Zidovudine Tablets

Name: Zidovudine Tablets

Adverse Reactions


The following adverse reactions are discussed in greater detail in other sections of the labeling:

  • Hematologic toxicity, including neutropenia and anemia [see Boxed Warning, Warnings and Precautions (5.1)].
  • Symptomatic myopathy [see Boxed Warning, Warnings and Precautions (5.3)].
  • Lactic acidosis and severe hepatomegaly with steatosis [see Boxed Warning, Warnings and Precautions (5.4)].
  •  Hepatic decompensation in patients co-infected with HIV-1 and hepatitis C [see Warnings and Precautions (5.5)]. 

Clinical Trials Experience


Because clinical trials are conducted under widely varying conditions, adverse reaction rates observed in the clinical trials of a drug cannot be directly compared with rates in the clinical trials of another drug and may not reflect the rates observed in practice.

Adults

The frequency and severity of adverse reactions associated with the use of zidovudine are greater in patients with more advanced infection at the time of initiation of therapy.

Table 3 summarizes adverse reactions reported at a statistically significant greater incidence for subjects receiving oral zidovudine in a monotherapy trial.


Table 3. Percentage (%) of Subjects with Adverse Reactions (Greater than or Equal to 5% Frequency) in Asymptomatic HIV-1 Infection (ACTG 019)
Adverse Reaction Zidovudine 500 mg/day
(n = 453)
Placebo
(n = 428)
aNot statistically significant versus placebo.
 Body as a whole
 
 
     Asthenia
9%a
6%
     Headache
63%
53%
     Malaise
53%
45%
 Gastrointestinal
 
 
     Anorexia
20%
11%
     Constipation
6%a
4%
     Nausea
51%
30%
     Vomiting
17%
10%

In addition to the adverse reactions listed in Table 3, adverse reactions observed at an incidence of greater than or equal to 5% in any treatment arm in clinical trials (NUCA3001, NUCA3002, NUCB3001, and NUCB3002) were abdominal cramps, abdominal pain, arthralgia, chills, dyspepsia, fatigue, insomnia, musculoskeletal pain, myalgia, and neuropathy. Additionally, in these trials hyperbilirubinemia was reported at an incidence of less than or equal to 0.8%.

Selected laboratory abnormalities observed during a clinical trial of monotherapy with oral zidovudine are shown in Table 4.


Table 4. Frequencies of Selected (Grade 3/4) Laboratory Abnormalities in Subjects with Asymptomatic HIV-1 Infection (ACTG 019)
Test
(Abnormal Level)
Zidovudine 500 mg/day
(n = 453)
Placebo
(n = 428)
ULN = Upper limit of normal.
 Anemia (Hgb <8 g/dL)
1%
<1%
 Granulocytopenia (<750 cells/mm3)
2%
2%
 Thrombocytopenia (platelets <50,000/mm3)
0%
<1%
 ALT (>5 x ULN)
3%
3%
 AST (>5 x ULN)
1%
2%

 
Pediatrics

The clinical adverse reactions reported among adult recipients of zidovudine may also occur in pediatric patients.



Trial ACTG 300: Selected clinical adverse reactions and physical findings with a greater than or equal to 5% frequency during therapy with EPIVIR® (lamivudine) oral suspension 4 mg per kg twice daily plus zidovudine 160 mg per m2 3 times daily compared with didanosine in therapy-naive (less than or equal to 56 days of antiretroviral therapy) pediatric subjects are listed in Table 5.  


