSDB record page.
Drug Warnings
Hydroxyurea is a highly toxic drug with a low therapeutic index, and a therapeutic response is not likely to occur without some evidence of toxicity. Hydroxyurea therapy may be complicated by severe, sometimes life-threatening or fatal, adverse effects. The drug must be used only under constant supervision by clinicians experienced in therapy with cytotoxic agents or the use of this agent for sickle cell anemia.
Hydroxyurea should be admin with caution to patients who have recently received other cytotoxic drugs or irradiation therapy, since bone marrow depression is likely in these patients. In addition, an exacerbation of post-irradiation erythema may occur.
Hepatotoxicity, in some cases resulting in fatal hepatic failure, has been reported in patients with HIV infection receiving hydroxyurea in combination with antiretroviral agents. Fatal hepatotoxicity occurred most frequently in patients receiving combination therapy with hydroxyurea, didanosine, and stavudine. Elevation of serum concentrations of hepatic enzymes has been reported in patients receiving hydroxyurea.
Cutaneous vasculitic toxicities, including vasculitic ulcerations and gangrene, have occurred in patients receiving hydroxyurea for myeloproliferative disorders, particularly in patients who have received or who are receiving interferon therapy.
For more Drug Warnings (Complete) data for HYDROXYUREA (37 total), please visit the HSDB record page.
Pharmacodynamics
The correlation between hydroxyurea concentrations, reduction of crisis rate, and increase in HbF, is not known.
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
CH4N2O2
Molecular Weight
76.05
Exact Mass
76.027
Elemental Analysis
C, 15.79; H, 5.30; N, 36.83; O, 42.07
CAS #
127-07-1
Related CAS #
127-07-1
PubChem CID
3657
Appearance
White to off-white solid powder
Density
1.5±0.1 g/cm3
Boiling Point
222.1±23.0 °C at 760 mmHg
Melting Point
135-140 °C
Flash Point
88.1±22.6 °C
Vapour Pressure
0.0±1.0 mmHg at 25°C
Index of Refraction
1.501
LogP
-1.8
Hydrogen Bond Donor Count
3
Hydrogen Bond Acceptor Count
2
Rotatable Bond Count
0
Heavy Atom Count
5
Complexity
42.9
Defined Atom Stereocenter Count
0
SMILES
O([H])N([H])C(N([H])[H])=O
InChi Key
VSNHCAURESNICA-UHFFFAOYSA-N
InChi Code
InChI=1S/CH4N2O2/c2-1(4)3-5/h5H,(H3,2,3,4)
Chemical Name
hydroxyurea
Synonyms
NCI-c04831; NSC-32065; NCI c04831; NSC 32065; NCIc04831; NSC32065; Biosupressin; Carbamoyl oxime; Droxia; Hydroxycarbamide
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)
DMSO: 15~50 mg/mL (197.2~657.5 mM)
Water: ~15 mg/mL (~197.2 mM)
Ethanol: <1 mg/mL
Solubility (In Vivo)
Solubility in Formulation 1: ≥ 2.5 mg/mL (32.87 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 (32.87 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 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 (32.87 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 25.0 mg/mL clear DMSO stock solution to 900 μL of corn oil and mix evenly.


