MOBILE VIEW  | 

AIDS ANTIVIRAL NUCLEOSIDES

Classification   |    Detailed evidence-based information

Therapeutic Toxic Class

    A) AIDS pyrimidine and thymidine nucleoside antiviral drugs are potent inhibitors in vitro of the replication of HIV in human T cells. Following activation to the triphosphate form, these drugs inhibit reverse transcriptase (viral DNA polymerase), which is essential in HIV's replicative cycle. These agents have potent activity against a broad spectrum of retroviruses, including human immunodeficiency virus type 2, human T-cell lymphotropic virus Type 1, animal lentiviruses, and murine retroviruses.

Specific Substances

    A) GENERAL TERMS
    1) Antiviral nucleosides, AIDS
    2) Nucleosides, AIDS antiviral
    3) HIV, AIDS antiviral nucleosides
    ABACAVIR
    1) ((1S,4R)-4-(2-Amino-6-(cyclopropylamino)-9H-purin-9-yl) cyclopent-2-enyl)methanol succinate
    2) 1592U89
    3) 1592
    4) ABC
    5) Ziagen
    6) Molecular Formula: C14-H18-N6-O, C4-H6-O4
    7) CAS 136470-78-5 (abacavir)
    8) CAS 168146-84-7 (abacavir succinate)
    ADEFOVIR
    1) ([2-(6-Amino-9H-purin-9-yl)ethoxy]-methyl)phosphonic acid; 9-[2-(Phosphonomethoxy)ethyl]adenine
    2) Bis-POM PMEA (adefovir dipivoxil)
    3) GS-0393 (adefovir)
    4) GS-0840 (adefovir dipivoxil)
    5) Phosphonylmethoxyethyl-a (adefovir)
    6) PMEA (adefovir)
    7) Preveon(R) (adefovir)
    8) CAS 106941-25-7 (adefovir)
    9) CAS 142340-99-6 (adefovir dipivoxil)
    EMTRICITABINE
    1) 524W91
    2) BW 524W91
    3) Coviracil(R)
    4) FTC
    LAMIVUDINE
    1) 2-(Hydroxymethyl)-1,3-oxathiolan-5-yl cytosine
    2) 3TC
    3) GR-109714X
    4) Molecular Formula: C1-H11-N3-O3-S
    5) CAS 131086-21-0
    6) CAS 134678-17-4
    STAVUDINE
    1) 2,3-didehydro-3-deoxythymidine
    2) d4T
    3) Molecular Formula: C10-H12-N2-O4
    4) CAS 3056-17-5
    TENOFOVIR
    1) (R)-PMPA
    2) PMPA
    3) GS-1278
    4) PMPA-IV
    5) Tenofovir disoproxil fumarate
    6) Tenofovir disoproxyl fumarate
    7) Oral PMPA
    8) Bis-POC-PMPA
    ZALCITABINE
    1) 2,3-Dideoxycytidine
    2) DDC
    3) ddC
    4) ddCyd
    5) Dideoxycytidine
    6) NSC 606170
    7) Ro-24-2027
    8) Ro-24-2027/000
    9) Molecular Formula: C9-H13-N3-O3
    10) CAS 7481-89-2

    1.2.1) MOLECULAR FORMULA
    1) ABACAVIR SULFATE: (C14H18N6O)2.H2SO4
    2) ADEFOVIR DIPIVOXIL: C20H32N5O8P
    3) EMTRICITABINE: C8H10FN3O3S
    4) LAMIVUDINE: C8H11N3O3S
    5) STAVUDINE: C10H12N2O4
    6) TENOFOVIR ALAFENAMIDE FUMARATE: C21H29O5N6P.1/2(C4H4O4)
    7) TENOFOVIR DISOPROXIL FUMARATE: C19H30N5O10P.C4H4O4

Available Forms Sources

    A) FORMS
    1) ABACAVIR
    a) As a single agent, abacavir is available as 300 mg tablets and as a 20 mg/mL oral solution (Prod Info ZIAGEN(R) oral tablets, solution, 2008).
    b) As part of a combination regimen, abacavir is available in combination with lamivudine as 600 mg abacavir/300 mg lamivudine tablets (Prod Info EPZICOM(R) oral tablets, 2009).
    c) As part of a combination regimen, abacavir is available in combination with lamivudine and zidovudine as 300 mg abacavir/150 mg lamivudine/300 mg zidovudine tablets (Prod Info TRIZIVIR(R) oral tablets, 2009).
    2) EMTRICITABINE
    a) As a single agent, emtricitabine is available as 200 mg capsules and as a 10 mg/mL oral solution (Prod Info EMTRIVA(R) capsules, oral solution, 2008).
    b) As part of a combination regimen, emtricitabine is available in combination with tenofovir as 200 mg emtricitabine/300 mg tenofovir disoproxil fumarate tablets (Prod Info TRUVADA(R) oral tablets, 2009).
    3) LAMIVUDINE
    a) As a single agent, lamivudine is available as 150 mg and 300 mg tablets and as a 10 mg/mL oral solution (Prod Info EPIVIR(R) oral tablets, solution, 2009).
    b) As part of a combination regimen, lamivudine is available in combination with zidovudine as 150 mg lamivudine/300 mg zidovudine tablets (Prod Info COMBIVIR(R) oral tablets, 2009).
    4) STAVUDINE
    a) Stavudine is available as 15, 20, 30 and 40 mg capsules and as a 1 mg/mL oral solution (Prod Info ZERIT(R) oral capsules, solution, 2009).
    5) TENOFOVIR
    a) As a single agent, tenofovir is available as 300 mg tablets containing tenofovir disoproxil fumarate, which is equivalent to 245 mg of tenofovir disoproxil (Prod Info VIREAD(R) oral tablets, 2010).
    b) As part of a combination regimen, tenofovir is available in combination with emtricitabine as 200 mg emtricitabine/300 mg tenofovir disoproxil fumarate tablets (Prod Info TRUVADA(R) oral tablets, 2009).
    6) ZALCITABINE is no longer available in the United States.
    7) DIDANOSINE and ZIDOVUDINE are covered in separate managements. Please refer to the individual managements for further information.
    B) USES
    1) AIDS antiretroviral agents are used in the treatment of HIV 1 and HIV 2 infectious processes in order to inhibit the replication of the virus. Evidence is lacking to show that these drugs affect latent or nonreplicating viruses (JEF Reynolds , 2000).

Life Support

    A) This overview assumes that basic life support measures have been instituted.