Table 5. Selected Clinical Adverse Reactions and Physical Findings (Greater than or Equal to 5% Frequency) in Pediatric Subjects in Trial ACTG 300
Adverse Reaction EPIVIR plus Zidovudine
(n = 236)
Didanosine
(n = 235)
aIncludes pain, discharge, erythema, or swelling of an ear.
 Body as a whole
 
 
     Fever
25%
32%
 Digestive
 
 
     Hepatomegaly
11%
11%
     Nausea & vomiting
8%
7%
     Diarrhea
8%
6%
     Stomatitis
6%
12%
     Splenomegaly
5%
8%
 Respiratory
 
 
     Cough
15%
18%
     Abnormal breath sounds/wheezing
7%
9%
 Ear, Nose, and Throat
 
 
     Signs or symptoms of earsa
7%
6%
     Nasal discharge or congestion
8%
11%
 Other
 
 
     Skin rashes
12%
14%
     Lymphadenopathy
9%
11%

Selected laboratory abnormalities experienced by therapy-naive (less than or equal to 56 days of antiretroviral therapy) pediatric subjects are listed in Table 6.


Table 6. Frequencies of Selected (Grade 3/4) Laboratory Abnormalities in Pediatric Subjects in Trial ACTG 300
Test (Abnormal Level) EPIVIR plus Zidovudine Didanosine
ULN = Upper limit of normal.
ANC = Absolute neutrophil count.
 Neutropenia (ANC <400 cells/mm3)
8%
3%
 Anemia (Hgb <7 g/dL)
4%
2%
 Thrombocytopenia (platelets <50,000/mm3)
1%
3%
 ALT (>10 x ULN)
1%
3%
 AST (>10 x ULN)
2%
4%
 Lipase (>2.5 x ULN)
3%
3%
 Total amylase (>2.5 x ULN)
3%
3%

Macrocytosis was reported in the majority of pediatric subjects receiving zidovudine 180 mg per m2 every 6 hours in open-label trials. Additionally, adverse reactions reported at an incidence of less than 6% in these trials were congestive heart failure, decreased reflexes, ECG abnormality, edema, hematuria, left ventricular dilation, nervousness/irritability, and weight loss.

Use for the Prevention of Maternal-Fetal Transmission of HIV-1

In a randomized, double-blind, placebo-controlled trial in HIV-1-infected women and their neonates conducted to determine the utility of zidovudine for the prevention of maternal-fetal HIV-1 transmission, zidovudine syrup at 2 mg per kg was administered every 6 hours for 6 weeks to neonates beginning within 12 hours following birth. The most commonly reported adverse reactions were anemia (hemoglobin less than 9 g per dL) and neutropenia (less than 1,000 cells per mm3). Anemia occurred in 22% of the neonates who received zidovudine and in 12% of the neonates who received placebo. The mean difference in hemoglobin values was less than 1 g per dL for neonates receiving zidovudine compared with neonates receiving placebo. No neonates with anemia required transfusion and all hemoglobin values spontaneously returned to normal within 6 weeks after completion of therapy with zidovudine. Neutropenia in neonates was reported with similar frequency in the group that received zidovudine (21%) and in the group that received placebo (27%). The long-term consequences of in utero and infant exposure to zidovudine are unknown.

Postmarketing Experience

The following adverse reactions have been identified during post-approval use of zidovudine. Because these reactions are reported voluntarily from a population of unknown size, it is not always possible to reliably estimate their frequency or establish a causal relationship to drug exposure.

 

Body as a Whole

Back pain, chest pain, flu-like syndrome, generalized pain, redistribution/accumulation of body fat [see Warnings and Precautions (5.7)].


Cardiovascular


Cardiomyopathy, syncope.


Eye


Macular edema.


Gastrointestinal


Constipation,dysphagia, flatulence, oral mucosa pigmentation, mouth ulcer.


General


Sensitization reactions including anaphylaxis and angioedema, vasculitis.


Hematologic


Aplastic anemia, hemolytic anemia, leukopenia, lymphadenopathy, pancytopenia with marrow hypoplasia, pure red cell aplasia.


Hepatobiliary


Hepatitis, hepatomegaly with steatosis, jaundice, lactic acidosis, pancreatitis.


Musculoskeletal


Increased CPK, increased LDH, muscle spasm, myopathy and myositis with pathological changes (similar to that produced by HIV-1 disease), rhabdomyolysis, tremor.


