Open menu Close menu Open Search Close search
DRUGS INDEX | MANUFACTURERS INDEX | ANATOMY | GEOGRAPHY | USA STATISTICS | CHINA STATISTICS | RELIGION | JOBS

Viread Tablets (Gilead)



  • Warnings
  • Description
  • Chemical Structure
  • Clinical Pharmacology
  • Indications and Usage
  • Clinical Studies
  • Contraindications
  • Warnings (2)
  • Precautions
  • Drug Interactions
  • Adverse Reactions
  • Overdosage
  • Dosage and Administration
  • How Supplied
  • Patient Package Insert



    - Drugs index
    - Manufacturers
    - Feedback

  • WARNING

    LACTIC ACIDOSIS AND SEVERE HEPATOMEGALY WITH STEATOSIS, INCLUDING FATAL CASES, HAVE BEEN REPORTED WITH THE USE OF NUCLEOSIDE ANALOGS ALONE OR IN COMBINATION WITH OTHER ANTIRETROVIRALS (SEE WARNINGS ).

    VIREAD IS NOT INDICATED FOR THE TREATMENT OF CHRONIC HEPATITIS B VIRUS (HBV) INFECTION AND THE SAFETY AND EFFICACY OF VIREAD HAVE NOT BEEN ESTABLISHED IN PATIENTS CO-INFECTED WITH HBV AND HIV. SEVERE ACUTE EXACERBATIONS OF HEPATITIS B HAVE BEEN REPORTED IN PATIENTS WHO ARE CO-INFECTED WITH HBV AND HIV AND HAVE DISCONTINUED VIREAD. HEPATIC FUNCTION SHOULD BE MONITORED CLOSELY WITH BOTH CLINICAL AND LABORATORY FOLLOW-UP FOR AT LEAST SEVERAL MONTHS IN PATIENTS WHO DISCONTINUE VIREAD AND ARE CO-INFECTED WITH HIV AND HBV. IF APPROPRIATE, INITIATION OF ANTI-HEPATITIS B THERAPY MAY BE WARRANTED (SEE WARNINGS ).

    DESCRIPTION

    VIREAD® is the brand name for tenofovir disoproxil fumarate (a prodrug of tenofovir) which is a fumaric acid salt of bis -isopropoxycarbonyloxymethyl ester derivative of tenofovir. In vivo tenofovir disoproxil fumarate is converted to tenofovir, an acyclic nucleoside phosphonate (nucleotide) analog of adenosine 5'-monophosphate. Tenofovir exhibits activity against HIV-1 reverse transcriptase.

    The chemical name of tenofovir disoproxil fumarate is 9-[( R )-2-[[bis[[(isopropoxycarbonyl)oxy]methoxy]phosphi-nyl]methoxy]propyl]adenine fumarate (1:1). It has a molecular formula of C 19 H 30 N 5 O 10 P · C 4 H 4 O 4 and a molecular weight of 635.52. It has the following structural formula:

    Tenofovir disoproxil fumarate is a white to off-white crystalline powder with a solubility of 13.4 mg/mL in distilled water at 25 °C. It has an octanol/phosphate buffer (pH 6.5) partition coefficient (log p) of 1.25 at 25 °C.

    VIREAD tablets are for oral administration. Each tablet contains 300 mg of tenofovir disoproxil fumarate, which is equivalent to 245 mg of tenofovir disoproxil, and the following inactive ingredients: croscarmellose sodium, lactose monohydrate, magnesium stearate, microcrystalline cellulose, and pregelatinized starch. The tablets are coated with a light blue colored film (Opadry II Y-30-10671-A) that is made of FD&C blue #2 aluminum lake, hydroxypropyl methylcellulose 2910, lactose monohydrate, titanium dioxide, and triacetin.

    In this insert, all dosages are expressed in terms of tenofovir disoproxil fumarate except where otherwise noted.

    Microbiology

    Mechanism of Action:    Tenofovir disoproxil fumarate is an acyclic nucleoside phosphonate diester analog of adenosine monophosphate. Tenofovir disoproxil fumarate requires initial diester hydrolysis for conversion to tenofovir and subsequent phosphorylations by cellular enzymes to form tenofovir diphosphate. Tenofovir diphosphate inhibits the activity of HIV-1 reverse transcriptase by competing with the natural substrate deoxyadenosine 5'-triphosphate and, after incorporation into DNA, by DNA chain termina-tion. Tenofovir diphosphate is a weak inhibitor of ma-mmalian DNA polymerases (alpha), (beta), and mitochondrial DNA polymerase (gamma).

    Antiviral Activity In Vitro:    The in vitro antiviral activity of tenofovir against laboratory and clinical isolates of HIV-1 was assessed in lymphoblastoid cell lines, primary monocyte/macrophage cells and peripheral blood lymphocytes. The IC 50 (50% inhibitory concentration) values for tenofovir were in the range of 0.04 µM to 8.5 µM. In drug combination studies of tenofovir with nucleoside reverse transcriptase inhibitors (abacavir, didanosine, lamivudine, stavudine, zalcitabine, zidovudine), non-nucleoside reverse transcriptase inhibitors (delavirdine, efavirenz, nevirapine), and protease inhibitors (amprenavir, indinavir, nelfinavir, ritonavir, saquinavir), additive to synergistic effects were observed. Tenofovir displayed antiviral activity in vitro against HIV-1 clades A, B, C, D, E, F, G and O (IC 50 values ranged from 0.5 µM to 2.2 µM). The IC 50 values of tenofovir against HIV-2 ranged from 1.6 µM to 4.9 µM.

    Drug Resistance:    HIV-1 isolates with reduced susceptibility to tenofovir have been selected in vitro. These viruses expressed a K65R mutation in reverse transcriptase and showed a 2-4 fold reduction in susceptibility to tenofovir.

    Tenofovir-resistant isolates of HIV-1 have also been recovered from some patients treated with tenofovir in combination with certain antiretroviral agents. In treatment-na[iuml ]ve patients treated with Viread + lamivudine + efavirenz, viral isolates from 8/47 (17%) patients with virologic failure through week 144 showed reduced susceptibility to tenofovir. In treatment-experienced patients, 14/304 (5%) of the VIREAD-treated patients with virologic failure through week 96 showed reduced susceptibility to tenofovir. Genotypic analysis of the resistant isolates showed a mutation in the HIV-1 reverse transcriptase gene resulting in the K65R amino acid substitution.

    Cross-resistance:    Cross-resistance among certain reverse transcriptase inhibitors has been recognized. The K65R mutation selected by tenofovir is also selected in some HIV-1 infected subjects treated with abacavir, didanosine, or zalcitabine. HIV isolates with this mutation also show reduced susceptibility to emtricitabine and lamivudine. Therefore, cross-resistance among these drugs may occur in patients whose virus harbors the K65R mutation. HIV-1 isolates from patients (N=20) whose HIV-1 expressed a mean of 3 zidovudine-associated reverse transcriptase mutations (M41L, D67N, K70R, L210W, T215Y/F or K219Q/E/N), showed a 3.1-fold decrease in the susceptibility to tenofovir. Multinucleoside resistant HIV-1 with a T69S double insertion mutation in the reverse transcriptase showed reduced susceptibility to tenofovir.

    Pharmacokinetics

    The pharmacokinetics of tenofovir disoproxil fumarate have been evaluated in healthy volunteers and HIV-1 infected individuals. Tenofovir pharmacokinetics are similar between these populations.

    Absorption:    VIREAD is a water soluble diester prodrug of the active ingredient tenofovir. The oral bioavailability of tenofovir from VIREAD in fasted patients is approximately 25%. Following oral administration of a single dose of VIREAD 300 mg to HIV-1 infected patients in the fasted state, maximum serum concentrations (C max ) are achieved in 1.0 ± 0.4 hrs. C max and AUC values are 296 ± 90 ng/mL and 2287 ± 685 ng·hr/mL, respectively.