Clinical Effects

    0.2.1) SUMMARY OF EXPOSURE
    A) USES: The nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs) are primarily used in the treatment of HIV-1 and HIV-2 infection. This class includes: emtricitabine, lamivudine, stavudine, abacavir, and tenofovir; zidovudine and didanosine are in this class but are covered in separate managements, and zalcitabine is no longer manufactured. These agents are also used in the treatment of hepatitis B infection and human T-lymphocyte virus (HTLV) 1 and 2.
    B) PHARMACOLOGY: The NRTIs terminate HIV RNA to DNA transcription by acting as substrates for the HIV reverse transcriptase and terminating DNA elongation. These agents prevent cell infection, but have no effect on already infected cells.
    C) TOXICOLOGY: Toxicological effects are generally extensions of adverse effects.
    D) EPIDEMIOLOGY: Overdose is uncommon and severe sequelae from acute overdose are rare. Adverse effects and drug interactions, however, are common.
    E) WITH THERAPEUTIC USE
    1) COMMON: The most common adverse effects from all NRTIs are nausea, vomiting, headache, and malaise. Peripheral neuropathy and elevated transaminases have been reported with most NRTIs. Rash and hypersensitivity reactions are common and are usually self-limited when therapy is continued.
    2) Other adverse effects specific to each drug include: ABACAVIR: Hypersensitivity, nausea, vomiting, headache, and a possible increased risk of coronary artery disease. EMTRICITABINE: Rash, diarrhea, hypercholesterolemia, transaminitis, and mild rhabdomyolysis are common. Hepatic failure/steatosis, neutropenia, and lactic acidosis are rare. LAMIVUDINE: Headache and nausea are common. Pancreatitis is rare. STAVUDINE: Peripheral neuropathy in greater than 60% of patients receiving over 4 mg/kg daily. Lactic acidosis and transaminitis are common and do not require therapy or discontinuation unless severe. Dyslipidemia and insulin resistance have been associated with chronic stavudine and zidovudine. TENOFOVIR: Rash, headache, transaminitis, nausea, and vomiting are common. Hepatic failure/steatosis, renal failure, rhabdomyolysis, pancreatitis, and lactic acidosis are rare. ZALCITABINE: Peripheral neuropathy, stomatitis, pancreatitis, transaminitis, and rash.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: There are limited data regarding overdose of NRTIs. However, overdose appears to be largely well tolerated with very few reports of severe clinical effects despite over 2 decades of drug availability. Nausea or vomiting, neurologic symptoms (ie, ataxia, lethargy, nystagmus, peripheral neuropathy), signs of bone marrow toxicity (ie, anemia, leukopenia, thrombocytopenia), or an increase in liver enzymes have all been reported in NRTI overdose or chronic toxicity.
    2) SEVERE TOXICITY: Severe toxicity has been reported after therapeutic use but not after acute overdose, and may be manifested by pancreatitis, hepatic steatosis, acute renal failure (ie, tenofovir), neuropsychiatric abnormalities, or acidosis. Chronic therapeutic administration may lead to mitochondrial toxicity leading to lactic acidosis, with or without hepatic microsteatosis. Pancreatitis, neuropathy, and myopathy often accompany the syndrome. Severe neuropsychiatric effects (ie, seizures, mania) have been reported. Lactic acidosis has been reported in patients receiving both single and dual nucleoside analogue (NRTI) regimens for HIV infection. This is thought to cause multiorgan failure and most commonly occurs in persons on prolonged (more than 6 months) therapy.
    0.2.14) DERMATOLOGIC
    A) Dermatologic effects may include the development of skin rashes, eczema, impetigo, pruritus, excoriation, nail pigmentation (zidovudine and emtricitabine), sweating, and erythema. Abacavir has been noted to cause a life-threatening hypersensitivity reaction. Dermatologic effects are common. Stevens-Johnson syndrome is a rarely described complication.
    0.2.20) REPRODUCTIVE
    A) Most AIDS antiviral agents are classified as FDA pregnancy category C or B. Efavirenz/emtricitabine/tenofovir is classified as FDA pregnancy category D. Transient anemia and other blood abnormalities (neutropenia, thrombocytopenia, and lymphopenia), as well as hyperlactatemia, have been reported in zidovudine-exposed, but HIV-uninfected infants.
    B) An increased risk of severe or fatal lactic acidosis has been seen in pregnant women who take the combination of HIV drugs, stavudine and didanosine, with other antiretroviral agents. Pancreatitis is also a well-documented complication of stavudine and didanosine.
    0.2.21) CARCINOGENICITY
    A) DIDANOSINE
    1) At the time of this review, no data were available to assess the carcinogenic potential of this agent.
    B) ZIDOVUDINE
    1) Extremely large doses have been associated with vaginal neoplasms in mice and rats; the significance in humans is not known.

Laboratory Monitoring

    A) Monitor serum electrolytes and hepatic enzymes.
    B) Monitor serum lipase in patients with abdominal pain or severe acidosis.
    C) Lactic acid concentration and serum pH should be monitored in acidotic patients.
    D) Cardiac failure, likely due to acidosis, has been reported; therefore, cardiac monitoring is recommended in the setting of acidosis or chest pain.
    E) Sources of infection should be sought in patients with neutropenia or significant acidosis.

Treatment Overview

    0.4.2) ORAL/PARENTERAL EXPOSURE
    A) MANAGEMENT OF MILD TO MODERATE TOXICITY
    1) Supportive therapy remains the mainstay of care. Benzodiazepines or antipsychotics may be used for agitation or manic symptoms. Mild transaminitis can be monitored, discontinuation of therapy is not usually necessary. Therapy should be changed for persistently rising transaminases or evidence of hepatic synthetic dysfunction. Nausea and vomiting should be treated with antiemetics. Peripheral neuropathies are generally reversible with drug withdrawal and can be treated with pain management as needed. Asymptomatic elevation of lactic acid without systemic acidemia does not require discontinuation of the medication.
    B) MANAGEMENT OF SEVERE TOXICITY
    1) Supportive care is the mainstay of care. Aggressive fluid resuscitation should be initiated for severe lactic acidosis. Granulocyte colony stimulating factor may be considered for patients with agranulocytosis complicated by infection. Vasopressors may be necessary in cases with multi-organ failure. Withdrawal of the agent is imperative to improvement in severe adverse reactions. Riboflavin and L-carnitine may be useful in treating nucleoside reverse transcriptase inhibitor (NRTI)-associated lactic acidosis.
    C) DECONTAMINATION
    1) PREHOSPITAL: No prehospital decontamination is indicated. Prehospital care should focus on assessment of vital signs and general supportive care.
    2) HOSPITAL: Activated charcoal may be considered for patients that present early after overdose if they are awake, alert, and willing to drink the charcoal. Gastric lavage has no role in the management of NRTI overdose.
    D) AIRWAY MANAGEMENT
    1) Respiratory depression is not expected with overdose of NRTIs. However, coingestants must be considered and airway protection should be employed as needed for airway protection.
    E) ANTIDOTE
    1) There is no specific antidote for NRTI toxicity.
    F) ACIDOSIS
    1) Treat severe metabolic acidosis (pH less than 7.1) with sodium bicarbonate 1 to 2 mEq/kg. Anecdotal evidence suggests that riboflavin and L-carnitine may be useful in reversing NRTI-associated lactic acidosis. Riboflavin has been used at a dose of 50 mg/day orally or intravenously. L-carnitine has been used at a dose of 50 mg/kg/day as a 2-hour infusion divided in 3 doses for patients not receiving dialysis, or a continuous infusion of 100 mg/kg/day in patients receiving dialysis.
    G) ENHANCED ELIMINATION
    1) Hemodialysis and whole bowel irrigation have no role in the management of NRTI overdose.
    H) PATIENT DISPOSITION
    1) HOME CRITERIA: Suicidal patients and those with symptoms should be referred to a healthcare facility. Asymptomatic patients with inadvertent ingestion of NRTIs can be observed at home.
    2) OBSERVATION CRITERIA: Asymptomatic or mildly symptomatic patients should be observed for 4 to 6 hours, primarily monitoring signs of coingestant toxicity.
    3) ADMISSION CRITERIA: Patients with severe toxicity should be admitted. Patients with severe lactic acidosis, hepatic failure, or renal failure should be admitted to an intensive care setting.
    4) CONSULT CRITERIA: Infectious disease should be consulted if a change to anti-retroviral therapy is indicated. Consult a medical toxicologist for patients with severe toxicity or in whom the diagnosis is not clear.
    I) PITFALLS
    1) Failure to consider toxicity of co-medications due to drug-drug interactions. Failure to remove the offending agent in patients with severe adverse drug reactions.
    J) PHARMACOKINETICS
    1) Intracellular elimination half-lives range from 2 to 24 hours. NRTIs are transported into cells and phosphorylated into an active form for incorporation into the viral reverse transcriptase. The drugs can be dephosphorylated or directly catabolized intracellularly. Formation of intracellular active metabolites has not been fully characterized and therefore pharmacokinetic profiles cannot be accurately predicted.
    2) ABACAVIR: Bioavailability 83%, protein binding 50%, volume of distribution 0.86 L/kg, extensive hepatic metabolism with renal elimination of metabolites, half-life 1 to 1.5 hours.
    3) EMTRICITABINE: Bioavailability 93%, protein binding less than 4%, little hepatic metabolism (13%), renal excretion 86%, half-life 10 hours.
    4) LAMIVUDINE: Bioavailability 82% to 87%, moderate protein binding (less than 36%), volume of distribution 0.9 to 1.7 L/kg, 70% renal elimination of unchanged drug, half-life 2 to 7 hours.
    5) STAVUDINE: Bioavailability 86%, negligible protein binding, volume of distribution 46 L, limited hepatic metabolism, renal elimination approximately 40%, half-life 1.6 hours.
    6) TENOFOVIR: Bioavailability 25%, protein binding 7%, volume of distribution 1.2 to 1.3 L/kg, 32% excreted unchanged in urine, half-life 17 hours.
    K) TOXICOKINETICS
    1) No data are available regarding toxicokinetics.
    L) DIFFERENTIAL DIAGNOSIS
    1) Other etiologies of hepatic failure (ie, acetaminophen, iron, carbon tetrachloride, etc.) should be considered. Medical etiologies (ie, portal vein thrombosis, viral hepatitis, hepatic abscess, or Budd-Chiari malformation) should be ruled out in cases of hepatitis. Infection must be ruled out in cases predominated by lactic acidosis and organ dysfunction.