Nervous

Anxiety, confusion, depression, dizziness, loss of mental acuity, mania, paresthesia, seizures, somnolence, vertigo.


Reproductive System and Breast


Gynecomastia.


Respiratory

Dyspnea, rhinitis, sinusitis.


Skin and Subcutaneous Tissue

Changes in skin and nail pigmentation, pruritus, Stevens-Johnson syndrome, toxic epidermal necrolysis, sweating, urticaria.


Special Senses


Amblyopia, hearing loss, photophobia, taste perversion.


Renal and Urinary


Urinary frequency, urinary hesitancy.

Drug Interactions

Antiretroviral Agents


Stavudine

Concomitant use of zidovudine with stavudine should be avoided since an antagonistic relationship has been demonstrated in vitro.

Nucleoside Analogues Affecting DNA Replication

Some nucleoside analogues affecting DNA replication, such as ribavirin, antagonize the in vitro antiviral activity of zidovudine against HIV-1; concomitant use of such drugs should be avoided.

Doxorubicin


Concomitant use of zidovudine with doxorubicin should be avoided since an antagonistic relationship has been demonstrated in vitro.

Hematologic/Bone Marrow Suppressive/Cytotoxic Agents


Coadministration of ganciclovir, interferon alfa, ribavirin, and other bone marrow suppressive or cytotoxic agents may increase the hematologic toxicity of zidovudine.

Use in specific populations

Pregnancy

Pregnancy Exposure Registry

There is a pregnancy exposure registry that monitors pregnancy outcomes in women exposed to zidovudine during pregnancy. Healthcare providers are encouraged to register patients by calling the Antiretroviral Pregnancy Registry (APR) at 1-800-258-4263.


Risk Summary


Available data from the APR show no difference in the overall risk of birth defects for zidovudine compared with the background rate for birth defects of 2.7% in the Metropolitan Atlanta Congenital Defects Program (MACDP) reference population [see Data]. The APR uses the MACDP as the U.S. reference population for birth defects in the general population. The MACDP evaluates women and infants from a limited geographic area and does not include outcomes for births that occurred at less than 20 weeks gestation. The rate of miscarriage is not reported in the APR. The estimated background rate of miscarriage in clinically recognized pregnancies in the U.S. general population is 15% to 20%. The background risk for major birth defects and miscarriage for the indicated population is unknown. 


In an animal reproduction study, administration of oral zidovudine to female rats prior to mating and throughout gestation resulted in embryotoxicity at doses that produced systemic exposure (AUC) approximately 33 times higher than exposure at the recommended clinical dose. However, no embryotoxicity was observed after oral administration of zidovudine to pregnant rats during organogenesis at doses that produced systemic exposure (AUC) approximately 117 times higher than exposures at the recommended clinical dose. Administration of oral zidovudine to pregnant rabbits during organogenesis resulted in embryotoxicity at doses that produced systemic exposure (AUC) approximately 108 times higher than exposure at the recommended clinical dose. However, no embryotoxicity was observed at doses that produced systemic exposure (AUC) approximately 23 times higher than exposures at the recommended clinical dose [see Data].


Data


Human Data: Based on prospective reports to the APR of over 13,000 exposures to zidovudine during pregnancy resulting in live births (including over 4,000 exposed in the first trimester), there was no difference between the overall risk of birth defects for zidovudine compared with the background birth defect rate of 2.7% in a U.S. reference population of the MACDP. The prevalence of birth defects in live births was 3.2% (95% CI: 2.7% to 3.8%) following first trimester exposure to zidovudine-containing regimens and 2.8% (95% CI: 2.5% to 3.2%) following second/third trimester exposure to zidovudine-containing regimens.