    The pharmacokinetics of tenofovir are dose proportional over a VIREAD dose range of 75 to 600 mg and are not affected by repeated dosing.

    Effects of Food on Oral Absorption:    Administration of VIREAD following a high-fat meal (~700 to 1000 kcal containing 40 to 50% fat) increases the oral bioavailability, with an increase in tenofovir AUC 0-(infinity) of approximately 40% and an increase in C max of approximately 14%. However, administration of VIREAD with a light meal did not have a significant effect on the pharmacokinetics of tenofovir when compared to fasted administration of the drug. Food delays the time to tenofovir C max by approximately 1 hour. C max and AUC of tenofovir are 326 ± 119 ng/mL and 3324 ± 1370 ng·hr/mL following multiple doses of VIREAD 300 mg once daily in the fed state, when meal content was not controlled.

    Distribution:    In vitro binding of tenofovir to human plasma or serum proteins is less than 0.7 and 7.2%, respectively, over the tenofovir concentration range 0.01 to 25 µg/mL. The volume of distribution at steady-state is 1.3 ± 0.6 L/kg and 1.2 ± 0.4 L/kg, following intravenous administration of tenofovir 1.0 mg/kg and 3.0 mg/kg.

    Metabolism and Elimination:    In vitro studies indicate that neither tenofovir disoproxil nor tenofovir are substrates of CYP450 enzymes.

    Following IV administration of tenofovir, approximately 70-80% of the dose is recovered in the urine as unchanged tenofovir within 72 hours of dosing. Following single dose, oral administration of VIREAD, the terminal elimination half-life of tenofovir is approximately 17 hours. After multiple oral doses of VIREAD 300 mg once daily (under fed conditions), 32 ± 10% of the administered dose is recovered in urine over 24 hours.

    Tenofovir is eliminated by a combination of glomerular filtration and active tubular secretion. There may be competition for elimination with other compounds that are also renally eliminated.

    Special Populations

    There were insufficient numbers from racial and ethnic groups other than Caucasian to adequately determine potential pharmacokinetic differences among these populations.

    Tenofovir pharmacokinetics are similar in male and female patients.

    Pharmacokinetic studies have not been performed in children (<18 years) or in the elderly (>65 years).

    The pharmacokinetics of tenofovir following a 300 mg single dose of VIREAD have been studied in non-HIV infected patients with moderate to severe hepatic impairment. There were no substantial alterations in tenofovir pharmacokinetics in patients with hepatic impairment compared with unimpaired patients. No change in VIREAD dosing is required in patients with hepatic impairment.

    The pharmacokinetics of tenofovir are altered in patients with renal impairment (See WARNINGS , Renal Impairment ). In patients with creatinine clearance <50 mL/min or with end-stage renal disease (ESRD) requiring dialysis, C max , and AUC 0-(infinity) of tenofovir were increased (Table 1). It is recommended that the dosing interval for VIREAD be modified in patients with creatinine clearance <50 mL/min or in patients with ESRD who require dialysis (see DOSAGE AND ADMINISTRATION ).

    Table 1. Pharmacokinetic Parameters (Mean±SD) of Tenofovir * in Patients with
    varying Degrees of Renal Function
    Baseline Creatinine
    Clearance (mL/min)
    >80
    (N=3)
    50-80
    (N=10)
    30-49
    (N=8)
    12-29
    (N=11)
    C max (ng/mL)
    335.4 ± 31.8 330.4 ± 61.0 372.1 ± 156.1 601.6 ± 185.3
    AUC 0-(infinity) (ng·hr/mL)
    2184.5 ± 257.4 3063.8 ± 927.0 6008.5 ± 2504.7 15984.7 ± 7223.0
    CL/F (mL/min)
    1043.7 ± 115.4 807.7 ± 279.2 444.4 ± 209.8 177.0 ± 97.1
    CL renal (mL/min)
    243.5 ± 33.3 168.6 ± 27.5 100.6 ± 27.5 43.0 ± 31.2
    *300 mg, single dose of VIREAD

    Tenofovir is efficiently removed by hemodialysis with an extraction coefficient of approximately 54%. Following a single 300 mg dose of VIREAD, a four-hour hemodialysis session removed approximately 10% of the administered tenofovir dose.

    Drug Interactions

    At concentrations substantially higher (~300-fold) than those observed in vivo, tenofovir did not inhibit in vitro drug metabolism mediated by any of the following human CYP450 isoforms: CYP3A4, CYP2D6, CYP2C9 or CYP2E1. However, a small (6%) but statistically significant reduction in metabolism of CYP1A substrate was observed. Based on the results of in vitro experiments and the known elimination pathway of tenofovir, the potential for CYP450 mediated interactions involving tenofovir with other medicinal products is low (See Pharmacokinetics ).

    Tenofovir is primarily excreted by the kidneys by a combination of glomerular filtration and active tubular secretion. Co-administration of VIREAD with drugs that are eliminated by active tubular secretion may increase serum concentrations of either tenofovir or the co-administered drug, due to competition for this elimination pathway. Drugs that decrease renal function may also increase serum concentrations of tenofovir.

    VIREAD has been evaluated in healthy volunteers in combination with abacavir, adefovir dipivoxil, atazanavir, didanosine, efavirenz, emtricitabine, indinavir, lamivudine, lopinavir/ritonavir, methadone, oral contraceptives and ribavirin. Tables 2 and 3 summarize pharmacokinetic effects of co-administered drug on tenofovir pharmacokinetics and effects of VIREAD on the pharmacokinetics of co-administered drug.

    Table 4 summarizes the drug interaction between VIREAD and didanosine. When administered with multiple doses of VIREAD, the C max and AUC of didanosine 400 mg increased significantly. The mechanism of this interaction is unknown. When didanosine 250 mg enteric-coated capsules were administered with VIREAD, systemic exposures to didanosine were similar to those seen with the 400 mg enteric-coated capsules alone under fasted conditions.

    Table 2. Drug Interactions: Changes in Pharmacokinetic Parameters for
    Tenofovir 1 in the Presence of the Co-administered Drug
    Co-administered
    Drug
    Dose of
    Co-administered
    Drug (mg)
      
    N
    % Change of Tenofovir
    Pharmacokinetic Parameters 2
    (90% Cl)
    C max AUC C min
    Abacavir
    300 once 8 [iff ] [iff ] NC
    Adefovir dipivoxil
    10 once 22 [iff ] [iff ] NC
    Atazanavir 3
    400 once daily
    × 14 days
    33 up 14
    (up 8 to up 20)
    up 24
    (up 21 to up 28)
    up 22
    (up 15 to up 30)
    Didanosine
    (enteric-coated)
    400 once 25 [iff ] [iff ] [iff ]
    Didanosine
    (buffered)
    250 or 400 once
    daily × 7 days
    14 [iff ] [iff ] [iff ]
    Efavirenz
    600 once
    daily × 14 days
    29 [iff ] [iff ] [iff ]
    Emtricitabine
    200 once
    daily × 7 days
    17 [iff ] [iff ] [iff ]
    Indinavir
    800 three times
    daily × 7 days
    13 up 14
    (down 3 to up 33)
    [iff ] [iff ]
    Lamivudine
    150 twice daily
    × 7 days
    15 [iff ] [iff ] [iff ]
    Lopinavir/
    Ritonavir
    400/100 twice
    daily × 14 days
    24 [iff ] up 32
    (up 25 to up 38)
    up 51
    (up 37 to up 66)
    1. Patients received VIREAD 300 mg once daily.
    2. Increase = up; Decrease = down; No Effect = [iff ]; NC = Not Calculated
    3. REYATAZ® Prescribing Information (Bristol-Myers Squibb)

    Following multiple dosing to HIV-negative subjects receiving either chronic methadone maintenance therapy or oral contraceptives, or single doses of ribavirin, steady state tenofovir pharmacokinetics were similar to those observed in previous studies, indicating lack of clinically significant drug interactions between these agents and VIREAD.

