yingweiwo

Zidovudine (Azidothymidine)

Alias: Azidothymidine, NSC-602670, NSC602670,NSC 602670,Zidovudinu;BW A509U; Azidothymidine; 30516-87-1; 3'-Azido-3'-deoxythymidine; Retrovir; AZT; Thymidine, 3'-azido-3'-deoxy-; Zidovudinum; BWA 509U; BWA-509U; BWA509U; ZDV; AZT; Retrovir.
Cat No.:V1823 Purity: ≥98%
Zidovudine (also known as ZDV, Azidothymidine; Retrovir; Zidovudinum, NSC 602670) is a nucleoside analogue reverse transcriptase inhibitor (NRTI) which used to treat and prevent HIV.
Zidovudine (Azidothymidine)
Zidovudine (Azidothymidine) Chemical Structure CAS No.: 30516-87-1
Product category: Reverse Transcriptase
This product is for research use only, not for human use. We do not sell to patients.
Size Price Stock Qty
250mg
500mg
1g
2g
5g
10g
Other Sizes

Other Forms of Zidovudine (Azidothymidine):

  • Zidovudine O-β-D-glucuronide (3'-Azido-3'-deoxythymidine β-D-glucuronide)
  • Zidovudine diphosphate
  • AZT triphosphate
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

Zidovudine (also known as ZDV, Azidothymidine; Retrovir; Zidovudinum, NSC 602670) is a nucleoside analogue reverse transcriptase inhibitor (NRTI) which used to treat and prevent HIV. Zidovudine is the first effective agent for the management of HIV-1 infection and is approved by FDA as a drug for AIDs in 1987. As a nucleoside analogue, zidovudine inhibits the activity of the reverse transcriptase with its triphosphate structure. It can also be used to prevent HIV transmission, such as from mother to child during the period of birth or after a needle stick injury. Used by itself in HIV-infected patients, AZT slows HIV replication in patients, but does not stop it entirely.