A randomized, double-blind, placebo-controlled trial was conducted in HIV-1-infected pregnant women to determine the utility of zidovudine for the prevention of maternal-fetal HIV-1-transmission [see Clinical Studies(14.3)]. Zidovudine treatment during pregnancy reduced the rate of maternal-fetal HIV-1 transmission from 24.9% for infants born to placebo-treated mothers to 7.8% for infants born to mothers treated with zidovudine. There were no differences in pregnancy-related adverse events between the treatment groups. Of the 363 neonates that were evaluated, congenital abnormalities occurred with similar frequency between neonates born to mothers who received zidovudine and neonates born to mothers who received placebo. The observed abnormalities included problems in embryogenesis (prior to 14 weeks) or were recognized on ultrasound before or immediately after initiation of study drug.


Zidovudine has been shown to cross the placenta and concentrations in neonatal plasma at birth were essentially equal to those in maternal plasma at delivery [see Clinical Pharmacology (12.3)].

Animal Data: A study in pregnant rats (at 50, 150, or 450 mg per kg per day starting 26 days prior to mating through gestation to postnatal Day 21) showed increased fetal resorptions at doses that produced systemic exposures (AUC) approximately 33 times higher than exposure at the recommended daily human dose (300 mg twice daily). However, in an oral embryo-fetal development study in rats (at 125, 250, or 500 mg per kg per day on gestation Days 6 through 15), no fetal resorptions were observed at doses that produced systemic exposure (AUC) approximately 117 times higher than exposures at the recommended daily human dose. An oral embryo-fetal development study in rabbits (at 75, 150, or 500 mg per kg per day on gestation Day 6 through 18) showed increased fetal resorptions at the 500 mg per kg per day dose, which produced systemic exposures (AUC) approximately 108 times higher than exposure at the recommended daily human dose; however, no fetal resorptions were noted at doses up to 150 mg per kg per day, which produced systemic exposure (AUC) approximately 23 times higher than exposures at the recommended daily human dose. These oral embryo-fetal development studies in the rat and rabbit revealed no evidence of fetal malformations with zidovudine. In another developmental toxicity study, pregnant rats (dosed at 3,000 mg per kg per day from Days 6 through 15 of gestation) showed marked maternal toxicity and an increased incidence of fetal malformations at exposures greater than 300 times the recommended daily human dose based on AUC. However, there were no signs of fetal malformations at doses up to 600 mg per kg per day.

Lactation

Risk Summary


The Centers for Disease Control and Prevention recommend that HIV-1-infected mothers in the United States not breastfeed their infants to avoid risking postnatal transmission of HIV-1 infection. Zidovudine is present in human milk. There is no information on the effects of zidovudine on the breastfed infant or the effects of the drug on milk production. Because of the potential for (1) HIV-1 transmission (in HIV-negative infants), (2) developing viral resistance (in HIV-positive infants), and (3) serious adverse reactions in a breastfed infant instruct mothers not to breastfeed if they are receiving zidovudine.

Pediatric Use


Zidovudine has been studied in HIV-1-infected pediatric subjects aged at least 6 weeks who had HIV-1-related symptoms or who were asymptomatic with abnormal laboratory values indicating significant HIV-1-related immunosuppression. Zidovudine has also been studied in neonates perinatally exposed to HIV-1 [see Dosage and Administration (2.2), Adverse Reactions (6.1), Clinical Pharmacology (12.3), Clinical Studies (14.2), (14.3)].

Geriatric Use


Clinical studies of zidovudine did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects. Other reported clinical experience has not identified differences in responses between the elderly and younger patients. In general, dose selection for an elderly patient should be cautious, reflecting the greater frequency of decreased hepatic, renal, or cardiac function, and of concomitant disease or other drug therapy.

Renal Impairment


Unchanged zidovudine and its glucuronide metabolite (formed in the liver) are primarily eliminated from the body by renal excretion. In patients with severely impaired renal function (CrCl less than 15 mL per min), dosage reduction is recommended [see Dosage and Administration (2.5), Clinical Pharmacology (12.3)].