    Table 3.     Drug Interactions: Changes in Pharmacokinetic Parameters for
    Co-administered Drug in the Presence of VIREAD
    Co-administered
    Drug
    Dose of
    Co-administered
    Drug (mg)
      
    N
    % Change of Co-administered Drug
    Pharmacokinetic Parameters 1
    (90% Cl)
    C max AUC C min
    Abacavir
    300 once 8 up 12
    (down 1 to up 26)
    [iff ] NA
    Adefovir dipivoxil
    10 once 22 [iff ] [iff ] NA
    Efavirenz
    600 once daily
    × 14 days
    30 [iff ] [iff ] [iff ]
    Emtricitabine
    200 once daily
    × 7 days
    17 [iff ] [iff ] up 20
    (up 12 to up 29)
    Indinavir
    800 three times
    daily × 7 days
    12 down 11
    (down 30 to up 12)
    [iff ] [iff ]
    Lamivudine
    150 twice daily
    × 7 days
    15 down 24
    (down 34 to down 12)
    [iff ] [iff ]
    Lopinavir
    Lopinavir/Ritonavir
    400/100 twice
    daily × 14 days
    24 [iff ] [iff ] [iff ]
    Methadone 2
    40-110 once daily
    × 14 days 3
    13 [iff ] [iff ] [iff ]
    Oral
    Contraceptives 4
    Ethinyl Estradiol/
    Norgestimate
    (Ortho-Tricyclen®)
    Once daily
    × 7 days
    20 [iff ] [iff ] [iff ]
    Ribavirin
    600 once 22 [iff ] [iff ] NA
    Ritonavir
    Lopinavir/Ritonavir
    400/100 twice
    daily × 14 days
    24 [iff ] [iff ] [iff ]
    Atazanavir 5
    400 once daily
    × 14 days
    34 down 21
    (down 27 to down 14)
    down 25
    (down 30 to down 19)
    down 40
    (down 48 to down 32)
    Atazanavir 5
    Atazanavir/Ritonavir
    300/100 once
    daily × 42 days
    10 down 28
    (down 50 to up 5)
    down 25 6
    (down 42 to down 3)
    down 23 6
    (down 46 toup 10)
    1. Increase = up; Decrease = down; No Effect = [iff ]; NA = Not Applicable
    2. R-(active), S-and total methadone exposures were equivalent when dosed alone or with VIREAD.
    3. Individual subjects were maintained on their stable methadone dose. No pharmacodynamic alterations (opiate toxicity or withdrawal signs or symptoms) were reported.
    4. Ethinyl estradiol and 17-deacetyl norgestimate (pharmacologically active metabolite) exposures were equivalent when dosed alone or with VIREAD.
    5. REYATAZ Prescribing Information (Bristol-Myers Squibb)
    6. In HIV-infected patients, addition of tenofovir DF to atazanavir 300 mg plus ritonavir 100 mg, resulted in AUC and Cmin values of atazanavir that were 2.3- and 4-fold higher than the respective values observed for atazanavir 400 mg when given alone.

    Table 4.     Drug Interactions: Pharmacokinetic Parameters for Didanosine in the Presence of VIREAD
    Didanosine 1
    Dose (mg)/
    Method of
    Administration 2
    VIREAD Method
    of Administration 2
    N % Difference (90% CI) vs. Didanosine 400 mg
    alone, Fasted 3
    C max AUC
    Buffered tablets      
    400 once daily 4 ×
    7 days
    Fasted 1 hour after
    didanosine
    14 up 28
    (up 11 to up 48)
    up 44
    (up 31 to up 59)
    Enteric coated capsules
          
    400 once,
    fasted
    With food, 2 hr after
    didanosine
    26 up 48
    (up 25 to up 76)
    up 48
    (up 31 to up 67)
    400 once,
    with food
    Simultaneously
    with didanosine
    26 up 64
    (up 41 to up 89)
    up 60
    (up 44 to up 79)
    250 once,
    fasted
    With food, 2 hr after
    didanosine
    28 down 10
    (down 22 to up 3)
    [iff ]
    250 once,
    fasted
    Simultaneously
    with didanosine
    28 [iff ] up 14
    (0 to up 31)
    250 once,
    with food
    Simultaneously
    with didanosine
    28 down 29
    (down 39 to down 18)
    down 11
    (down 23 to up 2)
    1.See PRECAUTIONS regarding use of didanosine with VIREAD.
    2.Administration with food was with a light meal (~373 kcal, 20% fat).
    3.Increase = up; Decrease = down; No Difference = [iff ]
    4.Includes 4 subjects weighing <60 kg receiving ddl 250 mg.

    INDICATIONS AND USAGE

    VIREAD is indicated in combination with other antiretroviral agents for the treatment of HIV-1 infection. This indication is based on analyses of plasma HIV-1 RNA levels and CD4 cell counts in controlled studies of VIREAD in treatment-na[iuml ]ve adults and in treatment-experienced adults.

    Additional important information regarding the use of VIREAD for the treatment of HIV-1 infection:

    • There are no study results demonstrating the effect of VIREAD on clinical progression of HIV-1.
    • The use of VIREAD should be considered for treating adult patients with HIV-1 strains that are expected to be susceptible to tenofovir as assessed by laboratory testing or treatment history (See Description of Clinical Studies ).

    Description of Clinical Studies

    Treatment-Na[iuml ]ve Patients

    Study 903: VIREAD + Lamivudine +Efavirenz Compared to Stavudine + Lamivudine + Efavirenz

    Data through 144 weeks are reported for Study 903, a double-blind, active-controlled multicenter study comparing VIREAD (300 mg QD) administered in combination with lamivudine and efavirenz versus stavudine, lamivudine, and efavirenz in 600 antiretroviral-na[iuml ]ve patients. Patients had a mean age of 36 years (range 18-64), 74% were male, 64% were Caucasian and 20% were Black. The mean baseline CD4 cell count was 279 cells/mm 3 (range 3-956) and median baseline plasma HIV-1 RNA was 77,600 copies/mL (range 417-5,130,000). Patients were stratified by baseline HIV-1 RNA and CD4 count. Forty-three percent of patients had baseline viral loads >100,000 copies/mL and 39% had CD4 cell counts <200 cells/mm 3 . Treatment outcomes through 144 weeks are presented in Table 5.

    Table 5.     Outcomes of Randomized Treatment (Study 903)
      At Week 48 At Week 144
    Outcomes
    VIREAD
    +3TC+EFV
    (N=299)
    Stavudine
    +3TC+EFV
    (N=301)
    VIREAD
    +3TC+EFV
    (N=299)
    Stavudine
    +3TC+EFV
    (N=301)
    % % % %
    Responder 1
    79% 82% 68% 62%
    Virologic failure 2
    6% 4% 10% 8%
         Rebound
    5% 3% 8% 7%
         Never suppressed
    0% 1% 0% 0%
         Added an
         antiretroviral agent
    1% 1% 2% 1%
    Death
    <1% 1% <1% 2%
    Discontinued due
    to adverse event
    6% 6% 8% 13%
    Discontinued for
    other reasons 3
    8% 7% 14% 15%
    1.Patients achieved and maintained confirmed HIV-1 RNA <400 copies/mL through Week 48 and 144.
    2.Includes confirmed viral rebound and failure to achieve confirmed <400 copies/mL through Week 48 and 144.
    3.Includes lost to follow-up, patient's withdrawal, noncompliance, protocol violation and other reasons.

    Achievement of plasma HIV-1 RNA concentrations of less than 400 copies/mL at week 144 was similar between the two treatment groups for the population stratified at baseline on the basis of HIV-1 RNA concentration (> or </= 100,000 copies/mL) and CD4 cell count (< or >/= 200 cells/mm 3 ). Through 144 weeks of therapy, 62% and 58% of patients in the VIREAD and stavudine arms, respectively achieved and maintained confirmed HIV-1 RNA <50 copies/mL. The mean increase from baseline in CD4 cell count was 263 cells/mm 3 for the VIREAD arm and 283 cells/mm 3 for the stavudine arm.