Range Of Toxicity

    A) TOXICITY: A full month supply of many of these agents has been ingested in overdose without clinical effects, though toxicity can occur at therapeutic doses with nucleoside reverse transcriptase inhibitors (NRTIs). LAMIVUDINE: No clinical signs or symptoms developed in an adult ingesting 6 grams of lamivudine. STAVUDINE: No acute toxicity was reported in patients treated with 12 to 24 times the recommended daily dosage. ZALCITABINE: Pediatric: Overdoses of 1.5 mg/kg have been reported; no sequelae developed.
    B) THERAPEUTIC DOSE: ABACAVIR: ADULT: 300 mg orally twice daily or 600 mg once daily. PEDIATRIC: 8 mg/kg orally twice daily. EMTRICITABINE: ADULT: 200 mg/day capsule, 240 mg/day oral solution. PEDIATRIC: 0 to 3 months of age: 3 mg/kg orally once daily; 3 months to 17 years of age: 6 mg/kg once daily oral solution, up to a maximum of 240 mg; children weighing more than 33 kg and can swallow whole capsule: 200 mg once daily. LAMIVUDINE: ADULT: 150 mg orally twice a day or 300 mg once daily. PEDIATRIC: 0 to 28-days-old: 2 mg/kg orally twice daily; 28 days or older: 4 mg/kg orally twice daily, maximum 150 mg twice daily. STAVUDINE: ADULT: less than 60 kg: 30 mg orally every 12 hours; 60 kg or more: 40 mg orally every 12 hours. PEDIATRIC: 0 to 13-days-old: 0.5 mg/kg/dose orally every 12 hours; 14-days-old and less than 30 kg: 1 mg/kg/dose orally every 12 hours ; 30 kg to less than 60 kg: 30 mg orally every 12 hours; 60 kg or more: 40 mg orally every 12 hours. TENOFOVIR: ADULT: 300 mg orally once daily. PEDIATRIC: at least 12-year-old and 35 kg or more: 300 mg orally once daily.

Summary Of Exposure

    A) USES: The nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs) are primarily used in the treatment of HIV-1 and HIV-2 infection. This class includes: emtricitabine, lamivudine, stavudine, abacavir, and tenofovir; zidovudine and didanosine are in this class but are covered in separate managements, and zalcitabine is no longer manufactured. These agents are also used in the treatment of hepatitis B infection and human T-lymphocyte virus (HTLV) 1 and 2.
    B) PHARMACOLOGY: The NRTIs terminate HIV RNA to DNA transcription by acting as substrates for the HIV reverse transcriptase and terminating DNA elongation. These agents prevent cell infection, but have no effect on already infected cells.
    C) TOXICOLOGY: Toxicological effects are generally extensions of adverse effects.
    D) EPIDEMIOLOGY: Overdose is uncommon and severe sequelae from acute overdose are rare. Adverse effects and drug interactions, however, are common.
    E) WITH THERAPEUTIC USE
    1) COMMON: The most common adverse effects from all NRTIs are nausea, vomiting, headache, and malaise. Peripheral neuropathy and elevated transaminases have been reported with most NRTIs. Rash and hypersensitivity reactions are common and are usually self-limited when therapy is continued.
    2) Other adverse effects specific to each drug include: ABACAVIR: Hypersensitivity, nausea, vomiting, headache, and a possible increased risk of coronary artery disease. EMTRICITABINE: Rash, diarrhea, hypercholesterolemia, transaminitis, and mild rhabdomyolysis are common. Hepatic failure/steatosis, neutropenia, and lactic acidosis are rare. LAMIVUDINE: Headache and nausea are common. Pancreatitis is rare. STAVUDINE: Peripheral neuropathy in greater than 60% of patients receiving over 4 mg/kg daily. Lactic acidosis and transaminitis are common and do not require therapy or discontinuation unless severe. Dyslipidemia and insulin resistance have been associated with chronic stavudine and zidovudine. TENOFOVIR: Rash, headache, transaminitis, nausea, and vomiting are common. Hepatic failure/steatosis, renal failure, rhabdomyolysis, pancreatitis, and lactic acidosis are rare. ZALCITABINE: Peripheral neuropathy, stomatitis, pancreatitis, transaminitis, and rash.
    F) WITH POISONING/EXPOSURE
    1) MILD TO MODERATE TOXICITY: There are limited data regarding overdose of NRTIs. However, overdose appears to be largely well tolerated with very few reports of severe clinical effects despite over 2 decades of drug availability. Nausea or vomiting, neurologic symptoms (ie, ataxia, lethargy, nystagmus, peripheral neuropathy), signs of bone marrow toxicity (ie, anemia, leukopenia, thrombocytopenia), or an increase in liver enzymes have all been reported in NRTI overdose or chronic toxicity.
    2) SEVERE TOXICITY: Severe toxicity has been reported after therapeutic use but not after acute overdose, and may be manifested by pancreatitis, hepatic steatosis, acute renal failure (ie, tenofovir), neuropsychiatric abnormalities, or acidosis. Chronic therapeutic administration may lead to mitochondrial toxicity leading to lactic acidosis, with or without hepatic microsteatosis. Pancreatitis, neuropathy, and myopathy often accompany the syndrome. Severe neuropsychiatric effects (ie, seizures, mania) have been reported. Lactic acidosis has been reported in patients receiving both single and dual nucleoside analogue (NRTI) regimens for HIV infection. This is thought to cause multiorgan failure and most commonly occurs in persons on prolonged (more than 6 months) therapy.