Biological Activity I Assay Protocols (From Reference)
Targets
HIV reverse transcriptase (NRTI)
ln Vitro
With EC50s of 17, 1311, 8, and 5 nM, respectively, zidovudine inhibits SVG, primary human fetal astrocytes (PFA), peripheral blood mononuclear cells (PBMC), and monocyte-derived macrophages (MDM). With EC90 values of 0.205 μM, 44.157 μM, 0.481 μM, and 0.219 μM, respectively, zidovudine inhibits SVG, PFA, PBMC, and MDM [1]. CRISPR/Cas9 genome editing has emerged as a dependable and efficient technique for precisely altering specific regions of the genome in living cells. CXCR4 is regarded as a key therapeutic target for AIDS since it functions as a coreceptor for HIV-1 infection. By attaching itself to the envelope protein gp120, CXCR4 facilitates viral entrance into human CD4+ cells. CRISPR/Cas9-mediated genome editing effectively disrupts the human CXCR4 gene, leading to HIV-1 resistance in human primary CD4+ T cells. High specificity and minimal off-target effects are displayed by Cas9-mediated CXCR4 ablation, which has no effect on cell division or proliferation [2].
ln Vivo
In wild-type mice, laser-induced choroidal neovascularization (CNV) was inhibited by NRTIs lamivudine (3TC), zidovudine (AZT), or abacavir (ABC) in comparison to the PBS vehicle. Day 3 following laser injury marked the peak of mean VEGF-A levels in the RPE/choroid. The eyes of mice treated with 3TC, AZT, and ABC had significantly lower levels of this protein than the wild-type mice's control eyes, but not the P2rx7-/- mice. [3].
Enzyme Assay
Production and quantitation of Env-pseudotyped luciferase reporter viruses[1]
Env-pseudotyped luciferase reporter viruses were produced by transfection of 293T cells with pCMVΔP1ΔenvpA, pHIV-1Luc, and either pcDNA3-VSVg or pSVIII-YU2 Env plasmids using Lipofectamine 2000 (Invitrogen) at a ratio of 1∶3∶1, as described previously. Viruses pseudotyped with the CCR5-using HIV-1 YU-2 envelope glycoproteins were used for infections of PBMC and MDM, whereas SVG cells and PFA were infected with viruses pseudotyped with the vesicular stomatitis virus G protein (VSV-G) in order to achieve sufficient levels of viral entry for the inhibition assays. The supernatants containing virus pseudotypes were harvested 48 h later, filtered through 0.45 µm filters, titrated on each of the different cell types (TCID50 values were calculated), and stored at −80°C.
Cell Assay
Cell viability assay[1]
ARV cytotoxicity was assessed in all cell types at 72 h post-drug exposure using the CellTitre-Glo Luminescent Cell Viability Assay (Promega, USA), according to the manufacturer's protocol.[1]
Virus inhibition assays[1]
Assays were performed in all cell types in the presence of titrating concentrations of ARV. 5,000 SVG, 2,500 PFA, 200,000 PBMC, or 50,000 MDM cells/well were seeded into triplicate wells of 96-well plates. Twenty-four hours later, the culture medium was removed and replaced with medium containing the ARV or DMSO (0.5% vol/vol), and equivalent TCID50 infectious units of luciferase reporter virus were added to the cells. After a 16 h incubation at 37°C, the initial viral inoculum was removed and replaced with culture medium containing the same ARV or DMSO (0.5% vol/vol) concentrations. At 72 h post infection, the medium was aspirated, the cells were lysed and HIV-1 infection measured using the Luciferase Assay System (Promega) according to manufacturer's instructions. Luminescence was measured using a FLUOStar Optima microplate reader (BMG Labtech, Germany). Inhibition curves and the 50% (EC50) and 90% (EC90) effective concentrations were determined by nonlinear regression analysis as previously described, using GraphPad Prism software.[1]
Animal Protocol
C57BL/6J (wild-type) and P2rx7-/- mice are used. The Nlrp3-/- mice are used. The NRTIs 3TC, AZT, and ABC or the P2X7 antagonist A438079 hydrochloride are dissolved in PBS. For CNV, each group of mice is injected once with 1 μL of NRTIs (3TC, 125 ng/μL; ABC, 183 ng/μL; AZT, 146 ng/μL), 1 μL of A438079 hydrochloride (3, 30, or 300 ng/μL), or the same volume of vehicle (PBS) into the vitreous humor using a 33-gauge needle immediately after laser injury. Another group of mice is injected with 3TC (125 ng) in combination with an anti-mouse VEGF polyclonal antibody (10 ng). Goat whole IgG (10 ng) is used as a biological control for the anti-mouse VEGF antibody. .
Mice
ADME/Pharmacokinetics
Absorption, Distribution and Excretion
Rapid and nearly complete absorption from the gastrointestinal tract following oral administration; however, because of first-pass metabolism, systemic bioavailability of zidovudine capsules and solution is approximately 65% (range, 52 to 75%). Bioavailability in neonates up to 14 days of age is approximately 89%, and it decreases to approximately 61% and 65% in neonates over 14 days of age and children 3 months to 12 years, respectively. Administration with a high-fat meal may decrease the rate and extent of absorption.