Solubility in Formulation 4: 100 mg/mL (1314.92 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 13.1492 mL 65.7462 mL 131.4924 mL
5 mM 2.6298 mL 13.1492 mL 26.2985 mL
10 mM 1.3149 mL 6.5746 mL 13.1492 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
Addition of JSP191 (C-kit Antibody) to Nonmyeloablative Hematopoietic Cell Transplantation for Sickle Cell Disease and Beta-Thalassemia
CTID: NCT05357482
Phase: Phase 1/Phase 2    Status: Recruiting
Date: 2024-12-02
Bomedemstat vs Hydroxyurea for Essential Thrombocythemia (MK-3543-007)
CTID: NCT06456346
Phase: Phase 3    Status: Recruiting
Date: 2024-11-27
Effects of Hydroxyurea and Metformin in Transfusion Dependent Beta-Thalassemia
CTID: NCT06606886
Phase: Phase 4    Status: Active, not recruiting
Date: 2024-11-22
Dosing Study of Radiation Combined With Tislelizumab and Pamiparib in Patients With Previously Treated Head and Neck Cancer
CTID: NCT05526924
Phase: Phase 1    Status: Recruiting
Date: 2024-11-22
Evaluation of HU-resistance in Adult Patients With Polycythemia Vera Who Meet PV-AIM Predictors
CTID: NCT05853458
Phase: Phase 4    Status: Recruiting
Date: 2024-11-18
View More

MITHRIDATE: Ruxolitinib Versus Hydroxycarbamide or Interferon as First Line Therapy in High Risk Polycythemia Vera
CTID: NCT04116502
Phase: Phase 3    Status: Recruiting
Date: 2024-11-18


Cerebrovascular Reserve and White Matter Disease in Patients with Chronic Anemia
CTID: NCT03715972
Phase:    Status: Completed
Date: 2024-10-30
A Phase 2 Open-label Study to Evaluate the Activity of Etavopivat on Transcranial Doppler Velocities in Pediatric Patients With Sickle Cell Disease Who Are at Increased Risk for Primary Stroke
CTID: NCT05953584
Phase: Phase 2    Status: Recruiting
Date: 2024-10-29
Chemotherapy and Locoregional Therapy Trial (Surgery or Radiation) for Patients With Head and Neck Cancer
CTID: NCT03107182
Phase: Phase 2    Status: Completed
Date: 2024-10-15
Hydroxyurea and EPO in Sickle Cell Disease
CTID: NCT05451940
Phase: Phase 1/Phase 2    Status: Active, not recruiting
Date: 2024-09-26
Nab-paclitaxel and Carboplatin Followed by Response-Based Local Therapy in Treating Patients With Stage III or IV HPV-Related Oropharyngeal Cancer
CTID: NCT02258659
Phase: Phase 2    Status: Completed
Date: 2024-09-24
A Trial for Prevention of Recurrent Ischemic Priapism in Men With Sickle Cell Disease: A Pilot Study
CTID: NCT05142254
Phase: Phase 2    Status: Completed
Date: 2024-09-20
Luspatercept With or Without Hydroxyurea for the Treatment of Myelodysplastic/Myeloproliferative Neoplasms With Ring Sideroblasts and Thrombocytosis or Unclassifiable With Ring Sideroblasts
CTID: NCT05005182
Phase: Phase 2    Status: Terminated
Date: 2024-09-19
Pharmacokinetics, Efficacy and Safety of Twice Daily Dosing Regimen of Hydroxycarbamide Dispersible Tablets in Children With Sickle Cell Disease
CTID: NCT06578507
Phase: Phase 2    Status: Not yet recruiting
Date: 2024-08-29
Stroke Prevention in Nigeria 2 Trial
CTID: NCT06526117
Phase: Phase 4    Status: Recruiting
Date: 2024-08-29
Study on Efficacy and Safety of Givinostat Versus Hydroxyurea in Patients With Polycythemia Vera
CTID: NCT06093672
Phase: Phase 3    Status: Recruiting
Date: 2024-08-28
ESCORT-HU Extension: European Sickle Cell Disease Cohort - Hydroxyurea - Extension Study
CTID: NCT04707235
Phase:    Status: Active, not recruiting
Date: 2024-08-27
SIKAMIC (SIklos on Kidney Function and AlbuMInuria Clinical Trial)
CTID: NCT03806452
Phase: Phase 2    Status: Completed
Date: 2024-08-27
T-Cell Depleted Alternative Donor Bone Marrow Transplant for Sickle Cell Disease (SCD) and Other Anemias
CTID: NCT03653338
Phase: Phase 1/Phase 2    Status: Recruiting
Date: 2024-08-09
Nonmyeloablative Haploidentical Peripheral Blood Mobilized Hematopoietic Precursor Cell Transplantation for Sickle Cell Disease