Hepatic Impairment


Zidovudine is primarily eliminated by hepatic metabolism and zidovudine concentrations appear to be increased in patients with impaired hepatic function, which may increase the risk of hematologic toxicity. Frequent monitoring of hematologic toxicities is advised. There are insufficient data to recommend dose adjustment of zidovudine in patients with impaired hepatic function or liver cirrhosis [see Dosage and Administration (2.6), Clinical Pharmacology (12.3)].

Zidovudine Tablets - Clinical Pharmacology

Mechanism of Action


Zidovudine is an antiretroviral agent [see Microbiology (12.4)].

Pharmacokinetics

Absorption and Bioavailability

In adults, following oral administration, zidovudine is rapidly absorbed and extensively distributed, with peak serum concentrations occurring within 0.5 to 1.5 hours. The AUC was equivalent when zidovudine was administered as Zidovudine Tablets or syrup compared with zidovudine capsules. The pharmacokinetic properties of zidovudine in fasting adult subjects are summarized in Table 7.

Table 7. Zidovudine Pharmacokinetic Parameters in Adult Subjects
a Median [range] for 50 paired samples drawn 1 to 8 hours after the last dose in subjects on chronic therapy with zidovudine.
b Approximate range.
Parameter
Mean ± SD (except where noted)
 Oral bioavailability (%)
64 ± 10 (n = 5)
 Apparent volume of distribution (L/kg)
1.6 ± 0.6 (n = 8)
 Cerebrospinal fluid (CSF):plasma ratioa
0.6 [0.04 to 2.62] (n = 39)
 Systemic clearance (L/h/kg)
1.6 ± 0.6 (n = 6)
 Renal clearance (L/h/kg)
0.34 ± 0.05 (n = 9)
 Elimination half-life (h)b
0.5 to 3 (n = 19)

Distribution

The apparent volume of distribution of zidovudine is 1.6 ± 0.6 L per kg (Table 7) and binding to plasma protein is low (less than 38%).

Metabolism and Elimination

Zidovudine is primarily eliminated by hepatic metabolism. The major metabolite of zidovudine is GZDV. GZDV AUC is about 3-fold greater than the zidovudine AUC. Urinary recovery of zidovudine and GZDV accounts for 14% and 74%, respectively, of the dose following oral administration. A second metabolite, 3′-amino-3′-deoxythymidine (AMT), has been identified in the plasma following single-dose IV administration of zidovudine. The AMT AUC was one-fifth of the zidovudine AUC. Pharmacokinetics of zidovudine were dose independent at oral dosing regimens ranging from 2 mg per kg every 8 hours to 10 mg per kg every 4 hours.

Effect of Food on Absorption

Zidovudine may be administered with or without food. The zidovudine AUC was similar when a single dose of zidovudine was administered with food.

Specific Populations

Renal Impairment: Zidovudine clearance was decreased resulting in increased zidovudine and GZDV half-life and AUC in subjects with impaired renal function (n = 14) following a single 200 mg oral dose (Table 8). Plasma concentrations of AMT were not determined. No dose adjustment is recommended for patients with CrCl greater than or equal to 15 mL per min. 

Table 8. Zidovudine Pharmacokinetic Parameters in Subjects with Severe Renal Impairmenta
a Data are expressed as mean ± standard deviation.
Parameter
Control Subjects
(Normal Renal Function)
(n = 6)
Subjects with Renal
Impairment
(n = 14)
 CrCl (mL/min)
120 ± 8
18 ± 2
 Zidovudine AUC (ng•h/mL)
1,400 ± 200
3,100 ± 300
 Zidovudine half-life (h)
1 ± 0.2
1.4 ± 0.1

Hemodialysis and Peritoneal Dialysis: The pharmacokinetics and tolerance of zidovudine were evaluated in a multiple-dose trial in subjects undergoing hemodialysis (n = 5) or peritoneal dialysis (n = 6) receiving escalating oral doses up to 200 mg 5 times daily for 8 weeks. Daily doses of 500 mg or less were well tolerated despite significantly elevated GZDV plasma concentrations. Apparent zidovudine oral clearance was approximately 50% of that reported in subjects with normal renal function. Hemodialysis and peritoneal dialysis appeared to have a negligible effect on the removal of zidovudine, whereas GZDV elimination was enhanced. A dosage adjustment is recommended for patients undergoing hemodialysis or peritoneal dialysis [see Dosage and Administration (2.5)].