Vital Signs

    3.3.3) TEMPERATURE
    A) WITH THERAPEUTIC USE
    1) FEVER: Chills and fever have been reported in 10% of patients receiving lamivudine plus zidovudine, and in 12% receiving zidovudine alone (Prod Info EPIVIR(R) oral tablets, solution, 2008). Stavudine treated patients also experienced chills and fever with use (Prod Info ZERIT(R) oral capsules, oral solution, 2006). Fever was reported in pediatric patients with a frequency of 19% following abacavir dosing (Prod Info Ziagen(TM), abacavir, 1998).

Heent

    3.4.4) EARS
    A) WITH THERAPEUTIC USE
    1) HEARING LOSS
    a) ZALCITABINE: Ototoxicity with hearing loss and tinnitus was reported in an AIDS patient taking alternating therapy with zalcitabine and zidovudine. On rechallenge with zalcitabine, hearing disturbance again recurred. The ototoxicity resolved on discontinuation of zalcitabine (Powderly et al, 1990).
    3.4.5) NOSE
    A) WITH THERAPEUTIC USE
    1) NASAL SYMPTOMS
    a) ZIDOVUDINE/LAMIVUDINE: Nasal signs and symptoms have been reported in 20% of adult patients receiving lamivudine plus zidovudine therapy; 11% of patients receiving zidovudine alone experienced these adverse effects (Prod Info EPIVIR(R) oral tablets, solution, 2008).
    3.4.6) THROAT
    A) WITH THERAPEUTIC USE
    1) ORAL ULCERATIONS -
    a) ZALCITABINE therapy has been associated with rare incidences of mouth ulcers, more than with other NRTIs (Carr & Cooper, 2000).

Cardiovascular

    3.5.2) CLINICAL EFFECTS
    A) HEART FAILURE
    1) WITH THERAPEUTIC USE
    a) Nucleoside analogs have been reported to exacerbate cardiomyopathy and congestive heart failure (Prod Info HIVID(R) oral tablets, 2002).
    B) CONDUCTION DISORDER OF THE HEART
    1) WITH THERAPEUTIC USE
    a) ZALCITABINE: Fewer than 3% of patients in zalcitabine clinical trials experienced the following: dysrhythmia, atrial fibrillation, hypertension, palpitations, tachycardia, and ventricular ectopy (Prod Info HIVID(R) oral tablets, 2002).
    3.5.3) ANIMAL EFFECTS
    A) ANIMAL STUDIES
    1) CARDIOMYOPATHY
    a) DIDEOXYADENOSINE (DDA): Both Fluoro-dideoxyadenosine (F-ddA) and dideoxyadenosine (ddA) (investigational drugs) produced dose-dependent cardiac lesions in rat studies. The deaminated catabolites of both agents were essentially non-toxic. Toxicity appeared related to Cmax rather than total exposure, with doses of 250 mg/kg producing significant cardiomyopathy. Toxicity was greater following treatment with F-ddA as opposed to ddA probably because F-ddA is deaminated 20 times more slowly than ddA (Donzanti et al, 1995).
    b) FddA and FddL: Myocardial degeneration and necrosis was evident at necropsy in rats administered FddA or FddL 500 to 1000 mg/kg/day either orally or intravenously (Comereski et al, 1993).

Respiratory

    3.6.2) CLINICAL EFFECTS
    A) DISORDER OF RESPIRATORY SYSTEM
    1) WITH THERAPEUTIC USE
    a) ZIDOVUDINE/LAMIVUDINE: Cough has been reported in 18% of adult patients receiving lamivudine plus zidovudine as compared to 13% in patients receiving zidovudine alone (Prod Info EPIVIR(R) oral tablets, solution, 2008).
    b) ABACAVIR: Respiratory distress may be one of the manifestations of hypersensitivity reactions associated with this agent. The potential of hypersensitivity reaction should be evaluated prior to the reinstitution of therapy in patients (Carr & Cooper, 2000).