As in adult patients, the major route of elimination was by metabolism to GZDV. After intravenous dosing, about 29% of the dose was excreted in the urine unchanged and about 45% of the dose was excreted as GZDV.
Apparent volume of distribution, HIV-infected patients, IV administration = 1.6 ± 0.6 L/kg
0.65 +/- 0.29 L/hr/kg [HIV-infected, Birth to 14 Days of Age]
1.14 +/- 0.24 L/hr/kg [HIV-infected, 14 Days to 3 Months of Age]
1.85 +/- 0.47 L/hr/kg [HIV-infected, 3 Months to 12 Years of Age]. The transporters, ABCB1, ABCC4, ABCC5, and ABCG2 are involved with the clearance of zidovudine.
In patients with impaired renal function, plasma concentrations of zidovudine may be increased and the half-life prolonged. In one study in adults with impaired renal function (creatinine clearances ranging from 6-31 ml/minute) without HIV infections beta half life of zidovudine averaged 1.4 hours and was similar to that reported for adults with HIV infections who had normal renal function. However, the beta half life of glucuronide in these adults with impaired renal function averaged 8 hours and was considerably prolonged compared with that reported for adults with HIV infections who had normal renal function. In one study in adults with hemophilia and HIV infections who had elevated serum concentrations of aspartate aminotransferase (serum glutamic-oxaloacetic transaminase), alanine aminotransferase (serum glutamic-pyruvic transaminase), pharmacokinetics of zidovudine after a single 300 mg oral dose showed considerable interindividual variation.
Following oral administration of zidovudine in patients with HIV infections, 63-95% of the dose is excreted in urine; approximately 14-18% of the dose is excreted as unchanged zidovudine and 72-74% is excreted as zidovudine 5'-O-glucuronide within 6 hours. Following iv administration of the drug in adults or children with HIV infections, approximately 18-29% of the dose is excreted in urine as unchanged drug and 45-60% is excreted as zidovudine 5'-O-glucuronide within 6 hours.
Zidovudine and 3'-azodp-3'-deoxy-5'-O-beta-d-glucopyranuronosylthymidine are eliminated principally in urine via both glomerular filtration and tubular secretion. Following oral or IV administration in adults with HIV infection, total body clearance of zidovudine averages 1.6 l/hr per kg (range: 0.8-2.7 l/hr per kg) and renal clearance of the drug averages 0.34 l/hr per kg. In children 3 months to 12 years of age, the total body clearance averaged 1.85 l/hr per kg. In one limited study in neonates and infants younger than 3 months of age, total body clearance of the drug averaged 0.65 l/hr per kg in those 14 days of age or younger and 1.14 l/hr per kg in those older than 14 days of age.
Zidovudine is rapidly metabolized via glucuronidation in the liver to zidovudine 5'-O-glucuronide (GAZT); the metabolite has an apparent elimination half life of 1 hour (range: 0.6-1.7 hours). Zidovudine 5'-O-glucuronide does not appear to have antiviral activity against HIV.
For more Absorption, Distribution and Excretion (Complete) data for ZIDOVUDINE (21 total), please visit the HSDB record page.
Metabolism / Metabolites
Hepatic. Metabolized by glucuronide conjugation to major, inactive metabolite, 3′-azido-3′-deoxy-5′- O-beta-D-glucopyranuronosylthymidine (GZDV). UGT2B7 is the primary UGT isoform that is responsible for glucuronidation. Compared to zidovudine, GZDV's area under the curve is approximately 3-fold greater. The cytochrome P450 isozymes are responsible for the reduction of the azido moiety to form 3'-amino-3'- deoxythymidine (AMT).
Zidovudine is rapidly metabolized via glucuronidation in the liver principally to 3-azido-3-deoxy-5-O-beta-d-glucopyranuronosylthymidine (GZDV; formerly GAZT); zidovudine is also metabolized to GZDV in renal microsomes. GZDV has an apparent elimination half-life of 1 hour (range: 0.6-1.7 hours) and does not appear to have antiviral activity against HIV. In addition, two other hepatic metabolites of zidovudine have been identified as 3-amino-3-deoxythymidine (AMT) and its glucuronide derivative (GAMT). Intracellularly, in both virus-infected and uninfected cells, zidovudine is converted to zidovudine monophosphate by cellular thymidine kinase; the monophosphate derivative is phosphorylated to zidovudine diphosphate via cellular dTMP kinase (thymidylate kinase) and then to zidovudine triphosphate via other cellular enzymes. Intracellular (host cell) conversion of zidovudine to the triphosphate derivative is necessary for the antiviral activity of the drug. Activation for antibacterial action, however, does not depend on phosphorylation within host cells but rather depends on conversion within bacterial cells.
The mechanisms of intestinal mucosal transport and metabolism of zidovudine and other thymidine analogs were studied. No zidovudine metabolites appeared in any part of the gastrointestinal tract. Other thymidine analogs were rapidly metabolized in the upper gastrointestinal tract, but not in the colon.