CTID: NCT03077542
Phase: Phase 1/Phase 2    Status: Active, not recruiting
Date: 2024-08-07
Paclitaxel Albumin-Stabilized Nanoparticle Formulation and Carboplatin Followed By Chemoradiation in Treating Patients With Recurrent Head and Neck Cancer
CTID: NCT01847326
Phase: Phase 1    Status: Completed
Date: 2024-07-31
Pharmacokinetics and Safety of Commonly Used Drugs in Lactating Women and Breastfed Infants
CTID: NCT03511118
Phase:    Status: Recruiting
Date: 2024-07-24
Hydroxyurea to Prevent Brain Injury in Sickle Cell Disease
CTID: NCT01389024
Phase: Phase 2    Status: Completed
Date: 2024-07-10
Mobile-Directly Observed Therapy on Adherence to Hydroxyurea
CTID: NCT02844673
Phase: Phase 2    Status: Completed
Date: 2024-06-12
Isatuximab in Combination With Chemotherapy in Pediatric Patients With Relapsed/Refractory Acute Lymphoblastic Leukemia or Acute Myeloid Leukemia
CTID: NCT03860844
Phase: Phase 2    Status: Terminated
Date: 2024-05-16
Promoting Utilization and Safety of Hydroxyurea Using Precision in Africa
CTID: NCT05285917
Phase: Phase 3    Status: Recruiting
Date: 2024-04-18
Reduced Intensity Conditioning for Non-Malignant Disorders Undergoing UCBT, BMT or PBSCT
CTID: NCT01962415
Phase: Phase 2    Status: Recruiting
Date: 2024-04-02
Management of Severe Acute Malnutrition in SCD, in Northern Nigeria
CTID: NCT03634488
Phase: Phase 2    Status: Completed
Date: 2024-03-27
Study of Stem Cell Transplant vs. Non-Transplant Therapies in High-Risk Myelofibrosis
CTID: NCT04217356
Phase:    Status: Recruiting
Date: 2024-03-22
Comparison Of Efficacy And Safety Of Thalidomide Vs Hydroxyurea In Thalassemia Patients: A Single-Centre Pilot Study.
CTID: NCT06239389
Phase: Phase 2    Status: Completed
Date: 2024-03-12
Efficacy of Combination of Hdroxyurea and Thalidomide Over Either Hydroxyurea or Thalidomide Alone in the Treatment of Transfusion Dependent Thalassemia in Children: A Quasi-Randomised Clinical Trial
CTID: NCT06299670
Phase: Phase 4    Status: Recruiting
Date: 2024-03-08
SACRED A Prospective Research Study to Reduce Stroke in Children With Sickle Cell Anemia
CTID: NCT02769845
Phase: N/A    Status: Active, not recruiting
Date: 2024-02-06
A Multicenter Study of Intensive Concomitant Chemoradiotherapy With Filgrastim (GCSF) for Patients With Locoregionally Advanced Head and Neck Cancer
CTID: NCT01693718
Phase: Phase 2    Status: Completed
Date: 2024-01-29
EXTEND EXpanding Treatment for Existing Neurological Disease
CTID: NCT02556099
Phase: Phase 2    Status: Active, not recruiting
Date: 2024-01-18
Alternative Dosing And Prevention of Transfusions (ADAPT)
CTID: NCT05662098
PhaseEarly Phase 1    Status: Recruiting
Date: 2024-01-10
Lung and Bone Marrow Transplantation for Lung and Bone Marrow Failure
CTID: NCT03500731
Phase: Phase 1/Phase 2    Status: Recru
A prospective open label, pharmacokinetic study of an oral hydroxyurea solution in children with sickle cell anemia
CTID: null
Phase: Phase 2    Status: GB - no longer in EU/EEA
Date: 2018-11-30
Phase 3 Study (PACIFICA): A Randomized, Controlled Phase 3 Study of Pacritinib Versus Physician’s Choice in Patients with Primary Myelofibrosis, Post Polycythemia Vera Myelofibrosis, or Post-Essential Thrombocythemia Myelofibrosis with Severe Thrombocytopenia (Platelet Counts <50,000/µL)
CTID: null
Phase: Phase 3    Status: GB - no longer in EU/EEA, Completed
Date: 2017-10-23
A Phase 2a, Randomised, Double-Blind, Placebo-Controlled Study of IMR-687 in Adult Patients with Sickle Cell Anaemia (Homozygous HbSS or Sickle-β0 Thalassemia)
CTID: null
Phase: Phase 2    Status: GB - no longer in EU/EEA
Date: 2017-10-19
Evaluation of the impact of renal function on the pharmacokinetics of hydroxyurea (Siklos®) in patients with sickle cell disease with normal renal function, with hyperfiltration, or with renal failure.