Neurologic

    3.7.2) CLINICAL EFFECTS
    A) SEIZURE
    1) ZALCITABINE: Seizures have occurred in less than 3% of patients in zalcitabine clinical trials (Prod Info HIVID(R) oral tablets, 2002).
    2) LAMIVUDINE: Drowsiness and seizures are a rare occurrence but have been reported following therapy with lamivudine (van Leeuwen et al, 1995; van Leeuwen et al, 1992). A causal relationship to therapy is uncertain.
    B) TOXIC ENCEPHALOPATHY
    1) ZALCITABINE: Dizziness, confusion, amnesia, depression, and paralysis have occurred in less than 3% of patients in zalcitabine clinical trials (Prod Info HIVID(R) oral tablets, 2002).
    C) NEUROPATHY
    1) WITH THERAPEUTIC USE
    a) Painful distal symmetrical peripheral neuropathy is the major dose-limiting toxicity of the nucleoside analogs (most often seen with didanosine, stavudine, and zalcitabine). It often progresses to severe pain requiring narcotics. It generally is abrupt in onset and rapidly progresses. Histologically, axonal degeneration is evident (Anon, 1989; Cooley et al, 1990; Lambert et al, 1990) Yarchoan et al, 1990; (Merigan et al, 1989; Merigan & Skowron, 1990; Skowron, 1995; Murray et al, 1995; Petersen et al, 1995; Simpson & Tagliati, 1995; Prod Info Zerit(R),, 2002; Moyle & Sadler, 1998; Moore et al, 2000; Carr & Cooper, 2000; Falco et al, 2002).
    1) INCIDENCE: Distal pain, numbness, paresthesias, and reduced reflexes/power occurs in 10% to 30% of NRTI treated patients, with the following decreasing order of incidence: zalcitabine = stavudine > didanosine > lamivudine (Carr & Cooper, 2000).
    2) The painful neuropathic syndrome consists of tingling, burning, or aching in the lower extremities. A painful sensorimotor peripheral neuropathy with a stocking glove distribution is described, and occurs particularly at night but gradually progresses to interfere with walking, sleep, and routine daily activities. There are no associated neurologic deficits except for occasional diminished vibratory sensation and decreased ankle reflexes (Lambert et al, 1990; Prod Info Hivid(R), zalcitabine, 1996). The onset of neuropathy usually ranges from 55 to 201 days after initiation of therapy and is usually reversible after discontinuation of the drug (Cooley et al, 1990; Lambert et al, 1990).
    3) Verma et al (1999) reported a fulminant axonal neuropathy with lactic acidosis and acute hyperglycemia in an HIV-infected patient treated with NRTI therapy. Motor deficit progressed to total paralysis of all limbs. Following discontinuation of NRTI therapy, lactic acidosis slowly resolved over 2 weeks, but neurologic deficit was irreversible, and the patient died following multiple organ failure (Verma et al, 1999).
    4) OTOTOXICITY, with tinnitus and hearing loss, has been associated with therapeutic use of NRTIs, although this is uncommon. Mitochondrial DNA toxicity as well as mitochondrial DNA mutations associated with aging and HIV-1 infection may all contribute to ototoxicity (Simdon et al, 2001).
    5) OPTIC NEUROPATHY: Late onset Leber's hereditary optic neuropathy in a 57-year-old man in the setting of nucleoside analogue toxicity has been reported. A total exposure to nucleoside analogues of 9 years was documented. The authors speculated that antiretroviral therapy played a role in producing disease expression at a late stage (Luzhansky et al, 2001).
    b) STAVUDINE: 19% to 24% of stavudine treated patients have required dosage reductions due to painful peripheral neuropathies (Petersen et al, 1995). Stavudine-induced peripheral neuropathy is dose-dependent (Moyle & Sadler, 1998).
    c) ZALCITABINE: All patients in a high-dose zalcitabine clinical trial experienced painful, predominantly sensorimotor peripheral neuropathy. Mean onset was 7.7 weeks (Berger et al, 1993). After stopping zalcitabine, some patients experience a period of symptom intensification, referred to as "coasting", lasting for several weeks to months (Moyle & Sadler, 1998). Severe peripheral neuropathy necessitating discontinuation of therapy occurs in about 10% of patients. Carey (2000) reports that the incidence of peripheral neuropathy is infrequent when zalcitabine is used in combination with other antiretrovirals (Carey, 2000).
    1) Acoustic neuropathy, with bilateral deafness and tinnitus, was reported to accompany peripheral neuropathy in a 46-year-old man taking therapeutic doses of zalcitabine. The neuropathy resolved within 4 months of stopping the drug (Martinez & French, 1993).
    d) Risk factors predisposing patients to peripheral neuropathies include: history of peripheral nervous system disease, heavy alcohol consumption, or a low serum cobalamin level (Fichtenbaum et al, 1995).
    e) LAMIVUDINE: Lamivudine is reported to be the least neurotoxic of this class of drugs. However, it has been reported to potentiate an underlying peripheral neuropathy in a 33-year-old man with AIDS (Cupler & Dalakas, 1995).
    D) HEADACHE
    1) WITH THERAPEUTIC USE
    a) ABACAVIR: Headache has been reported in pediatric patients with a frequency of 16% (Prod Info Ziagen(TM), abacavir, 1998).
    b) ZIDOVUDINE/LAMIVUDINE: Headache is a common effect, occurring in 35% of patients on combination lamivudine and zidovudine, and 27% on zidovudine monotherapy (Prod Info EPIVIR(R) oral tablets, solution, 2008).

Gastrointestinal

    3.8.2) CLINICAL EFFECTS
    A) PANCREATITIS
    1) WITH THERAPEUTIC USE
    a) INCIDENCE: Pancreatitis, with abdominal pain and elevated amylase has been reported to occur at a rate of <1% to 6% of patients in the following decreasing order of drug use: didanosine > lamivudine/zalcitabine (Carr & Cooper, 2000). Alcohol and pentamidine may aggravate pancreatitis resulting from NRTIs. Pancreatitis is a well-documented adverse effect of stavudine and didanosine ((Anon, 2001)).
    b) ZALCITABINE: Fatal pancreatitis has been reported following zalcitabine therapy, however, this is not a common effect (Prod Info HIVID(R) oral tablets, 2002).
    c) ZIDOVUDINE/ZALCITABINE: Fatal hemorrhagic pancreatitis was reported in a 45-year-old woman treated concurrently with zidovudine and zalcitabine (Aponte-Cipriani et al, 1993).
    d) It has been suggested that the development of hypertriglyceridemia may serve as a marker for patients at risk of developing pancreatitis (Tal & Dall, 1993).
    B) DIARRHEA
    1) WITH THERAPEUTIC USE
    a) LAMIVUDINE: Diarrhea, which does not appear to be dose dependent, may be severe enough to necessitate discontinuance of medication, and may be accompanied by nausea and vomiting in patients receiving lamivudine (Pluda et al, 1995).
    b) STAVUDINE: Kline et al (1995) reported a 68% incidence of diarrhea, which was not dose-related, in pediatric patients (Kline et al, 1995).
    C) NAUSEA AND VOMITING
    1) WITH THERAPEUTIC USE
    a) ABACAVIR causes nausea, vomiting, diarrhea and loss of appetite in adults and children with a frequency of greater than 10% (Prod Info Ziagen(TM), abacavir, 1998). In clinical trials of abacavir alone, gastrointestinal disturbance was the primary adverse effect in adults (Foster & Faulds, 1998).
    b) ZIDOVUDINE/LAMIVUDINE: Nausea has occurred in 33% of patients treated with lamivudine plus zidovudine and 29% of zidovudine mono therapy patients in 1 study (Prod Info EPIVIR(R) oral tablets, solution, 2008).
    2) WITH POISONING/EXPOSURE
    a) LAMIVUDINE/EFAVIRENZ: A 17-year-old developed lower abdominal pain and then began vomiting 5 hours after ingesting lamivudine 3.9 g and efavirenz 15.6 g (Boscacci et al, 2006).
    D) ULCER OF ESOPHAGUS
    1) WITH THERAPEUTIC USE
    a) ZALCITABINE: Therapy with zalcitabine has been associated with rare esophageal ulceration(Prod Info HIVID(R) oral tablets, 2002; Indorf & Pegram, 1992). Local pill-induced injury has been suggested as the etiology of zalcitabine-induced esophageal ulceration.
    b) LAMIVUDINE: Oral ulcerations/lesions have been reported in patients receiving lamivudine (van Leeuwen et al, 1995; Pluda et al, 1995).