Hepatic. Metabolized by glucuronide conjugation to major, inactive metabolite, 3′-azido-3′-deoxy-5′- O-beta-D-glucopyranuronosylthymidine (GZDV). UGT2B7 is the primary UGT isoform that is responsible for glucuronidation. Compared to zidovudine, GZDV's area under the curve is approximately 3-fold greater. The cytochrome P450 isozymes are responsible for the reduction of the azido moiety to form 3'-amino-3'- deoxythymidine (AMT).
Route of Elimination: As in adult patients, the major route of elimination was by metabolism to GZDV. After intravenous dosing, about 29% of the dose was excreted in the urine unchanged and about 45% of the dose was excreted as GZDV.
Half Life: Elimination half life, HIV-infected patients, IV administration = 1.1 hours (range of 0.5 - 2.9 hours)
Biological Half-Life
Elimination half life, HIV-infected patients, IV administration = 1.1 hours (range of 0.5 - 2.9 hours)
The plasma half-life of zidovudine in adults averages approximately 0.53 hours following oral or IV administration. Following IV administration of zidovudine in adults or children, plasma concentrations of the drug appear to decline in a biphasic manner. Half-life in adults is less than 10 minutes in the initial phase and 1 hour in the terminal phase. Following IV administration over 1 hour of a single 80-, 120-, or 160-mg/sq m , dose in children 1-13 years of age with symptomatic HIV infection, the alpha half-life of zidovudine averaged 0.16-0.25 hours and the beta half-life averaged 1-1.7 hours. Plasma half-life of zidovudine generally is longer in neonates than in older children and adults but decreases with neonatal maturity. In one limited study in neonates and infants younger than 3 months of age, plasma half-life of zidovudine averaged 3.1 hours in those 14 days of age or younger and 1.9 hours in those older than 14 days of age. In a study in premature neonates (26-32 weeks gestation; birthweight 0.7-1.9 kg), the serum half-life of zidovudine averaged 7.3 hours at an average postnatal age of 6.3 days and averaged 4.4 hours at an average postnatal age of 17.7 days.
The value for half-life of zidovudine is 1-2 hr.
Toxicity/Toxicokinetics
Toxicity Summary
Zidovudine, a structural analog of thymidine, is a prodrug that must be phosphorylated to its active 5ду_-triphosphate metabolite, zidovudine triphosphate (ZDV-TP). It inhibits the activity of HIV-1 reverse transcriptase (RT) via DNA chain termination after incorporation of the nucleotide analogue. It competes with the natural substrate dGTP and incorporates itself into viral DNA. It is also a weak inhibitor of cellular DNA polymerase ‘± and ‘_.
Toxicity Data
Male rat(po): LD50 = 3.1 g/kg
Female rat(po): LD50 = 3.7 g/kg
Male mice: LD50 = 3.6 g/kg
Female mice(po): LD50 = 3.1 g/kg
LD50 is 3084 mg/kg (orally in mice).
Interactions
Concomitant use of probenecid may produce substantially higher and prolonged serum concentrations of zidovudine.
In at least one study, concomitant use of acetaminophen reportedly resulted in an increased risk of granulocytopenia in patients receiving zidovudine; this potentiation of hematologic toxicity appeared to correlate with the duration of acetaminophen use.
Neurotoxicity (profound drowsiness and lethargy), which recurred on rechallenge, has been reported in at least one HIV-infected patient who received acyclovir and zidovudine concomitantly. Neurotoxicity was evident within 30-60 days after initiation of IV acyclovir therapy, persisted with some improvement when acyclovir was administered orally, and resolved following discontinuance of acyclovir in this patient. Acyclovir and zidovudine have been used concomitantly in other HIV-infected patients without evidence of increased toxicity. Although the clinical importance is unclear, there is some evidence that acyclovir may potentiate the antiretroviral effect of zidovudine in vitro; acyclovir alone has only minimal antiretroviral activity.
Both ganciclovir and zidovudine alone produce direct, dose-dependent inhibitory effects on myeloid and erythroid progenitor cells, and combined use of the drugs increases the risk of hematologic toxicity and may result in additive or synergistic myelotoxic effects. In several studies in patients with AIDS and cytomegalovirus infections, profound, intolerable myelosuppression, evidenced principally as severe neutropenia, occurred in all patients receiving ganciclovir (5 mg/kg iv 1-4 times daily) concomitantly with zidovudine (200 mg orally every 4 hours); anemia also occurred in many of these patients. Severe hematologic toxicity, which required a reduction in zidovudine dosage, also occurred in more than 80% of patients receiving ganciclovir (5 mg/kg iv 1-2 times daily) concomitantly with zidovudine (100 mg orally every 4 hours).
For more Interactions (Complete) data for ZIDOVUDINE (18 total), please visit the HSDB record page.
Non-Human Toxicity Values
LD50 Rat iv > 750 mg/kg
LD50 Mice iv > 3000 mg/kg
References