    Through 144 weeks, eleven patients in the VIREAD group and nine patients in the stavudine group experienced a new CDC Class C event.

    Resistance Analyses of VIREAD in Treatment Naïve Patients

    Genotypic analyses of patients with virologic failure showed development of efavirenz-associated and lamivudine-associated mutations to occur most frequently and with no difference between the treatment arms. The K65R mutation occurred in 8 patients on the VIREAD arm and in 2 patients on the stavudine arm. Of the 8 patients who developed K65R in the VIREAD arm through 144 weeks, 7 of these occurred in the first 48 weeks of treatment and one at week 96. Other mutations resulting in resistance to VIREAD were not identified in this study.

    Treatment-Experienced Patients

    Study 907:   VIREAD + Standard Background Therapy (SBT) Compared to Placebo + SBT

    Study 907 was a 24 week, double-blind placebo-controlled multicenter study of VIREAD added to a stable background regimen of antiretroviral agents in 550 treatment-experienced patients. After 24 weeks of blinded study treatment, all patients continuing on study were offered open-label VIREAD for an additional 24 weeks. Patients had a mean baseline CD4 cell count of 427 cells/mm 3 (range 23-1385), median baseline plasma HIV-1 RNA of 2340 (range 50-75,000) copies/mL, and mean duration of prior HIV-1 treatment was 5.4 years. Mean age of the patients was 42 years, 85% were male and 69% were Caucasian, 17% Black and 12% Hispanic.

    Changes from baseline in log 10 copies/mL plasma HIV-1 RNA levels over time up to week 48 are presented below in Figure 1.

    The percent of patients with HIV-1 RNA <400 copies/mL and outcomes of patients through 48 weeks are summarized in Table 6.

    Table 6.     Outcomes of Randomized Treatment (Study 907)
      0-24 weeks 0-48 weeks 24-48 weeks
    Outcomes VIREAD
    (N=368)
    %
    Placebo
    (N=182)
    %
    VIREAD
    (N=368)
    %
    Placebo
    Crossover to
    VIREAD
    (N=170)
    %
    HIV-1 RNA <400 copies/mL 1
    40% 11% 28% 30%
    Virologic failure 2
    53% 84% 61% 64%
    Discontinued due to adverse event
    3% 3% 5% 5%
    Discontinued for other reasons 3
    3% 3% 5% 1%
    1.Patients with HIV-1 RNA <400 copies/mL and no prior study drug discontinuation at Week 24 and 48 respectively.
    2.Patients with HIV-1 RNA >/=400 copies/mL efficacy failure or missing HIV -1 RNA at Week 24 and 48 respectively.
    3.Includes lost to follow up, patient withdrawal, noncompliance, protocol violation and other reasons.

    At 24 weeks of therapy, there was a higher proportion of patients in the VIREAD arm compared to the placebo arm with HIV-1 RNA <50 copies/mL (19% and 1%, respectively). Mean change in absolute CD4 counts by week 24 was +11 cells/mm 3 for the VIREAD group and -5 cells/mm 3 for the placebo group. Mean change in absolute CD4 counts by week 48 was +4 cells/mm 3 for the VIREAD group.

    Through week 24, one patient in the VIREAD group and no patients in the placebo arm experienced a new CDC Class C event.

    Genotypic Analyses of VIREAD in Patients with Previous Antiretroviral Therapy

    The virologic response to VIREAD therapy has been evaluated with respect to baseline viral genotype (N=222) in treatment experienced patients participating in two controlled trials.

    In two clinical studies, 94% of the participants evaluated had baseline HIV-1 isolates expressing at least one NRTI mutation. These included resistance mutations associated with zidovudine (M41L, D67N, K70R, L210W, T215Y/F or K219Q/E/N), the abacavir/emtricitabine/lamivudine-associated mutation (M184V), and others. In addition the majority of participants evaluated had mutations associated with either PI or NNRTI use. Virologic responses for patients in the genotype substudy were similar to the overall study results.

    Several exploratory analyses were conducted to evaluate the effect of specific mutations and mutational patterns on virologic outcome. Descriptions of numerical differences in HIV-1 RNA response are displayed in Table 7. Because of the large number of potential comparisons, statistical testing was not conducted.

    Varying degrees of cross-resistance of VIREAD to pre-existing zidovudine-associated mutations were observed and appeared to depend on the number of specific mutations. VIREAD-treated patients whose HIV-1 expressed 3 or more zidovudine-associated mutations that included either the M41L or L210W reverse transcriptase mutation showed reduced responses to VIREAD therapy; however, these responses were still improved compared with placebo. The presence of the D67N, K70R, T215Y/F or K219Q/E/N mutation did not appear to affect responses to VIREAD therapy. The HIV-1 RNA responses by number and type of baseline zidovudine-associated mutations are shown in Table 7.

    Table 7. HIV-1 RNA Response at Week 24 by Number of Baseline Zidovudine-Associated Mutations (Intent-To-Treat) 1
    Number of baseline
    zidovudine-associated
    mutations 2
    Change in HIV-1 RNA 3 (N)
    VIREAD 300 mg Placebo
    None -0.80 (68) -0.11 (29)
    Any  -0.50 (154) 0 (81)
    1-2 -0.66 (55) -0.04 (33)
    >/=3 including M41L or L210W -0.21 (57) +0.01 (29)
    >/=3 without M41L or L210W -0.67 (42) +0.07 (19)
    1.Genotypic testing performed by Virco Laboratories and Visible Genetics TruGene™ technology.
    2.M41L, D67N, K70R, L210W, T215Y/F or K219Q/E/N in RT.
    3.Average HIV-1 RNA change from baseline through week 24 (DAVG 24 ) in log 10 copies/mL.

    In the protocol defined analyses, virologic response to VIREAD was not reduced in patients with HIV-1 that expressed the abacavir/emtricitabine/lamivudine-associated M184V mutation. In the absence of zidovudine-associated mutations, patients with the M184V mutation receiving VIREAD showed a -0.84 log10 copies/mL decrease in their HIV-1 RNA relative to placebo. In the presence of zidovudine-associated mutations, the M184V mutation did not affect the mean HIV-1 RNA responses to VIREAD treatment. HIV-1 RNA responses among these patients were durable through week 48.

    Phenotypic Analyses of VIREAD in Patients with Previous Antiretroviral Therapy

    The virologic response to VIREAD therapy has been evaluated with respect to baseline phenotype (N=100) in treatment-experienced patients participating in two controlled trials. Phenotypic analysis of baseline HIV-1 from patients in these studies demonstrated a correlation between baseline susceptibility to VIREAD and response to VIREAD therapy. Table 8 summarizes the HIV-1 RNA response by baseline VIREAD susceptibility.

    Table 8.     HIV-1 RNA Response at Week 24 by Baseline VIREAD Susceptibility (Intent-To-Treat) 1
    Baseline VIREAD Susceptibility   2 Change in HIV-1 RNA 3 (N)
    </=1 -0.74 (35)
    >1 and </=3 -0.56 (49)
    >3 and </=4 -0.3 (7)  
    </=4 -0.61 (91)
    >4 -0.12 (9)
    1.Tenofovir susceptibility was determined by recombinant phenotypic Antivirogram™ assay (Virco).
    2.Fold change in susceptibility from wild-type.
    3.Average HIV-1 RNA change from baseline through week 24 (DAVG 24 ) in log 10 copies/mL.

    CONTRAINDICATIONS

    VIREAD is contraindicated in patients with previously demonstrated hypersensitivity to any of the components of the product.