Hepatic

    3.9.2) CLINICAL EFFECTS
    A) LIVER ENZYMES ABNORMAL
    1) Asymptomatic elevation in liver aminotransferase serum concentrations (with normal bilirubin) are more common than serious liver toxicity with symptoms. This occurs in 5% to 15% of all patients treated with NRTIs (except abacavir) (Carr & Cooper, 2000).
    2) ZIDOVUDINE/LAMIVUDINE: Increased liver enzymes (5 times the upper limit) have been reported equally in patients receiving lamivudine plus zidovudine as compared to zidovudine alone. This is a small percent of patients, but may increase in overdose cases (Prod Info EPIVIR(R) oral tablets, solution, 2008).
    3) ZIDOVUDINE/ZALCITABINE: Two nurses, receiving zidovudine and zalcitabine for prophylaxis following needlestick injuries, developed elevated liver enzymes after 3 weeks of therapy associated with pruritic rashes and lymphocytopenia (Henry et al, 1996).
    4) STAVUDINE: Modest elevations in serum transaminases were commonly observed in controlled clinical trials of stavudine (Prod Info ZERIT(R) oral capsules, oral solution, 2006; Browne et al, 1993; Sandstrom & Oberg, 1993; Wallace et al, 1996).
    B) HEPATIC FAILURE
    1) WITH THERAPEUTIC USE
    a) A case series of 6 patients with acute liver failure associated with nucleoside analogue HIV-1 therapy has been reported. Five of the 6 died. Development of acute liver failure was sudden and did not appear to be related to duration or type of antiretroviral therapy, and was not predicted by monitoring of liver function. Median duration of therapy was 12.5 months and median time to onset of liver failure following introduction of new therapy was 8 weeks. A predominantly hepatocellular pattern was noted. Liver biopsy revealed mitochondrial toxicity in 1 case and confluent hepatocellular necrosis, inflammation and cholestasis in the other cases (Clark et al, 2002).
    b) Fatal portal hypertension, liver failure, and mitochondrial dysfunction have been reported in a patient who had recovered from HIV-1 nucleoside analogue-induced hepatitis and lactic acidemia more than 18 months earlier. The authors suggest a delayed progression to non-cirrhotic portal hypertension and chronic liver failure with persistent mitochondrial dysfunction following a recovery from nucleoside-analogue-induced acute hepatitis and lactic acidosis (Carr et al, 2001).
    c) ZALCITABINE: Rare cases of liver failure, lactic acidosis, and hepatomegaly with steatosis have been reported with zalcitabine therapy (Prod Info HIVID(R) oral tablets, 2002).
    C) TOXIC HEPATITIS
    1) WITH THERAPEUTIC USE
    a) LAMIVUDINE has been reported to cause a reactivation of Hepatitis B in a 29-year-old man with chronic HBV infection (Honkoop et al, 1995).
    D) STEATOSIS OF LIVER
    1) WITH THERAPEUTIC USE
    a) Typically, liver disease due to NRTI therapy is associated with lactic acidosis and massive liver steatosis and failure. The incidence of liver failure and lactic acidosis in a retrospective study of a cohort of HIV-infected patients treated with NRTI was reported to be 1.3 per 1000 person-years of follow-up (Bleeker-Rovers et al, 2000). The manufacturers of lamivudine and stavudine have issued warnings concerning lactic acidosis and severe hepatomegaly with steatosis, including fatal cases, with the therapeutic use of these drugs (Prod Info EPIVIR-HBV(R) oral tablets, oral solution, 2004; Prod Info ZERIT(R) oral capsules, oral solution, 2006).
    1) Hepatic steatosis with lactic acidosis has occurred following therapeutic doses of the nucleoside reverse transcriptase inhibitors (NRTI) (Fouty et al, 1998; Mokrzycki et al, 2000; Miller et al, 2000; Lonergan et al, 2000; Megarbane et al, 2001). It has been proposed that hepatic steatosis and lactic acidosis results from impaired mitochondrial DNA replication due to NRTI-mediated inhibition of DNA polymerase gamma and possible dietary deficiencies. Fatal lactic acidosis and severe hepatomegaly with steatosis have been reported following stavudine, didanosine and abacavir therapy (Prod Info ZERIT(R) oral capsules, oral solution, 2006; Mokrzycki et al, 2000; Brivet et al, 2000; Carr et al, 2000; Bleeker-Rovers et al, 2000).
    b) The syndrome of lactic acidosis and hepatic steatosis, a complication of nucleoside reverse-transcriptase inhibitors, has been associated with riboflavin deficiency. Luzzati et al (1999) reported a patient with severe lactic acidosis and hepatic steatosis who responded to riboflavin treatment (50 mg/day) with rapid recovery (within 4 days) (Luzzati et al, 1999).

Genitourinary

    3.10.2) CLINICAL EFFECTS
    A) RENAL TUBULAR DISORDER
    1) WITH THERAPEUTIC USE
    a) ADEFOVIR: At doses of 120 mg/day for greater than 6 months, 40% of patients in clinical trials developed signs of incipient renal tubular damage, a Fanconi-like syndrome (Gilden, 1998).
    b) TENOFOVIR
    1) Seven HIV-infected patients developed renal tubular injury from 5 weeks to 16 months after receiving polytherapy that included tenofovir. Some of the renal disorders were consistent with Fanconi syndrome. Laboratory abnormalities returned to baseline or were significantly improved within 4 months after discontinuing tenofovir (Peyriere et al, 2004).
    2) CASE REPORT: A 41-year-old patient with HIV and hepatitis B presented with proteinuria and an elevated serum creatinine concentration (from 1 to 1.5 mg/dL). The patient's medication regimen, for the past 4 years, consisted of emtricitabine, tenofovir, atazanavir, and ritonavir. In addition to the patient's elevated serum creatinine concentration, laboratory data revealed elevated serum albumin and serum protein concentrations, urinalysis was positive for protein and glucose, and urine sediment microscopy demonstrated cigar-shaped renal epithelial cells, all of which is consistent with proximal tubular injury and Fanconi syndrome. Renal biopsy of the patient also showed acute tubular necrosis, primarily involving the proximal tubules. Further investigation of the patient's history revealed that the patient first developed proximal tubular dysfunction, as evidenced by the presence of orthoglycemic glycosuria , approximately 2 years following initiation of tenofovir therapy, suggesting that the patient's renal dysfunction is secondary to tenofovir toxicity (Agarwala et al, 2010).
    3) CASE SERIES: A series of 22 patients with HIV were identified with tenofovir-associated renal toxicity. Six of the 22 patients were on first-line antiretroviral therapy (ART). Twenty patients were taking tenofovir in combination with ritonavir and a protease inhibitor, and 5 patients were taking didanosine concomitantly. According to the available data from 19 of the 22 patients, all had normal serum creatinine concentration (median 84 mcmol/L; range 68 to 111 mcmol/L), with an increase in the serum creatinine concentration after initiating therapy with tenofovir (median rise 88 mcmol/L; range 5 to 659 mcmol/L). After discontinuation of tenofovir therapy, the serum creatinine concentration decreased in 95.5% of the patients, with the serum creatinine concentration returning to normal in 59% of the patients. All 22 patients also developed proteinuria at presentation, which significantly decreased following discontinuation of tenofovir. Nineteen of the 22 patients developed hypophosphatemia following initiation of tenofovir therapy. After discontinuing therapy, phosphate concentrations returned to normal in 18 patients. Thirteen of 14 patients also showed a reduction in the capacity of the proximal tubule to reabsorb urinary phosphate, indicating that the hypophosphatemia may be secondary to proximal tubule dysfunction. In addition to the tubular proteinuria and the hypophosphatemia, glycosuria, in the absence of diabetes, was present in 8 of the 9 patients tested, confirming a diagnosis of Fanconi Syndrome secondary to tenofovir toxicity (Woodward et al, 2009).