[1]. The NRTIs lamivudine, stavudine and zidovudine have reduced HIV-1 inhibitory activity in astrocytes. PLoS One. 2013 Apr 16;8(4):e62196.

[2]. Genome editing of CXCR4 by CRISPR/cas9 confers cells resistant to HIV-1 infection. Sci Rep. 2015 Oct 20;5:15577.

[3]. Nucleoside Reverse Transcriptase Inhibitors Suppress Laser-Induced Choroidal Neovascularization in Mice. Invest Ophthalmol Vis Sci. 2015 Nov;56(12):7122-9.

Additional Infomation
Therapeutic Uses
Anti-HIV Agents; Antimetabolites; Antimetabolites, Antineoplastic; Reverse Transcriptase Inhibitors
Zidovudine is indicated in combination with other antiretroviral agents for the treatment of HIV infection. ... /Included in US product labeling/
Zidovudine is indicated for the prevention of mother-to-child transmission of HIV-1 infection as part of a regimen that includes oral zidovudine beginning between 14 and 34 weeks gestation, continuous intravenous infusion of zidovudine during labor, and administration of zidovudine syrup to the neonate for the first 6 weeks of life. However, transmission to infants may still occur in some cases despite the use of this regimen. /Included in US product labeling/
Zidovudine has been used prophylactically in health care workers at risk of acquiring HIV infection after occupational exposure to the virus. Risk of transmission from a single needlestick is approximately 0.3%. Efficacy, and optimal dose and duration of prophylactic treatment are unknown at this time; however, HIV infection has occurred in persons who received zidovudine prophylaxis after a needlestick or other parenteral exposure. /NOT included in US product labeling/
For more Therapeutic Uses (Complete) data for ZIDOVUDINE (7 total), please visit the HSDB record page.
Drug Warnings
Adverse systemic effects reported with IV zidovudine are similar to those reported with oral zidovudine. However, clinical experience with IV zidovudine has been more limited than experience with oral zidovudine and the drug has generally been administered IV only for short periods of time. Long-term IV zidovudine therapy (i.e., longer than 2-4 weeks) has not been evaluated in adults and may enhance adverse hematologic effects.
The most common adverse effects of zidovudine are hematologic effects (i.e., anemia, neutropenia), nausea, and headache. Because HIV-infected patients receiving zidovudine generally have serious underlying disease with multiple baseline symptomatology and clinical abnormalities and because many adverse effects that occurred in zidovudine-treated patients also occurred in patients receiving placebo, many reported effects may not be directly attributable to zidovudine. The frequency and severity of adverse effects associated with use of zidovudine in adults are greater in patients with more advanced disease at the time of initiation of therapy. In one study in asymptomatic patients receiving 100 mg of the drug orally 5 times daily for an average of longer than 1 year (range: 4 months to 2 years), only nausea occurred more frequently in patients receiving zidovudine than in those receiving placebo. Adverse effects reported with use of zidovudine in women, IV drug users, and racial minorities are similar to those reported with use of the drug in white males.
Four patients with the acquired immunodeficiency syndrome, and a history of Pneumocystis carinii pneumonia developed severe pancytopenia (hemoglobin, less than 85 g/l; granulocytes, less than or equal to 0.5 X 10(9)/L; platelets, less than or equal to 30 X 10(9)/L) 12 to 17 weeks after the initiation of azidothymidine (AZT) therapy. The bone marrow was markedly hypocellular in three patients and moderately hypocellular in the fourth. Partial bone marrow recovery was documented within 4 to 5 weeks in three patients, but no marrow recovery has yet occurred in one patient during the more than 6 months since AZT treatment was discontinued.
Hematologic toxicity is causally related to zidovudine therapy, being directly related to dosage and duration of therapy with the drug, and has been reported most frequently in patients with advanced symptomatic HIV infection or low pretreatment hemoglobin concentrations, neutrophil counts, and helper/inducer (CD4+, T4+) T-cell counts. Patients with low serum folate or vitamin B12 concentrations may be at increased risk for developing bone marrow toxicity during zidovudine therapy. There also are limited data suggesting that bone marrow of patients with fulminant acquired immunodeficiency syndrome (AIDS) may be more sensitive to zidovudine-induced toxicity than that of patients with less advanced disease (e.g., AIDS-related complex (ARC)).
For more Drug Warnings (Complete) data for ZIDOVUDINE (43 total), please visit the HSDB record page.
Pharmacodynamics
Zidovudine is a nucleoside reverse transcriptase inhibitor (NRTI) with activity against Human Immunodeficiency Virus Type 1 (HIV-1). Zidovudine is phosphorylated to active metabolites that compete for incorporation into viral DNA. They inhibit the HIV reverse transcriptase enzyme competitively and act as a chain terminator of DNA synthesis. The lack of a 3'-OH group in the incorporated nucleoside analogue prevents the formation of the 5' to 3' phosphodiester linkage essential for DNA chain elongation, and therefore, the viral DNA growth is terminated.
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
C10H13N5O4
Molecular Weight
267.24
Exact Mass
267.096
Elemental Analysis
C, 44.94; H, 4.90; N, 26.21; O, 23.95
CAS #
30516-87-1
Related CAS #
117675-21-5 (glucuronide); 106060-89-3 (diphosphate); 92586-35-1 (triphosphate)
PubChem CID
35370
Appearance
White to off-white solid
Melting Point
113-115 °C(lit.)
Index of Refraction
47 ° (C=1, H2O)
LogP
-0.53
Hydrogen Bond Donor Count
2
Hydrogen Bond Acceptor Count
6
Rotatable Bond Count
3
Heavy Atom Count
19
Complexity
484
Defined Atom Stereocenter Count
3
SMILES
O=C(C(C)=CN1[C@@H](C2)O[C@@H]([C@H]2N=[N+]=[N-])CO)NC1=O
InChi Key
HBOMLICNUCNMMY-XLPZGREQSA-N
InChi Code
InChI=1S/C10H13N5O4/c1-5-3-15(10(18)12-9(5)17)8-2-6(13-14-11)7(4-16)19-8/h3,6-8,16H,2,4H2,1H3,(H,12,17,18)/t6-,7+,8+/m0/s1
Chemical Name
1-[(2R,4S,5S)-4-azido-5-(hydroxymethyl)oxolan-2-yl]-5-methylpyrimidine-2,4-dione
Synonyms
Azidothymidine, NSC-602670, NSC602670,NSC 602670,Zidovudinu;BW A509U; Azidothymidine; 30516-87-1; 3'-Azido-3'-deoxythymidine; Retrovir; AZT; Thymidine, 3'-azido-3'-deoxy-; Zidovudinum; BWA 509U; BWA-509U; BWA509U; ZDV; AZT; Retrovir.
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:53 mg/mL (198.3 mM)
Water:53 mg/mL (198.3 mM)
Ethanol: N/A
Solubility (In Vivo)
Solubility in Formulation 1: ≥ 2.5 mg/mL (9.35 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 (9.35 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 (9.35 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: 18 mg/mL (67.4 mM)

Solubility in Formulation 5: 20 mg/mL (74.84 mM) in PBS (add these co-solvents sequentially from left to right, and one by one), clear solution; with ultrasonication (<60°C).

 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 3.7420 mL 18.7098 mL 37.4195 mL
5 mM 0.7484 mL 3.7420 mL 7.4839 mL
10 mM 0.3742 mL 1.8710 mL 3.7420 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
Natural History Study of the KSHV Inflammatory Cytokine Syndrome (KICS)
CTID: NCT01419561
Phase: Phase 2    Status: Recruiting
Date: 2024-12-02
Virotherapy and Natural History Study of KHSV-Associated Multricentric Castleman s Disease With Correlates of Disease Activity
CTID: NCT00092222
Phase: Phase 2    Status: Active, not recruiting
Date: 2024-11-25
Belinostat Therapy With Zidovudine for Adult T-Cell Leukemia-Lymphoma
CTID: NCT02737046
Phase: Phase 2    Status: Active, not recruiting
Date: 2024-08-19
A Study to Evaluate the Pharmacokinetics, Safety, Tolerability, and Antiviral Activity of Rilpivirine (TMC278) in Human Immunodeficiency Virus Infected Adolescents and Children Aged Greater Than or Equal to 6 Years
CTID: NCT00799864
Phase: Phase 2    Status: Completed
Date: 2024-06-25
Inhibition of Reverse Transcription in Aicardi-Goutières Syndrome
CTID: NCT04731103
Phase: Phase 2    Status: Completed
Date: 2024-05-09
View More