    WARNINGS

    Lactic Acidosis/Severe Hepatomegaly with Steatosis

    Lactic acidosis and severe hepatomegaly with steatosis, including fatal cases, have been reported with the use of nucleoside analogs alone or in combination with other antiretrovirals. A majority of these cases have been in women. Obesity and prolonged nucleoside exposure may be risk factors. Particular caution should be exercised when administering nucleoside analogs to any patient with known risk factors for liver disease; however, cases have also been reported in patients with no known risk factors. Treatment with VIREAD should be suspended in any patient who develops clinical or laboratory findings suggestive of lactic acidosis or pronounced hepatotoxicity (which may include hepatomegaly and steatosis even in the absence of marked transaminase elevations).

    Patients Co-infected with HIV and Hepatitis B Virus

    It is recommended that all patients with HIV be tested for the presence of chronic hepatitis B virus (HBV) before initiating antiretroviral therapy. VIREAD is not indicated for the treatment of chronic HBV infection and the safety and efficacy of VIREAD have not been established in patients co-infected with HBV and HIV. Severe acute exacerbations of hepatitis B have been reported in patients who are co-infected with HBV and HIV and have discontinued VIREAD. Hepatic function should be monitored closely with both clinical and laboratory follow-up for at least several months in patients who discontinue VIREAD and are co-infected with HIV and HBV. If appropriate, initiation of anti-hepatitis B therapy may be warranted.

    Renal Impairment

    Tenofovir is principally eliminated by the kidney. Dosing interval adjustment is recommended in all patients with creatinine clearance <50 mL/min (see DOSAGE AND ADMINISTRATION ). No safety data are available in patients with renal dysfunction who received VIREAD using these dosing guidelines.

    Renal impairment, including cases of acute renal failure and Fanconi syndrome (renal tubular injury with severe hypophosphatemia), has been reported in association with the use of VIREAD (see Adverse Reactions - Post Marketing Experience ). The majority of these cases occurred in patients with underlying systemic or renal disease, or in patients taking nephrotoxic agents, however, some cases occurred in patients without identified risk factors.

    VIREAD should be avoided with concurrent or recent use of a nephrotoxic agent. Patients at risk for, or with a history of, renal dysfunction and patients receiving concomitant nephrotoxic agents should be carefully monitored for changes in serum creatinine and phosphorus.

    PRECAUTIONS

    Drug Interactions

    When administered with VIREAD, C max and AUC of didanosine administered as either the buffered or enteric-coated formulation increased significantly (see Table 4). The mechanism of this interaction is unknown. Higher didanosine concentrations could potentiate didanosine-associated adverse events, including pancreatitis and neuropathy. In adults weighing >60kg, the didanosine dose should be reduced to 250 mg when it is co-administered with VIREAD. Data are not available to recommend a dose adjustment of didanosine for patients weighing <60 kg. When co-administered, VIREAD and didanosine EC may be taken under fasted conditions or with a light meal (<400 kcal, 20% fat). Co-administration of didanosine buffered tablet formulation with VIREAD should be under fasted conditions. Co-administration of VIREAD and didanosine should be undertaken with caution and patients receiving this combination should be monitored closely for didanosine-associated adverse events. Didanosine should be discontinued in patients who develop didanosine-associated adverse events.

    Since tenofovir is primarily eliminated by the kidneys, co-administration of VIREAD with drugs that reduce renal function or compete for active tubular secretion may increase serum concentrations of tenofovir and/or increase the concentrations of other renally eliminated drugs. Some examples include, but are not limited to adefovir dipivoxil, cidofovir, acyclovir, valacyclovir, ganciclovir and valganciclovir.

    Higher tenofovir concentrations could potentiate VIREAD-associated adverse events, including renal disorders.

    Atazanavir and lopinavir/ritonavir have been shown to increase tenofovir concentrations. The mechanism of this interaction is unknown. Patients receiving atazanavir and lopinavir/ritonavir and VIREAD should be monitored for VIREAD-associated adverse events. VIREAD should be discontinued in patients who develop VIREAD-associated adverse events.

    VIREAD decreases the AUC and C min of atazanavir. When coadministered with VIREAD, it is recommended that atazanavir 300 mg is given with ritonavir 100 mg. Atazanavir without ritonavir should not be coadministered with VIREAD.

    Bone Effects

    In study 903 through 144 weeks, decreases from baseline in bone mineral density (BMD) were seen at the lumbar spine and hip in both arms of the study. At week 144, there was a significantly greater mean percentage decrease from baseline in BMD at the lumbar spine in patients receiving VIREAD + lamivudine + efavirenz (-2.2% ± 3.9) compared with patients receiving stavudine + lamivudine + efavirenz (-1.0% ± 4.6). Changes in BMD at the hip were similar between the two treatment groups (-2.8% ± 3.5 in the VIREAD group vs. -2.4% ± 4.5 in the stavudine group). In both groups, the majority of the reduction in BMD occurred in the first 24-48 weeks of the study and this reduction was sustained through week 144. Twenty-eight percent of VIREAD-treated patients vs. 21% of the stavudine-treated patients lost at least 5% of BMD at the spine or 7% of BMD at the hip. Clinically relevant fractures (excluding fingers and toes) were reported in 4 patients in the VIREAD group and 6 patients in the stavudine group. In addition, there were significant increases in biochemical markers of bone metabolism (serum bone-specific alkaline phosphatase, serum osteocalcin, serum C-telopeptide and urinary N-telopeptide) in the VIREAD group relative to the stavudine group, suggesting increased bone turnover. Serum parathyroid hormone levels and 1,25 Vitamin D levels were also higher in the VIREAD group. Except for bone specific alkaline phosphatase, these changes resulted in values that remained within the normal range. The effects of VIREAD-associated changes in BMD and biochemical markers on long-term bone health and future fracture risk are unknown.

    Bone monitoring should be considered for HIV infected patients who have a history of pathologic bone fracture or are at risk for osteopenia. Although the effect of supplementation with calcium and vitamin D was not studied, such supplementation may be beneficial for all patients. If bone abnormalities are suspected then appropriate consultation should be obtained.

    Fat Redistribution

    Redistribution/accumulation of body fat including central obesity, dorsocervical fat enlargement (buffalo hump), peripheral wasting, facial wasting, breast enlargement, and "cushingoid appearance" have been observed in patients receiving antiretroviral therapy. The mechanism and long-term consequences of these events are currently unknown. A causal relationship has not been established.

    Immune Reconstitution Syndrome

    Immune reconstitution syndrome has been reported in patients treated with combination antiretroviral therapy, including VIREAD. During the initial phase of combination antiretroviral treatment, patients whose immune system responds may develop an inflammatory response to indolent or residual opportunistic infections (such as Mycobacterium avium infection, cytomegalovirus, Pneumocystis jirovecii pneumonia (PCP), or tuberculosis), which may necessitate further evaluation and treatment.

    Animal Toxicology

    Tenofovir and tenofovir disoproxil fumarate administered in toxicology studies to rats, dogs and monkeys at exposures (based on AUCs) greater than or equal to 6 fold those observed in humans caused bone toxicity. In monkeys the bone toxicity was diagnosed as osteomalacia. Osteomalacia observed in monkeys appeared to be reversible upon dose reduction or discontinuation of tenofovir. In rats and dogs, the bone toxicity manifested as reduced bone mineral density. The mechanism(s) underlying bone toxicity is unknown.

    Evidence of renal toxicity was noted in 4 animal species. Increases in serum creatinine, BUN, glycosuria, proteinuria, phosphaturia and/or calciuria and decreases in serum phosphate were observed to varying degrees in these animals. These toxicities were noted at exposures (based on AUCs) 2-20 times higher than those observed in humans. The relationship of the renal abnormalities, particularly the phosphaturia, to the bone toxicity is not known.

    Carcinogenesis, Mutagenesis, Impairment of Fertility

    Long-term oral carcinogenicity studies of tenofovir disoproxil fumarate in mice and rats were carried out at exposures up to approximately 16 times (mice) and 5 times (rats) those observed in humans at the therapeutic dose for HIV infection. At the high dose in female mice, liver adenomas were increased at exposures 16 times that in humans. In rats, the study was negative for carcinogenic findings at exposures up to 5 times that observed in humans at the therapeutic dose.