Hepatic Impairment: Data describing the effect of hepatic impairment on the pharmacokinetics of zidovudine are limited. However, zidovudine is eliminated primarily by hepatic metabolism and it appears that zidovudine clearance is decreased and plasma concentrations are increased in subjects with hepatic impairment. There are insufficient data to recommend dose adjustment of zidovudine in patients with impaired hepatic function or liver cirrhosis [see Dosage and Administration (2.6)].

Pediatric Patients: Zidovudine pharmacokinetics have been evaluated in HIV-1-infected pediatric subjects (Table 9).

Patients Aged 3 Months to 12 Years: Overall, zidovudine pharmacokinetics in pediatric patients older than 3 months are similar to those in adult patients. Proportional increases in plasma zidovudine concentrations were observed following administration of oral solution from 90 to 240 mg per m2 every 6 hours. Oral bioavailability, terminal half-life, and oral clearance were comparable to adult values. As in adult subjects, the major route of elimination was by metabolism to GZDV. After IV dosing, about 29% of the dose was excreted in the urine unchanged, and about 45% of the dose was excreted as GZDV [see Dosage and Administration (2.2)].

Patients Aged Less than 3 Months: Zidovudine pharmacokinetics have been evaluated in pediatric subjects from birth to 3 months of life. Zidovudine elimination was determined immediately following birth in 8 neonates who were exposed to zidovudine in utero. The half-life was 13 ± 5.8 hours. In neonates less than or equal to 14 days old, bioavailability was greater, total body clearance was slower, and half-life was longer than in pediatric subjects older than 14 days. For dose recommendations for neonates [see Dosage and Administration (2.3)]. 

Table 9. Zidovudine Pharmacokinetic Parameters in Pediatric Subjectsa
a Data presented as mean ± standard deviation except where noted.
b Median [range].
Parameter
Birth to 14 Days
Aged 14 Days
to 3 Months
Aged 3 Months
to 12 Years
 Oral bioavailability (%)
89 ± 19
(n = 15)
61 ± 19
(n = 17)
65 ± 24
(n = 18)
 CSF:plasma ratio
no data
no data
0.68 [0.03 to 3.25]b
(n = 38)
 CL (L/h/kg)
0.65 ± 0.29
(n = 18)
1.14 ± 0.24
(n = 16)
1.85 ± 0.47
(n = 20)
 Elimination half-life (h)
3.1 ± 1.2
(n = 21)
1.9 ± 0.7
(n = 18)
1.5 ± 0.7
(n = 21)

Pregnancy: Zidovudine pharmacokinetics have been studied in a Phase I trial of 8 women during the last trimester of pregnancy. Zidovudine pharmacokinetics were similar to those of nonpregnant adults. Consistent with passive transmission of the drug across the placenta, zidovudine concentrations in neonatal plasma at birth were essentially equal to those in maternal plasma at delivery [see Use in Specific Populations (8.1)].

Although data are limited, methadone maintenance therapy in 5 pregnant women did not appear to alter zidovudine pharmacokinetics.

Geriatric Patients: Zidovudine pharmacokinetics have not been studied in subjects over 65 years of age.

Gender: A pharmacokinetic trial in healthy male (n = 12) and female (n = 12) subjects showed no differences in zidovudine AUC when a single dose of zidovudine was administered as a 300 mg zidovudine tablet.

Drug Interactions

[See Drug Interactions (7).]