Acid-Base

    3.11.2) CLINICAL EFFECTS
    A) LACTIC ACIDOSIS
    1) WITH THERAPEUTIC USE
    a) Nucleoside reverse transcriptase inhibitors have been associated with an abrupt onset of lactic acidosis (type "B") and hepatic steatosis or an insidious onset in patients with AIDS. Lactic acidosis is uncommon, but when it occurs, it is not readily reversible after discontinuation of the drugs and with normal therapy and mortality rate is high. This syndrome may result in death from progressive lactic acidosis, hypotension, and multiorgan failure (Fouty et al, 1998; Mokrzycki et al, 2000; Brivet et al, 2000; Carr et al, 2000; Miller et al, 2000; Lonergan et al, 2000; ter Hofstede et al, 2000; Bleeker-Rovers et al, 2000; Coghlan et al, 2001; Carr et al, 2001; Megarbane et al, 2001; Falco et al, 2002).
    1) Initially, nausea, vomiting, and abdominal pain occur. Fatigue and bodyweight loss may predominate in insidious cases. A tender, enlarged liver may be palpable. This may be followed by dyspnea, tachypnea, and hyperventilation, liver and/or renal failure, clotting abnormalities, seizures, cardiac dysrhythmias, and death (Moyle, 2000).
    b) INCIDENCE: Some degree of lactic acidosis has been reported to occur in approximately 15% of patients receiving NRTI therapy. Differences in the relative incidence of lactic acidosis between different agents or different combinations have not been established (Carr & Cooper, 2000; Moyle, 2000). The presence of asymptomatic hyperlactatemia was studied in 83 HIV-infected, nucleoside analogue-treated patients. Two thirds had 2 or more risk factors for hyperlactatemia and 11% had more than 4 risk factors. Two patients had a lactate level more than 1.5 times the upper limit of normal. There were no reports of symptomatic hyperlactatemia or lactic acidosis during the study (Wohl et al, 2004).
    c) MORTALITY: Lactate serum levels of >10 mM appear to be associated with a higher mortality (Falco et al, 2002a). In a retrospective study of 39 patients (34 from previously published literature) with severe lactic acidosis related to nucleoside analogue therapy, an initial lactate level of greater than 9 mmol/L was useful in predicting mortality. In patients with initial lactate levels of greater than 9 mmol/L 3 of 20 survived, while 17 of the 19 patients with initial lactate of less than 9 mmol/L survived(Claessens et al, 2003).
    d) Lactic acidosis and hepatic steatosis were reported as a possible syndrome related to riboflavin deficiency in HIV-infected patients taking nucleoside reverse-transcriptases inhibitors. A 35-year-old pregnant patient with this syndrome was non-responsive to sodium bicarbonate. When therapy was changed to riboflavin (50 mg/day orally), she recovered rapidly (4 days) with a fall in blood lactate to normal values (Luzzati et al, 1999).
    e) PREGNANCY: The FDA has issued a warning concerning an increased risk of severe or fatal lactic acidosis in pregnant women who take the combination of HIV drugs, stavudine and didanosine, with other antiretroviral agents. Pancreatitis is also a well-documented complication of stavudine and didanosine (Prod Info ZERIT(R) oral capsules, oral solution, 2006).
    1) Three cases of fatal lactic acidosis, with and without pancreatitis, were reported in pregnant women taking stavudine and didanosine in combination with other antiretroviral drugs. Several nonfatal cases in pregnant women (with and without pancreatitis) were also reported. Although it has been suggested that women may be at an increased risk of developing lactic acidosis and liver toxicity, it is unclear if pregnancy potentiates these known adverse events ((Anon, 2001)).
    f) CASE REPORT: A 46-year-old woman, taking stavudine, lamivudine, indinavir and amitriptyline for 4 months, developed lactic acidosis (6.4 mmol/L), decreased BUN (1.8 mmol/L) and significant hepatic steatosis on abdominal CT. A riboflavin deficiency was suspected and confirmed. Therapy with 50 mg riboflavin resolved the lactic acidosis and raised her BUN into a normal range (Fouty et al, 1998).
    g) CASE REPORT: An HIV-infected patient taking NRTI was reported with fulminant axonal neuropathy with lactic acidosis and acute hyperglycemia. Liver function was normal. Serum lactate levels declined over a 2 week period following discontinuation of NRTI, but the neuropathy was irreversible, and the patient died due to multiple organ failure (Verma et al, 1999).
    h) CASE SERIES: Six cases of fatal lactic acidosis were reported in patients taking stavudine (6 patients), lamivudine (5 patients), and didanosine (1 patient). All patients had manifestations consistent with mitochondrial toxicity including lipodystrophy, myositis, fatty hepatitis, pancreatitis, and peripheral neuropathy. Although antiretroviral therapy was discontinued and aggressive treatment with fluid resuscitation, riboflavin, and bicarbonate was given, all patients died of persistent circulatory collapse (Sheng et al, 2004).

Hematologic

    3.13.2) CLINICAL EFFECTS
    A) THROMBOCYTOPENIC DISORDER
    1) WITH THERAPEUTIC USE
    a) ZALCITABINE has been associated with thrombocytopenia in only 1% to 3% of patients in clinical trials and appears to be dose related (Prod Info HIVID(R) oral tablets, 2002).
    b) LAMIVUDINE: Thrombocytopenia occurred in 23% of pediatric patients with abnormal baseline levels receiving lamivudine alone (Prod Info Epivir(R), lamivudine, 2001).
    c) STAVUDINE: No significant thrombocytopenia has been reported in clinical studies (Browne et al, 1993; Sandstrom & Oberg, 1993). The manufacturer reports an incidence of 3% to 5% thrombocytopenia (platelets < 50,000 cells/mm(3)) in a Phase 3 trial (Prod Info Zerit(R),, 2002).
    B) LEUKOPENIA
    1) WITH THERAPEUTIC USE
    a) ZALCITABINE: Neutropenia has been reported as a dose dependent adverse effect in 17% of patients in clinical trials of zalcitabine, and leukopenia in 13% (Prod Info HIVID(R) oral tablets, 2002).
    b) LAMIVUDINE: Dose related neutropenia has been reported. Doses of 20 mg/kg resulted in a trend toward progressively decreasing neutrophil counts in 1 study. This did not occur at lower doses. Neutropenia of less than 1000/mm(3) was reported in 2 of the 15 patients on the higher dose. Neutropenia occurred on rechallenge (Pluda et al, 1995).
    1) In pediatric patients receiving lamivudine alone, neutropenia occurred in 22% of patients with normal baselines (Prod Info Epivir(R), lamivudine, 2001).
    c) STAVUDINE: Bone marrow toxicity is not substantial following therapeutic stavudine in available studies (Browne et al, 1993; Dunkle et al, 1990; Sandstrom & Oberg, 1993; Skowron, 1995).
    C) MACROCYTIC ANEMIA
    1) WITH THERAPEUTIC USE
    a) LAMIVUDINE: In pediatric patients receiving lamivudine alone, anemia occurred in 24% who were anemic prior to initiation of therapy. Anemia occurred in 2% with prior normal baselines (Prod Info Epivir(R), lamivudine, 2001). A retrospective study, conducted by Khawcharoenporn et al (2007), found that lamivudine was strongly associated with macrocytosis (OR=24.6 [2.9 to 3223], p=0.001) (Khawcharoenporn et al, 2007).