    Tenofovir disoproxil fumarate was mutagenic in the in vitro mouse lymphoma assay and negative in an in vitro bacterial mutagenicity test (Ames test). In an in vivo mouse micronucleus assay, tenofovir disoproxil fumarate was negative when administered to male mice.

    There were no effects on fertility, mating performance or early embryonic development when tenofovir disoproxil fumarate was administered to male rats at a dose equivalent to 10 times the human dose based on body surface area comparisons for 28 days prior to mating and to female rats for 15 days prior to mating through day seven of gestation. There was, however, an alteration of the estrous cycle in female rats.

    Pregnancy

    Pregnancy category B:

    Reproduction studies have been performed in rats and rabbits at doses up to 14 and 19 times the human dose based on body surface area comparisons and revealed no evidence of impaired fertility or harm to the fetus due to tenofovir. There are, however, no adequate and well-controlled studies in pregnant women. Because animal reproduction studies are not always predictive of human response, VIREAD should be used during pregnancy only if clearly needed.

    Antiretroviral Pregnancy Registry:   To monitor fetal outcomes of pregnant women exposed to VIREAD, an Antiretroviral Pregnancy Registry has been established. Healthcare providers are encouraged to register patients by calling 1-800-258-4263.

    Nursing Mothers:    The Centers for Disease Control and Prevention recommend that HIV-infected mothers not breast-feed their infants to avoid risking postnatal transmission of HIV. Studies in rats have demonstrated that tenofovir is secreted in milk. It is not known whether tenofovir is excreted in human milk. Because of both the potential for HIV transmission and the potential for serious adverse reactions in nursing infants, mothers should be instructed not to breast-feed if they are receiving VIREAD.

    Pediatric Use

    Safety and effectiveness in pediatric patients have not been established.

    Geriatric Use

    Clinical studies of VIREAD did not include sufficient numbers of subjects aged 65 and over to determine whether they respond differently from younger subjects. In general, dose selection for the elderly patient should be cautious, keeping in mind the greater frequency of decreased hepatic, renal, or cardiac function, and of concomitant disease or other drug therapy.

    ADVERSE REACTIONS

    Clinical Trials:    More than 12,000 patients have been treated with VIREAD alone or in combination with other antiretroviral medicinal products for periods of 28 days to 215 weeks in Phase I-III clinical trials and expanded access studies. A total of 1,287 patients have received VIREAD 300 mg once daily in Phase I-III clinical trials; over 11,000 patients have received VIREAD in expanded access studies.

    Treatment-Naïve Patients

    Treatment-Emergent Adverse Events:    The most common adverse reactions seen in a double-blind comparative controlled study in which 600 treatment-na[iuml ]ve patients received VIREAD (N=299) or stavudine (N=301) in combination with lamivudine and efavirenz for 144 weeks (Study 903) were mild to moderate gastrointestinal events and dizziness.

    Mild adverse events (Grade 1) were common with a similar incidence in both arms, and included dizziness, diarrhea and nausea. Selected treatment-emergent moderate to severe adverse events are summarized in Table 9.

    Table 9.Selected Treatment-Emergent Adverse Events
    (Grades 2-4) Reported in >/=5% in Any Treatment Group in Study 903 (0-144 weeks)
      VIREAD
    +3TC+EFV
    d4T+3TC
    +EFV
      N=299 N=301
    Body as a Whole
       
       Headache
    14% 17%
       Pain
    13% 12%
       Fever
    8% 7%
       Abdominal Pain
    7% 12%
       Back Pain
    9% 8%
       Asthenia
    6% 7%
    Digestive System
       
       Diarrhea
    11% 13%
       Nausea
    8% 9%
       Dyspepsia
    4% 5%
       Vomiting
    5% 9%
    Metabolic Disorders
       
       Lipodystrophy 1
    1% 8%
    Musculoskeletal
       
       Arthralgia
    5% 7%
       Myalgia
    3% 5%
    Nervous System
       
       Depression
    11% 10%
       Insomnia
    5% 8%
       Dizziness
    3% 6%
       Peripheral neuropathy 2
    1% 5%
       Anxiety
    6% 6%
    Respiratory
       
       Pneumonia
    5% 5%
    Skin and Appendages
       
       Rash Event 3
    18% 12%
    1. Lipodystrophy represents a variety of investigator-described adverse events not a protocol-defined syndrome
    2.Peripheral neuropathy includes peripheral neuritis and neuropathy
    3.Rash event includes rash, pruritus, maculopapular rash, urticaria, vesiculobullous rash, and pustular rash.

    Laboratory Abnormalities:    With the exception of cholesterol and triglyceride elevations that were more common in the stavudine group (15% and 14%) compared with VIREAD (5% and 3%) respectively, laboratory abnormalities observed in this study occurred with similar frequency in the VIREAD and stavudine treatment arms. A summary of Grade 3 and 4 laboratory abnormalities is provided in Table 10.

    Table 10.     Grade 3/4 Laboratory Abnormalities
    Reported in >/= 1% of VIREAD-Treated
    Patients in Study 903 (0-144 weeks)
      VIREAD
    +3TC+EFV
    d4T+3TC
    +EFV
      N=299 N=301
    Any >/= Grade 3 Laboratory
    Abnormality
    36% 42%
    Total Cholesterol
    (> 300 mg/dL)
    5% 15%
    Creatine Kinase
    (M: >990 U/L)
    (F: >845 U/L)
    12% 12%
    Serum Amylase
    (>175 U/L)
    9% 8%
    AST
    (M: >180 U/L)
    (F: >170 U/L)
    5% 7%
    ALT
    (M: >215 U/L)
    (F: >170 U/L)
    4% 5%
    Hematuria (>100 RBC/HPF)
    7% 7%
    Neutrophil (<750/mm 3 )
    3% 1%
    Triglyceride (>750 mg/dL)
    3% 14%

    Treatment-Experienced Patients

    Treatment-Emergent Adverse Events:    The adverse reactions seen in treatment experienced patients were generally consistent with those seen in treatment naïve patients including mild to moderate gastrointestinal events, such as nausea, diarrhea, vomiting and flatulence. Less than 1% of patients discontinued participation in the clinical studies due to gastrointestinal adverse events (Study 907).

    A summary of moderate to severe, treatment-emergent adverse events that occurred during the first 48 weeks of Study 907 is provided in Table 11.

    Table 11.     Selected Treatment-Emergent Adverse Events (Grades 2-4) Reported
    in >/=3% in Any Treatment Group in Study 907 (0-48 weeks)
      VIREAD
    (N=368)
    (Week 0-24)
    Placebo
    (N=182)
    (Week 0-24)
    VIREAD
    (N=368)
    (Week 0-48)
    Placebo
    Crossover to
    VIREAD
    (N=170)
    (Week 24-48)
    Body as a Whole
           
         Asthenia
    7% 6% 11% 1%
         Pain
    7% 7% 12% 4%
         Headache
    5% 5% 8% 2%
         Abdominal Pain
    4% 3% 7% 6%
         Back Pain
    3% 3% 4% 2%
         Chest Pain
    3% 1% 3% 2%
         Fever
    2% 2% 4% 2%
    Digestive System
           
         Diarrhea
    11% 10% 16% 11%
         Nausea
    8% 5% 11% 7%
         Vomiting
    4% 1% 7% 5%
         Anorexia
    3% 2% 4% 1%
         Dyspepsia
    3% 2% 4% 2%
         Flatulence
    3% 1% 4% 1%
    Respiratory
           
        Pneumonia
    2% 0% 3% 2%
    Nervous System
           
         Depression
    4% 3% 8% 4%
         Insomnia
    3% 2% 4% 4%
         Peripheral
    3% 3% 5% 2%
         Neuropathy 1
           
         Dizziness
    1% 3% 3% 1%
    Skin and Appendage
           
         Rash Event 2
    5% 4% 7% 1%
         Sweating
    3% 2% 3% 1%
    Musculoskeletal
           
         Myalgia
    3% 3% 4% 1%
    Metabolic
           
         Weight Loss
    2% 1% 4% 2%
    1.Peripheral neuropathy includes peripheral neuritis and neuropathy.
    2.Rash event includes rash, pruritus, maculopapular rash, urticaria, vesiculobullous rash, and pustular rash.