Table 10. Effect of Coadministered Drugs on Zidovudine AUCa
↑ = Increase; ↓ = Decrease; ↔ = No significant change; AUC = Area under the concentration versus time curve; CI = Confidence interval.
a This table is not all inclusive.
b Estimated range of percent difference.
Note: ROUTINE DOSE MODIFICATION OF ZIDOVUDINE IS NOT WARRANTED WITH COADMINISTRATION OF THE FOLLOWING DRUGS.
Coadministered Drug
and Dose
Zidovudine
Oral Dose
n
Zidovudine
Concentrations
Concentration of
Coadministered
Drug
AUC
Variability
 
 Atovaquone 750 mg every 12 h with food
200 mg every 8 h
14
↑ 31%
Range:
23% to 78%b

 Clarithromycin 500 mg twice daily
100 mg every 4 h x 7 days
4
↓ 12%
Range:
↓34% to ↑14%b
Not Reported
 Fluconazole 400 mg daily
200 mg every 8 h
12
↑ 74%
95% CI:
54% to 98%
Not Reported
 Lamivudine 300 mg every 12 h
single 200 mg
12
↑ 13%
90% CI:
2% to 27%

 Methadone 30 to 90 mg daily
200 mg every 4 h
9
↑ 43%
Range:
16% to 64%b

 Nelfinavir 750 mg every 8 h x 7 to 10 days
single 200 mg
11
↓ 35%
Range:
28% to 41%b

 Probenecid 500 mg every 6 h x 2 days
2 mg/kg every 8 h x 3 days
3
↑ 106%
Range:
100% to 170%b
Not Assessed
 Rifampin 600 mg daily x 14 days
200 mg every 8 h x 14 days
8
↓ 47%
90% CI:
41% to 53%
Not Assessed
 Ritonavir 300 mg every 6 h x 4 days
200 mg every 8 h x 4 days
9
↓ 25%
95% CI:
15% to 34%

 Valproic acid 250 mg or 500 mg every 8 h x 4 days
100 mg every 8 h x 4 days
6
↑ 80%
Range:
64% to 130%b
Not Assessed

Phenytoin:  Phenytoin plasma levels have been reported to be low in some patients receiving zidovudine, while in one case a high level was documented. However, in a pharmacokinetic interaction trial in which 12 HIV-1-positive volunteers received a single 300 mg phenytoin dose alone and during steady-state zidovudine conditions (200 mg every 4 hours), no change in phenytoin kinetics was observed. Although not designed to optimally assess the effect of phenytoin on zidovudine kinetics, a 30% decrease in oral zidovudine clearance was observed with phenytoin.

Ribavirin:  In vitro data indicate ribavirin reduces phosphorylation of lamivudine, stavudine, and zidovudine. However, no pharmacokinetic (e.g., plasma concentrations or intracellular triphosphorylated active metabolite concentrations) or pharmacodynamic (e.g., loss of HIV-1/HCV virologic suppression) interaction was observed when ribavirin and lamivudine  (n = 18), stavudine (n = 10), or zidovudine (n = 6) were coadministered as part of a multi-drug regimen to HIV-1/HCV co-infected subjects [see Warnings and Precautions (5.5)].

Microbiology


Mechanism of Action 

Zidovudine is a synthetic nucleoside analogue. Intracellularly, zidovudine is phosphorylated to its active 5'-triphosphate metabolite, zidovudine triphosphate (ZDV-TP). The principal mode of action of ZDV-TP is inhibition of reverse transcriptase (RT) via DNA chain termination after incorporation of the nucleotide analogue. ZDV-TP is a weak inhibitor of the cellular DNA polymerases α and γ and has been reported to be incorporated into the DNA of cells in culture.

Antiviral Activity 

The antiviral activity of zidovudine against HIV-1 was assessed in a number of cell lines including monocytes and fresh human peripheral blood lymphocytes. The EC50 and EC90 values for zidovudine were 0.01 to 0.49 microM (1 microM = 0.27 mcg per mL) and 0.1 to 9 microM, respectively. HIV-1 from therapy-naive subjects with no amino acid substitutions associated with resistance gave median EC50 values of 0.011 microM (range: 0.005 to 0.11 microM) from Virco (n = 92 baseline samples) and 0.0017 microM (range0.006 to 0.0340 microM) from Monogram Biosciences (n = 135 baseline samples). The EC50 values of zidovudine against different HIV-1 clades (A-G) ranged from 0.00018 to 0.02 microM, and against HIV-2 isolates from 0.00049 to 0.004 microM. Zidovudine was not antagonistic to tested anti-HIV agents with the exception of stavudine where an antagonistic relationship with zidovudine has been demonstrated in cell culture. Ribavirin has been found to inhibit the phosphorylation of zidovudine in cell culture.