Early Infant HIV Treatment in Botswana
CTID: NCT02369406
Phase: Phase 2/Phase 3    Status: Active, not recruiting
Date: 2023-11-09


Antiretroviral Regime for Viral Eradication in Newborns
CTID: NCT02712801
Phase: Phase 4    Status: Completed
Date: 2022-09-28
Study of HIV-Infected and Uninfected Pregnant Woman/Child Dyads in Gaborone, Botswana
CTID: NCT03088410
Phase: Phase 4    Status: Completed
Date: 2022-07-20
Evaluating Strategies to Reduce Mother-to-Child Transmission of HIV Infection in Resource-Limited Countries
CTID: NCT01061151
Phase: Phase 3    Status: Completed
Date: 2022-02-11
Safety and Pharmacokinetic Study of Fixed Dose Combination of Zidovudine, Lamivudine, and Nevirapine in HIV-Infected Children in Thailand
CTID: NCT00672412
Phase: Phase 1/Phase 2    Status: Completed
Date: 2021-11-09
Reducing the Incidence of Nevirapine Resistance Mutations in Pregnant HIV Infected Women Who Receive Anti-HIV Drugs Prior to and After Giving Birth
CTID: NCT00109590
Phase: Phase 2    Status: Completed
Date: 2021-11-05
A Study of Foscarnet in the Treatment of HIV Infection in Patients Who Have Taken Zidovudine for a Long Time
CTID: NCT00001002
Phase: Phase 1    Status: Completed
Date: 2021-11-04
A Study of Pentamidine in the Prevention of Pneumocystis Carinii Pneumonia
CTID: NCT00001003
Phase: N/A    Status: Completed
Date: 2021-11-04
Antiviral Activity of and Resistance to Lamivudine in Combination With Zidovudine, Stavudine, or Didanosine
CTID: NCT00000838
Phase: Phase 2    Status: Completed
Date: 2021-11-04
Randomized, Double-Blind, Placebo-Controlled Trial of Nimodipine for the Neurological Manifestations of HIV-1
CTID: NCT00000738
Phase: Phase 1    Status: Completed
Date: 2021-11-04
A Study of Three Anti-HIV Drug Combinations in Patients Who Have Taken Amprenavir
CTID: NCT00001095
Phase: Phase 2    Status: Completed
Date: 2021-11-04
The Safety and Effectiveness of Injections of Human Recombinant Interferon-gamma in Patients With AIDS Who Have Taken Zidovudine
CTID: NCT00001112
Phase: Phase 1    Status: Completed
Date: 2021-11-04
A Randomized, Comparative Trial of Zidovudine (AZT) Versus 2',3'-Didehydro-3'-Deoxythymidine (Stavudine; d4T) in Children With HIV Infection
CTID: NCT00000789
Phase: Phase 2    Status: Completed
Date: 2021-11-04
A Study of AZT in HIV-Infected Patients With AIDS-Related Kaposi's Sarcoma
CTID: NCT00000994
Phase: Phase 3    Status: Completed
Date: 2021-11-04
A Comparison of Zidovudine (AZT) Used Alone or in Combination With Didanosine (ddI) or Dideoxycytidine (ddC) in HIV-Infected Patients
CTID: NCT00001022
Phase: Phase 3    Status: Completed
Date: 2021-11-04
Effectiveness of AZT and Nevirapine in Preventing HIV Transmission From Ugandan Mothers to Their Newborns
CTID: NCT00006396
Phase: Phase 3    Status: Completed
Date: 2021-11-04
The Safety and Effectiveness of Zidovudine (AZT) in the Treatment of HIV Infection in Patients With AIDS and Advanced ARC
CTID: NCT00000999
Phase: Phase 1    Status: Completed
Date: 2021-11-04
A Phase I Concentration-Controlled Trial to Assess the Safety, Tolerance, Pharmacokinetics and Development of Decreased HIV-1 Susceptibility to the Combination of Atevirdine Mesylate (U-87201E), Zidovudine (AZT), and Didanosine (ddI)
CTID: NCT00000742
Phase: Phase 1    Status: Completed
Date: 2021-11-04
A Comparative Study of a Combination of Zidovudine, Didanosine, and Double-Blinded Nevirapine Versus a Combination of Zidovudine and Didanosine
CTID: NCT00000770
Phase: Phase 2    Status: Completed
Date: 2021-11-04
Methadone Effects on Zidovudine (ZDV, AZT) Disposition
CTID: NCT00000800
Phase: Phase 1    Status: Completed
Date: 2021-11-04
A Study of Dideoxycytidine Plus Zidovudine (AZT) in HIV-Infected Children Who Have Taken AZT
CTID: NCT00001032
Phase: Phase 2    Status: Completed
Date: 2021-11-04
A Study of 141W94 Used Alone or in Combination With Zidovudine Plus 3TC in HIV-Infected Patients
CTID: NCT00001085
Phase: Phase 2    Status: Completed
Date: 2021-11-04
Granulocyte-Macrophage Colony-Stimulating Factor and Zidovudine: A Phase I Study of Concurrent Administration in Patients With AIDS and Severe ARC
CTID: NCT00000711
Phase: Phase 1    Status: Completed
Date: 2021-11-04
The Safety and Effectiveness of Hyperimmune Anti-HIV Intravenous Immunoglobulin (HVIG) Plus Zidovudine in HIV-Infected Infants
CTID: NCT00000961
Phase: Phase 2    Status: Terminated
Date: 2021-11-04
The Safety of Different Dose Levels of Zidovudine in HIV-Infected Children
CTID: NCT00000983
Phase: Phase 2    Status: Completed
Date: 2021-11-04
Comparison of New Anti-HIV Drug Combinations in HIV-Infected Children Who Have Taken Anti-HIV Drugs
CTID: NCT00001083
Phase: Phase 2    Status: Completed
Date: 2021-11-04
A Study of Zidovudine Plus Interleukin-2 in HIV-Infected Patients Who Have No Symptoms of Infection But Who Have Tender Lymph Nodes
CTID: NCT00001005
Phase: Phase 1    Status: Completed
Date: 2021-11-04
A Phase I Concentration-Targeted Multidose Study of Atevirdine Mesylate ( U-87201E ), AZT, and ddI or ddC
CTID: NCT00000753
Phase: Phase 1    Status: Completed
Date: 2021-11-04
A Randomized Phase II Study of Two Doses of Interferon Alfa-2a (IFN Alfa-2a) in Combination With
Studio PKCT - Pharmacokinetics of chemotherapy when given concurrently with antiretroviral (Protocol no. CSL01).
CTID: null
Phase: Phase 4    Status: Completed
Date: 2011-10-20
HIV postexposure prophylaxis with Darunavir/r (PEPDar)
CTID: null
Phase: Phase 3    Status: Completed
Date: 2011-07-26
Studio degli effetti immuno-virologici dell’interruzione di Maraviroc nei pazienti che stanno fallendo un regime contenente Maraviroc
CTID: null
Phase: Phase 2    Status: Prematurely Ended
Date: 2011-03-22