    Laboratory Abnormalities:    Laboratory abnormalities observed in this study occurred with similar frequency in the VIREAD and placebo-treated groups. A summary of Grade 3 and 4 laboratory abnormalities is provided in Table 12.

    Table 12. Grade 3/4 Laboratory Abnormalities Reported in >/=1% of VIREAD-Treated Patients in Study 907 (0-48 weeks)
      VIREAD
    (N=368)
    (Week 0-24)
    Placebo
    (N=182)
    (Week 0-24)
    VIREAD
    (N=368)
    (Week 0-48)
    Placebo
    Crossover to
    VIREAD
    (N=170)
    (Week 24-48)
      (%) (%) (%) (%)
    Any >/= Grade 3 Laboratory Abnormality
    25% 38% 35% 34%
    Triglycerides (>750 mg/dL)
    8% 13% 11% 9%
    Creatine Kinase
    (M: >990U/L)
    (F: >845 U/L)
    7% 14% 12% 12%
    Serum Amylase (>175 U/L)
    6% 7% 7% 6%
    Urine Glucose (>/=3+)
    3% 3% 3% 2%
    AST
    (M: >180 U/L)
    (F: >170 U/L)
    3% 3% 4% 5%
    ALT
    (M: >215 U/L)
    (F: >170 U/L)
    2% 2% 4% 5%
    Serum Glucose (>250 U/L)
    2% 4% 3% 3%
    Neutrophils (<750/mm 3 )
    1% 1% 2% 1%

    Post Marketing Experience:    In addition to adverse events reported from clinical trials, the following events have been identified during post-approval use of VIREAD. Because they are reported voluntarily from a population of unknown size, estimates of frequency cannot be made. These events have been chosen for inclusion due to a combination of their seriousness, frequency of reporting or potential causal connection to VIREAD.

    IMMUNE SYSTEM DISORDERS

    Allergic reaction

    METABOLISM AND NUTRITION DISORDERS

    Hypophosphatemia, Lactic acidosis

    RESPIRATORY, THORACIC, AND MEDIASTINAL DISORDERS

    Dyspnea

    GASTROINTESTINAL DISORDERS

    Abdominal pain, Increased amylase, Pancreatitis

    HEPATOBILIARY DISORDERS

    Increased liver enzymes, Hepatitis

    RENAL AND URINARY DISORDERS

    Renal insufficiency, Renal failure, Acute renal failure, Fanconi syndrome, Proximal tubulopathy, Proteinuria, Increased creatinine, Acute tubular necrosis, Nephrogenic diabetes insipidus

    OVERDOSAGE

    Limited clinical experience at doses higher than the therapeutic dose of VIREAD 300 mg is available. In Study 901, 600 mg tenofovir disoproxil fumarate was administered to 8 patients orally for 28 days. No severe adverse reactions were reported. The effects of higher doses are not known.

    If overdose occurs the patient must be monitored for evidence of toxicity, and standard supportive treatment applied as necessary.

    Tenofovir is efficiently removed by hemodialysis with an extraction coefficient of approximately 54%. Following a single 300 mg dose of VIREAD, a four-hour hemodialysis session removed approximately 10% of the administered tenofovir dose.

    DOSAGE AND ADMINISTRATION

    The dose of VIREAD is 300 mg once daily taken orally, without regard to food.

    Dose Adjustment for Renal Impairment

    Significantly increased drug exposures occurred when VIREAD was administered to patients with moderate to severe renal impairment (See PHARMACOKINETICS ). The dosing interval of VIREAD should be adjusted in patients with baseline creatinine clearance <50 mL/min using the recommendations in Table 13. The safety and effectiveness of these dosing interval adjustment recommendations have not been clinically evaluated, therefore, clinical response to treatment and renal function should be closely monitored in these patients.

    Table 13.      Dosage Adjustment for Patients with Altered Creatinine Clearance
      Creatinine Clearance (mL/min) 1 Hemodialysis Patients
      >/=50 30-49 10-29  
    Recommended 300 mg
    Dosing Interval
    Every 24
    hours
    Every 48
    hours
    Twice
    a week
    Every 7 days or after a total of approximately 12 hours of
    dialysis 2
    1.Calculated using ideal (lean) body weight.
    2.Generally once weekly assuming three hemodialysis sessions a week of approximately 4 hours duration. VIREAD should be administered following completion of dialysis.

    The pharmacokinetics of tenofovir have not been evaluated in non-hemodialysis patients with creatinine clearance <10 mL/min; therefore, no dosing recommendation is available for these patients.

    HOW SUPPLIED

    VIREAD is available as tablets. Each tablet contains 300 mg of tenofovir disoproxil fumarate, which is equivalent to 245 mg of tenofovir disoproxil. The tablets are almond-shaped, light blue, film-coated, and debossed with "GILEAD" and "4331" on one side and with "300" on the other side. They are packaged as follows: Bottles of 30 tablets (NDC 61958-0401-1) containing a desiccant (silica gel canister or sachet) and closed with child-resistant closure.

    Store at 25 °C (77 °F), excursions permitted to 15-30 °C (59-86 °F) (see USP Controlled Room Temperature).

    Do not use if seal over bottle opening is broken or missing.

    Gilead Sciences, Inc.

    Foster City, CA 94404

    May 2005 VIREAD is a registered trademark of Gilead Sciences, Inc

    ©2001-2005, Gilead Sciences, Inc.

    9427-G0701-26-GS03

    GILEAD

    VIREAD®

    (tenofovir disoproxil fumarate) Tablets

    Patient Information

    VIREAD (VEER ee ad)

    Generic Name:   tenofovir disoproxil fumarate (te NOE' fo veer dye soe PROX il FYOU-mar-ate)

    Read this leaflet carefully before you start taking VIREAD. Also, read it each time you get your VIREAD prescription refilled, in case something has changed. This information does not take the place of talking with your healthcare provider when you start this medicine and at check ups. You should stay under a healthcare provider's care when taking VIREAD. Do not change or stop your medicine without first talking with your healthcare provider. Talk to your healthcare provider if you have any questions about VIREAD.

    What is VIREAD and how does it work?

    VIREAD is a type of medicine called an HIV-1 (human immunodeficiency virus) nucleotide analog reverse transcriptase inhibitor (NRTI). VIREAD is always used in combination with other anti-HIV medicines to treat people with HIV-1 infection. VIREAD is for adults age 18 and older.

    HIV infection destroys CD4 (T) cells, which are important to the immune system. After a large number of T cells are destroyed, acquired immune deficiency syndrome (AIDS) develops.

    VIREAD helps to block HIV-1 reverse transcriptase, a chemical in your body (enzyme) that is needed for HIV-1 to multiply. VIREAD lowers the amount of HIV-1 in the blood (called viral load) and may help to increase the number of T cells (called CD4 cells). Lowering the amount of HIV-1 in the blood lowers the chance of death or infections that happen when your immune system is weak (opportunistic infections).

    Does VIREAD cure HIV-1 or AIDS?

    VIREAD does not cure HIV-1 infection or AIDS. The long-term effects of VIREAD are not known at this time. People taking VIREAD may still get opportunistic infections or other conditions that happen with HIV-1 infection. Opportunistic infections are infections that develop because the immune system is weak. Some of these conditions are pneumonia, herpes virus infections, and Mycobacterium avium complex (MAC) infections.

    Does VIREAD reduce the risk of passing HIV-1 to others?