Resistance 

HIV-1 isolates with reduced susceptibility to zidovudine have been selected in cell culture and were also recovered from subjects treated with zidovudine. Genotypic analyses of the isolates selected in cell culture and recovered from zidovudine-treated subjects showed thymidine analog mutation (TAMs) substitutions in the HIV-1 RT (M41L, D67N, K70R, L210W, T215Y or F, and K219E/R/H/Q/N/Q) that confer zidovudine resistance. In general, higher levels of resistance were associated with greater number of substitutions. In some subjects harboring zidovudine-resistant virus at baseline, phenotypic sensitivity to zidovudine was restored by 12 weeks of treatment with lamivudine and zidovudine.

Cross-Resistance 

Cross-resistance has been observed among NRTIs. TAM substitutions are selected by zidovudine and confer cross-resistance to abacavir, didanosine, stavudine, and tenofovir.

PACKAGE LABEL-PRINCIPAL DISPLAY PANEL - 300 mg (60 Tablet Bottle)


NDC 65862-024-60
Zidovudine Tablets,
USP 300 mg
Rx only           60 Tablets
AUROBINDO



PACKAGE LABEL-PRINCIPAL DISPLAY PANEL - 300 mg Blister Carton (6 x 10 Unit-dose)


NDC 65862-024-10
Zidovudine Tablets, USP
300 mg
Rx only           60 Tablets (6 x 10 Unit-dose)
AUROBINDO

ZIDOVUDINE 
zidovudine tablet, film coated
Product Information
Product Type HUMAN PRESCRIPTION DRUG LABEL Item Code (Source) NDC:65862-024
Route of Administration ORAL DEA Schedule     
Active Ingredient/Active Moiety
Ingredient Name Basis of Strength Strength
ZIDOVUDINE (ZIDOVUDINE) ZIDOVUDINE 300 mg
Inactive Ingredients
Ingredient Name Strength
HYPROMELLOSE 2910 (5 MPA.S)  
MAGNESIUM STEARATE  
CELLULOSE, MICROCRYSTALLINE  
POLYETHYLENE GLYCOL 400  
SODIUM STARCH GLYCOLATE TYPE A POTATO  
TITANIUM DIOXIDE  
HYPROMELLOSE 2910 (6 MPA.S)  
Product Characteristics
Color WHITE Score no score
Shape ROUND (Biconvex) Size 10mm
Flavor Imprint Code D;11
Contains     
Packaging
# Item Code Package Description
1 NDC:65862-024-60 60 TABLET, FILM COATED in 1 BOTTLE
2 NDC:65862-024-10 6 BLISTER PACK in 1 CARTON
2 10 TABLET, FILM COATED in 1 BLISTER PACK
Marketing Information
Marketing Category Application Number or Monograph Citation Marketing Start Date Marketing End Date
ANDA ANDA077267 09/19/2005
Labeler - Aurobindo Pharma Limited (650082092)
Establishment
Name Address ID/FEI Operations
Aurobindo Pharma Limited 918917642 ANALYSIS(65862-024), MANUFACTURE(65862-024)
Establishment
Name Address ID/FEI Operations
Aurobindo Pharma Limited 650381903 ANALYSIS(65862-024), MANUFACTURE(65862-024)
Establishment
Name Address ID/FEI Operations
Aurobindo Pharma Limited 918917662 API MANUFACTURE(65862-024)
Revised: 04/2017   Aurobindo Pharma Limited
(web3)