CTID: null
Phase: Phase 4    Status: Completed
Date: 2015-02-06
A Randomized Phase III study of Decitabine (DAC) with or without Hydroxyurea (HY) versus HY in patients with advanced proliferative Chronic Myelomonocytic Leukemia (CMML)
CTID: null
Phase: Phase 3    Status: Ongoing, Completed
Date: 2014-08-12
A randomized, open-label, multicenter, controlled, parallel arm, phase III study assessing the efficacy and safety of AOP2014 vs. Hydroxyurea in patients with Polycythemia Vera
CTID: null
Phase: Phase 3    Status: Completed
Date: 2013-08-15
Polycythemia Vera Symptom Study Evaluating Ruxolitinib Versus Hydroxyurea in a Randomized, Multicenter, Double-Blind, Double-Dummy, Phase 3 Efficacy and Safety Study of Patient Reported Outcomes
CTID: null
Phase: Phase 3    Status: Completed
Date: 2012-11-06
Randomized Trial of Pegylated Interferon Alfa-2a versus Hydroxyurea Therapy in the Treatment of High Risk Polycythemia Vera and High Risk Essential Thrombocythemia
CTID: null
Phase: Phase 3    Status: Ongoing, Prematurely Ended, Completed
Date: 2012-02-15
The induction of apoptosis by anti-psoriatic treatments
CTID: null
Phase: Phase 4    Status: Completed
Date: 2011-11-08
Risk-adapted, MRD-directed therapy for young adults with newly diagnosed acute myeloid leukemia.
CTID: null
Phase: Phase 2    Status: Completed
Date: 2011-10-26
DALIAH
CTID: null
Phase: Phase 3    Status: Ongoing
Date: 2011-09-12
Randomized, open label, multicenter phase III study of Efficacy and Safety in Polycythemia vera subjects who are resistant to or intolerant of hydroxyurea: JAK iNhibitor INC424 tablets versus best available care (The RESPONSE Trial)
CTID: null
Phase: Phase 3    Status: Completed
Date: 2011-02-08
Devenir de la spermatogénèse chez les hommes drépanocytaires traités médicalement (HYDREP).
CTID: null
Phase: Phase 3    Status: Ongoing
Date: 2010-04-20
Essai de phase 2-3 randomisé, comparant deux modalités de réirradiation après chirurgie en territoire irradié des carcinomes des voies aéro-digestives supérieures :
CTID: null
Phase: Phase 2, Phase 3    Status: Ongoing
Date: 2010-02-04
STUDY OF HYDROXYUREA MECHANISMS OF ACTION ON LEUKOCYTE ACTIVATION IN PATIENTS WITH CHRONIC MYELOPROLIFERATIVE SYNDROMES.
CTID: null
Phase: Phase 4    Status: Completed
Date: 2009-07-08
Temozolomide in the chemo-immuno-therapy of refractory acute leukaemia of adult patients: The TRIAC protocol
CTID: null
Phase: Phase 3    Status: Prematurely Ended
Date: 2009-05-26
Imatinib piu` idrossiurea nel trattamento dei meningiomi recidivati o in progressione: studio randomizzato di fase II.