    VIREAD does not reduce the risk of passing HIV-1 to others through sexual contact or blood contamination. Continue to practice safe sex and do not use or share dirty needles.

    Who should not take VIREAD?

    Together with your healthcare provider, you need to decide whether VIREAD is right for you.

    Do not take VIREAD if

    • you are allergic to VIREAD or any of its ingredients

    What should I tell my healthcare provider before taking VIREAD?

    Tell your healthcare provider

    • If you are pregnant or planning to become pregnant:    The effects of VIREAD on pregnant women or their unborn babies are not known.
    • If you are breast-feeding:    Do not breast-feed if you are taking VIREAD. Do not breast-feed if you have HIV. If you are a woman who has or will have a baby, talk with your healthcare provider about the best way to feed your baby. If your baby does not already have HIV, there is a chance that the baby can get HIV through breast-feeding.
    • If you have kidney or bone problems
    • If you have liver problems including Hepatitis B Virus infection
    • Tell your healthcare provider about all your medical conditions
    • TELL YOUR HEALTHCARE PROVIDER ABOUT ALL THE MEDICINES YOU TAKE, INCLUDING PRESCRIPTION AND NON-PRESCRIPTION MEDICINES AND DIETARY SUPPLEMENTS. VIREAD MAY INCREASE THE AMOUNT OF VIDEX® (DIDANOSINE) IN YOUR BLOOD. YOU MAY NEED TO BE FOLLOWED MORE CAREFULLY IF YOU ARE TAKING THESE TWO DRUGS TOGETHER. IF YOU ARE TAKING VIDEX (DIDANOSINE) AND VIREAD TOGETHER YOUR HEALTHCARE PROVIDER MAY NEED TO REDUCE YOUR DOSE OF VIDEX (DIDANOSINE).
      VIREAD MAY DECREASE THE AMOUNT OF REYATAZ® (ATAZANAVIR) IN YOUR BLOOD. IF YOU ARE TAKING VIREAD AND REYATAZ (ATAZANAVIR) TOGETHER YOU SHOULD ALSO BE TAKING NORVIR® (RITONAVIR).
      IT IS A GOOD IDEA TO KEEP A COMPLETE LIST OF ALL THE MEDICINES THAT YOU TAKE. MAKE A NEW LIST WHEN MEDICINES ARE ADDED OR STOPPED. GIVE COPIES OF THIS LIST TO ALL OF YOUR HEALTHCARE PROVIDERS EVERY TIME YOU VISIT YOUR HEALTHCARE PROVIDER OR FILL A PRESCRIPTION.

    How should I take VIREAD?

    • Stay under a healthcare provider's care when taking VIREAD. Do not change your treatment or stop treatment without first talking with your healthcare provider.
    • Take VIREAD every day exactly as your healthcare provider prescribed it. Follow the directions from your healthcare provider, exactly as written on the label. Set up a dosing schedule and follow it carefully.
    • The usual dose of VIREAD is 1 tablet once a day, in combination with other anti-HIV medicines. If you have kidney problems, your healthcare provider may recommend that you take VIREAD less frequently.
    • VIREAD may be taken with or without a meal.
    • When your VIREAD supply starts to run low, get more from your healthcare provider or pharmacy. This is very important because the amount of virus in your blood may increase if the medicine is stopped for even a short time. The virus may develop resistance to VIREAD and become harder to treat.
    • Only take medicine that has been prescribed specifically for you. Do not give VIREAD to others or take medicine prescribed for someone else.

    What should I do if I miss a dose of VIREAD?

    It is important that you do not miss any doses. If you miss a dose of VIREAD, take it as soon as possible and then take your next scheduled dose at its regular time. If it is almost time for your next dose, do not take the missed dose. Wait and take the next dose at the regular time. Do not double the next dose.

    What happens if I take too much VIREAD?

    If you suspect that you took more than the prescribed dose of VIREAD, contact your local poison control center or emergency room right away.

    As with all medicines, VIREAD should be kept out of reach of children.

    What should I avoid while taking VIREAD?

    • Do not breast-feed. See "What should I tell my healthcare provider before taking VIREAD?"

    What are the possible side effects of VIREAD?

    • Clinical studies:   The most common side effects of VIREAD are: diarrhea, nausea, vomiting, and flatulence (intestinal gas).
    • Marketing experience:   Other side effects reported since VIREAD has been marketed include: weakness, inflammation of the pancreas, low blood phosphate, dizziness, shortness of breath, and rash.
    • Some patients treated with VIREAD have had kidney problems. If you have had kidney problems in the past or need to take another drug that can cause kidney problems, your healthcare provider may need to perform additional blood tests.
    • Laboratory tests show changes in the bones of patients treated with VIREAD. It is not known whether long-term use of VIREAD will cause damage to your bones. If you have had bone problems in the past, your healthcare provider may need to perform additional tests or may suggest additional medication.
    • Some patients taking antiviral drugs like VIREAD have developed a condition called lactic acidosis (a buildup in the blood of lactic acid, the same substance that causes your muscles to burn during heavy exercise). Symptoms of lactic acidosis include nausea, vomiting, unusual or unexpected stomach discomfort, and weakness. If you notice these symptoms or if your medical condition changes suddenly, call your healthcare provider right away.
    • Changes in body fat have been seen in some patients taking anti-HIV medicine. These changes may include increased amount of fat in the upper back and neck ("buffalo hump"), breast, and around the main part of your body (trunk). Loss of fat from the legs, arms and face may also happen. The cause and long term health effects of these conditions are not known at this time.
    • If you have hepatitis B virus (HBV) infection, you may have a "flare-up" of hepatitis B, in which the disease suddenly returns in a worse way than before if you stop taking VIREAD. VIREAD is not for the treatment of hepatitis B virus infection.
    • There have been other side effects in patients taking VIREAD. However, these side effects may have been due to other medicines that patients were taking or to the illness itself. Some of these side effects can be serious.
    • This list of side effects is not complete. If you have questions about side effects, ask your healthcare provider. You should report any new or continuing symptoms to your healthcare provider right away. Your healthcare provider may be able to help you manage these side effects.

    How do I store VIREAD?

    • Keep VIREAD and all other medications out of reach of children.
    • Store VIREAD at room temperature 77 °F (25 °C). It should remain stable until the expiration date printed on the label.
    • Do not keep your medicine in places that are too hot or cold.
    • Do not keep medicine that is out of date or that you no longer need. If you throw any medicines away make sure that children will not find them.

    General advice about prescription medicines:

    TALK TO YOUR HEALTHCARE PROVIDER IF YOU HAVE ANY QUESTIONS ABOUT THIS MEDICINE OR YOUR CONDITION. MEDICINES ARE SOMETIMES PRESCRIBED FOR PURPOSES OTHER THAN THOSE LISTED IN A PATIENT INFORMATION LEAFLET. IF YOU HAVE ANY CONCERNS ABOUT THIS MEDICINE, ASK YOUR HEALTHCARE PROVIDER. YOUR HEALTHCARE PROVIDER OR PHARMACIST CAN GIVE YOU INFORMATION ABOUT THIS MEDICINE THAT WAS WRITTEN FOR HEALTH CARE PROFESSIONALS. DO NOT USE THIS MEDICINE FOR A CONDITION FOR WHICH IT WAS NOT PRESCRIBED. DO NOT SHARE THIS MEDICINE WITH OTHER PEOPLE.

    DO NOT USE IF SEAL OVER BOTTLE OPENING IS BROKEN OR MISSING.

    May 2005

    ©2001-2005, Gilead Sciences, Inc.

    GILEAD








    - To bookmark this page (add it to your favorites), please click the image to the left.
    - If you wish to link to this page, you can do so by referring to the URL address below this line.

    https://theodora.com/drugs/viread_tablets_gilead.html

    This page was last modified 30-JUN-07
    Copyright © 1995-2021 ITA all rights reserved.