BREATHER (PENTA 16): Short-cycle therapy (SCT) (5 days on/ 2 days off) in young people with chronic HIV-infection
CTID: null
Phase: Phase 2, Phase 3    Status: Completed
Date: 2010-08-11
Efficacy of atazanavir/ritonavir monotherapy as maintenance in patients with viral suppression. Randomized, open label non inferiority trial. (MODAt STUDY)
CTID: null
Phase: Phase 3    Status: Completed
Date: 2010-07-26
MoLO study - Evaluation of cost/efficacy ratio of monotherapy with lopinavir/ritonavir versus standard in patients treated with protesi inhibotors in virologic suppressison.
CTID: null
Phase: Phase 3    Status: Prematurely Ended
Date: 2010-03-24
Randomised trial comparing the introduction of an immediate or deferred new HAART regimen in failing HIV infected patients: the role of lamivudine monotherapy
CTID: null
Phase: Phase 3    Status: Completed
Date: 2009-03-13
Study on Pharmacokinetics of newly developed ANtiretroviral agents in HIV-infected pregNAnt women (PANNA)
CTID: null
Phase: Phase 4    Status: Trial now transitioned, Ongoing, GB - no longer in EU/EEA
Date: 2009-02-13
A Phase III, Randomised, Open- Label Study Comparing the Safety and Efficacy of Switching Stavudine or Zidovudine to Tenofovir Disoproxil Fumarate versus Continuing Stavudine or Zidovudine in Virologically Suppressed HIV-Infected Children Taking Highly Active Antiretroviral Therapy.
CTID: null
Phase: Phase 3    Status: Completed
Date: 2007-12-18
Estudio piloto, aleatorizado, nacional, multicéntrico, de eficacia inmunológica, comparando un régimen TARGA de tres fármacos (Tenofovir + Emtricitabina + Lopinavir/r) y cuatro fármacos (Tenofovir + Emtricitabina + Zidovudina + Lopinavir/r) en primera línea para el tratamiento de pacientes con infección por VIH avanzada
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2007-12-15
A randomised open-label study comparing the safety and efficacy of three different combination antiretroviral regimens as initial therapy for HIV infection.
CTID: null
Phase: Phase 3, Phase 4    Status: Ongoing, Completed
Date: 2007-07-10
The Liverpool HIV TDM Registry: Studying influences upon plasma HIV drug exposure
CTID: null
Phase: Phase 4    Status: Completed
Date: 2007-04-26
Host genetic factors influencing drug disposition and response to HIV treatment
CTID: null
Phase: Phase 4    Status: Completed
Date: 2007-03-29
Prévention de la transmission du VIH-1 de la mère à l’enfant sans utilisation d’analogue nucléosidique
CTID: null
Phase: Phase 2    Status: Ongoing
Date: 2007-03-09
A Randomized, Controlled, Open-Label, 48-Week Study of Continuing Successfully Suppressive Treatment in HIV-1 Infected Adults with First-Line Twice-Daily Zidovudine and Lamivudine-Based Regimens versus Proactively Replacing of Zidovudine and Lamivudine by Once-Daily Emtricitabine and Tenofovir Disoproxil Fumarate to Prevent Progression of or Reverse Peripheral Lipoatrophy.
CTID: null
Phase: Phase 4    Status: Ongoing, Completed
Date: 2006-07-26
Strategic long term, immunologically driven treatment interruptions in previously naive patients starting HAART: a controlled, randomized, multicenter study
CTID: null
Phase: Phase 3    Status: Ongoing
Date: 2006-06-29
THERAPEUTIC SIMPLIFICATION WITH THYMIDINE ANALOGUR SPARING REGIMENS IN PATIENTS ON EFFECTIVE HAART: A CONTROLLED, RANDOMIZED STUDY
CTID: null
Phase: Phase 3    Status: Completed
Date: 2005-10-11
Estudio piloto unicéntrico, abierto, aleatorizado para evaluar la evolución inmuno-virológica y clínica de una combinación con análogos a nucleósidos/nucléotidos (TrizivirÒ +Tenofovir) en pacientes multirresistentes en fracaso virológico
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2005-09-02
Eficacia virológica, inmunológica y trascendencia clínica del efecto de la lamivudina en pacientes portadores de la mutación M184V/I con fracaso terapéutico.
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2005-05-10
A phase 3, open label, randomised, parallel group study to compare the effect on prevention and resolution of treatment related adverse events of a simplified, once daily regimen of a fixed dose combination tablet of emtricitabine and tenofovir DF versus twice daily co-formulated zidovudine and lamivudine (Combivir®) or zidovudine and lamivudine, in virologically suppressed, HIV infected patients taking efavirenz.
CTID: null
Phase: Phase 3    Status: Completed
Date: 2005-05-06
A RANDOMIZED STUDY, POSPECTIVE, ON THREE DIFFERENT STRATEGY OF HIV POSITIVE NAIVE FOR THE ANTIRETROVIRAL THERAPY
CTID: null
Phase: Phase 3    Status: Completed
Date: 2005-04-05
Pilot study randomised and open to compare the switch to trizivir against the previous treatment in chronic HIV-1 infected patients with liver cirrhosis secondary to Hepatitis C coinfection
CTID: null
Phase: Phase 3, Phase 4    Status: Ongoing
Date: 2005-01-25
Ensayo para evaluar la eficacia, seguridad y tolerabilidad, de una estrategia de simplificacion temprana a Trizivir, en pacientes con supresion virologica tras tratamiento antiretroviral con Combivir mas Lopinavir/Ritonavir (Kaletra).
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2004-11-26
ESTUDIO PILOTO PARA EVALUAR LA EFICACIA Y TOLERANCIA DE LA COMBINACION DE TRIZIVIR Y TENOFOVIR EN PACIENTES INFECTADOS POR EL VIH, SIN TRATAMENTO ANTIRRETROVIRAL PREVIO (NAÏVES)
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2004-11-18
NATIONAL, MULTICENTER, RANDOMISED, OPEN STUDY TO VALUATE THE EFFICACY OF DIFFERENT THERAPEUTIC STRATEGIES TO AVOID THE IMMUNOLOGIC FAILURE IN MULTIRESISTENT HIV-1 INFECTED PATIENTS.
CTID: null
Phase: Phase 3    Status: Prematurely Ended
Date: 2004-09-15
A pilot clinical trial of reverse transcriptase inhibitors in children with Aicardi-Goutières syndrome (AGS)
CTID: null
Phase: Phase 2    Status: Completed
Date:

Biological Data
  • NRTIs suppressed laser-induced CNV volume in wild-type mice. Intravitreous administration of 3TC, AZT, and ABC significantly suppressed laser-induced CNV volume in wild-type mice. Data are means ± SEM (n = 7–13 per group). *P < 0.05 by one-way ANOVA and Dunn method. Right: Representative images of FITC-isolectin CNV lesions in RPE-choroid flat mounts. Scale bar: 100 μm. Invest Ophthalmol Vis Sci . 2015 Nov;56(12):7122-9.
  • NRTIs did not suppress laser-induced CNV in P2rx7−/− mice. Intravitreous injection of 3TC, AZT, and ABC did not suppress the laser-induced CNV in P2rx7−/− mice compared with control (PBS). Data are means ± SEM (n = 4 per group). *P < 0.05 by one-way ANOVA and Dunn method. N.S., not significant. Right: Representative images of FITC-isolectin CNV lesions in RPE-choroid flat mounts. Scale bar: 100 μm. Invest Ophthalmol Vis Sci . 2015 Nov;56(12):7122-9.
  • Coadministration of 3TC with anti-mouse VEGF antibody did not reduce CNV. Intravitreous injection of an anti-mouse VEGF antibody (10 ng) reduced laser-induced CNV volume in wild-type and P2rx7−/− mice compared with an IgG control (10 ng). Intravitreous injection of 3TC in combination with the anti-mouse VEGF antibody suppressed laser-induced CNV to a similar extent in wild-type and P2rx7−/− mice. Data are means ± SEM (n = 4–6 per group). *P < 0.05 versus control by one-way ANOVA and Student-Newman-Keuls post hoc test. Bottom: Representative images of FITC-isolectin CNV lesions in RPE-choroid flat mounts. Scale bar: 100 μm. Invest Ophthalmol Vis Sci . 2015 Nov;56(12):7122-9.
Contact Us