CTID: null
Phase: Phase 2    Status: Prematurely Ended
Date: 2009-05-08
A large-scale trial testing the intensity of CYTOreductive therapy to prevent cardiovascular events In patients with Polycythemia Vera (PV) – CYTO-PV.
CTID: null
Phase: Phase 3    Status: Prematurely Ended
Date: 2008-05-23
A randomized double-blind dose-finding multi-centre phase IIa study with VS411 for HIV-1 infection.
CTID: null
Phase: Phase 2    Status: Completed
Date: 2008-02-07
A randomised trial to compare ASPIRIN versus HYDROXYUREA/ASPIRIN in 'intermediate risk' primary thrombocythaemia and ASPIRIN only with observation in 'Low risk' primary thrombocythaemia.
CTID: null
Phase: Phase 3    Status: Completed
Date: 2007-07-05
Glivec® (imatinib mesylate)/Litalir® (hydroxyurea) plus initial radiotherapy after surgery in patients with newly diagnosed glioblastoma multiforme followed by Glivec® and Litalir® – A phase I/II safety evaluation study.
CTID: null
Phase: Phase 2    Status: Completed
Date: 2007-02-09
Estudio fase II, abierto, multicéntrico, no comparativo, que evalúa la eficacia de Glivec® con Hidroxiurea en pacientes con Glioblastoma Multiforme (GBM) en progresión, que no reciben fármacos anticonvulsivantes con capacidad de inducción enzimática
CTID: null
Phase: Phase 2    Status: Prematurely Ended, Completed
Date: 2006-01-17
Estudio fase II, abierto, multicéntrico, no comparativo, que evalúa la eficacia de Glivec® con Hidroxiurea en pacientes con Glioblastoma Multiforme (GBM) en progresión, que reciben fármacos anticonvulsivantes con capacidad de inducción enzimática
CTID: null
Phase: Phase 2    Status: Prematurely Ended, Completed
Date: 2006-01-17
A Phase IIIb, randomised, open label study to compare the safety, efficacy and tolerability of anagrelide hydrochloride versus hydroxyurea in high-risk essential thrombocythaemia patients.
CTID: null
Phase: Phase 3    Status: Completed
Date: 2005-07-07
Glivec (imatinib mesylate) in combination with hydroxyurea or hydroxyurea alone as an oral therapy in temozolomide resistant progressive glioblastoma patients
CTID: null
Phase: Phase 3    Status: Prematurely Ended
Date: 2004-12-21
A SINGLE BLIND, MULTI-CENTE, RANDOMIZED MULTINATIONAL PHASE III STUDY TO COMPARE THE EFFICACY AND TOLERABILITY OF ANAGRELIDE VS HYDROXYUREA IN PATIENTS WITH ESSENTIAL THROMBOCYTHAEMIA
CTID: null
Phase: Phase 3    Status: Completed
Date: 2004-07-12
Evaluation systématique des troubles respiratoires au cours du sommeil chez l'enfant drépanocytaire : apport du traitement par Hydroxycarbamide lors de l'identification d'une hypoxémie isolée - essai clinique multicentrique randomisé contre placebo, double aveugle
CTID: null
Phase: Phase 2    Status: Completed
Date:
An open-label, non-comparative, multicentre study to evaluate the acceptability of a new paediatric formulation of hydroxycarbamide in children with sickle cell disease
CTID: null
Phase: Phase 2    Status: Completed
Date:

Biological Data
  • Effect of hydroxyurea treatment on fractional fetal hemoglobin (HbF) (high-performance liquid chromatography analysis), fold induction of Gγ:Aγ-globin mRNA (real-time one-step quantitative competitive reverse transcriptase-polymerase chain reaction), and Gγ:Aγ-globin ratio (Triton X-100 acid urea polyacrylamide gel electrophoresis) in β-thalassemia/hemoglobin E (β-thal/HbE) patients in vitro vs in vivo. Exp Hematol . 2005 Dec;33(12):1486-92.
Contact Us