Dermatologic

    3.14.1) SUMMARY
    A) Dermatologic effects may include the development of skin rashes, eczema, impetigo, pruritus, excoriation, nail pigmentation (zidovudine and emtricitabine), sweating, and erythema. Abacavir has been noted to cause a life-threatening hypersensitivity reaction. Dermatologic effects are common. Stevens-Johnson syndrome is a rarely described complication.
    3.14.2) CLINICAL EFFECTS
    A) ERUPTION
    1) WITH THERAPEUTIC USE
    a) DESCRIPTION: Hypersensitivity may manifest as an erythematous, maculopapular, pruritic, and confluent rash with or without fever. The rash usually begins after 1 to 3 weeks of therapy and is most prominent on the body and arms. Chills and fever, myalgias, and arthralgias are often prominent and may precede the rash, particularly with abacavir. Rash or fever occurring greater than 8 weeks after onset of therapy is usually due to another agent (Carr & Cooper, 2000).
    b) ABACAVIR: Abacavir has been reported to induce life-threatening hypersensitivity syndromes in 5% to 8% of patients, manifested by cutaneous, gastrointestinal and/or respiratory symptoms. Risk factors include female gender, non-African ethnicity, and certain genetic HLA haplotypes. With drug re-challenge, multi-organ failure and death have occurred (Stekler et al, 2006; Carr & Cooper, 2000).
    c) ZALCITABINE: A transient symptom complex of cutaneous eruptions (maculovesicular in nature), fever, malaise, and aphthous ulcers have been a relatively frequent complication of zalcitabine therapy in AIDS patients. This usually occurs during the first 4 to 6 weeks of therapy (McNeely et al, 1989) Yarchoan et al, 1988; (Merigan et al, 1989; Merigan & Skowron, 1990; Broder, 1990).
    d) LAMIVUDINE: Skin rashes and/or pruritus have been reported in approximately 10% of patients during oral therapy with lamivudine (Pluda et al, 1995; van Leeuwen et al, 1995).
    e) LAMIVUDINE: Henoch-Schonlein purpura has been reported in 1 patient following therapy with oral lamivudine in doses of 12 mg/kg/day, resulting in discontinuance of therapy (Pluda et al, 1995).
    2) WITH POISONING/EXPOSURE
    a) LAMIVUDINE: A 17-year-old developed a generalized, pruritic, maculopapular rash 1 day after ingesting lamivudine 3.9 g and efavirenz 15.6 g (Boscacci et al, 2006).
    B) STEVENS-JOHNSON SYNDROME
    1) WITH THERAPEUTIC USE
    a) INCIDENCE: Stevens-Johnson syndrome or toxic epidermal necrolysis develops in less than 0% to 5% of NRTI treated patients. These have not been reported with abacavir therapy (Carr & Cooper, 2000).
    C) DISCOLORATION OF SKIN
    1) WITH THERAPEUTIC USE
    a) TRICITABINE/EMTRICITABINE can cause hyperpigmentation of the skin and nails (Luther & Glesby, 2007).
    D) ALOPECIA
    1) WITH THERAPEUTIC USE
    a) LAMIVUDINE: Hair loss has been reported in 5 of 16 patients in a lamivudine clinical trial (Fong, 1994).

Musculoskeletal

    3.15.2) CLINICAL EFFECTS
    A) DRUG-INDUCED MYOPATHY
    1) WITH THERAPEUTIC USE
    a) STAVUDINE: Creatine phosphokinase was greater than 5 times normal in 7 out of 41 patients in a clinical trial of stavudine (Murray et al, 1995).
    b) ZIDOVUDINE/LAMIVUDINE: During a clinical trial, 8% of patients receiving lamivudine plus zidovudine experienced myalgias (Prod Info EPIVIR(R) oral tablets, solution, 2008).
    B) JOINT PAIN
    1) WITH THERAPEUTIC USE
    a) ZALCITABINE: Dose related arthralgias have been reported during zalcitabine therapy (Merigan & Skowron, 1990; Broder, 1990).
    C) BONE PAIN
    1) WITH THERAPEUTIC USE
    a) TENOFOVIR: Bone pain occurred in 12 of 22 patients diagnosed with tenofovir-associated renal toxicity. Osteomalacia was confirmed in 7 patients via an isotope bone scan. All 7 patients had an elevated alkaline phosphatase concentration after initiating therapy with tenofovir (Woodward et al, 2009).

Endocrine

    3.16.2) CLINICAL EFFECTS
    A) HYPERGLYCEMIA
    1) WITH THERAPEUTIC USE
    a) CASE REPORT: Verma et al (1999) reported acute hyperglycemia (blood glucose ranging between 128 and 251 mg%) with fulminant axonal neuropathy and lactic acidosis in a patient taking NRTI therapy. A toxic neuropathic reaction was suggested by the parallel association of lactic acidosis and hyperglycemia with mixed sensory and motor axonal degeneration (Verma et al, 1999).

Immunologic

    3.19.2) CLINICAL EFFECTS
    A) ACUTE ALLERGIC REACTION
    1) The NRTI, abacavir, is a common antiretroviral drug that causes hypersensitivity. Hypersensitivity reactions are uncommon with other NRTIs. Abacavir-induced hypersensitivity rash occurs in approximately 3% of patients with an average onset of 9 days (Carr & Cooper, 2000). Drug hypersensitivity in HIV-1 infected patients is about 100 times more common than in the general population. Carr & Cooper (2000) report the following typical clinical features of HIV-associated drug hypersensitivity:
    1) Morbilliform/maculopapular rash
    2) Fever (often precedes rash)
    3) Myalgias, fatigue
    4) Mucosal ulceration
    2) ZALCITABINE: Two cases of zalcitabine-induced hypersensitivity syndrome have been reported. Symptoms in both cases included diffuse maculopapular eruption, fever, lymphadenopathy, significant eosinophilia (70% in one case), facial swelling, and an exfoliative dermatitis (Tancrede-Bohin et al, 1996).
    3) ABACAVIR has been reported to cause hypersensitivity syndrome reactions in 2% to 3% of clinical trial recipients. Onset of this reaction is typically within the first 6 weeks of drug therapy. Reactions have consisted of drug fever with nausea/vomiting, malaise and/or rash (Foster & Faulds, 1998; Dobkin, 1999). Patients exhibiting a hypersensitivity syndrome should have abacavir therapy discontinued and the patient should NOT be re-challenged with abacavir.
    a) Abnormal laboratory values have included: lymphopenia, increased liver function tests, and occasionally increased creatine kinase levels or thrombocytopenia. With continued dosing, symptoms worsen, but resolve within a few days on discontinuation of therapy. With drug re-challenge, multi-organ failure and death may result. Fatalities have been reported.
    b) Initial presentation of fatal hypersensitivity reactions has included respiratory symptoms of dyspnea, cough, or pharyngitis. Delays in diagnosis of hypersensitivity can result in continuation of abacavir, or re-introduction of the drug, resulting in life-threatening hypotension and death (Anon, 2000).

Reproductive

    3.20.1) SUMMARY
    A) Most AIDS antiviral agents are classified as FDA pregnancy category C or B. Efavirenz/emtricitabine/tenofovir is classified as FDA pregnancy category D. Transient anemia and other blood abnormalities (neutropenia, thrombocytopenia, and lymphopenia), as well as hyperlactatemia, have been reported in zidovudine-exposed, but HIV-uninfected infants.
    B) An increased risk of severe or fatal lactic acidosis has been seen in pregnant women who take the combination of HIV drugs, stavudine and didanosine, with other antiretroviral agents. Pancreatitis is also a well-documented complication of stavudine and didanosine.
    3.20.2) TERATOGENICITY
    A) SPINAL MALFORMATION
    1) Two case reports describe HIV-positive women treated with combination antiretroviral therapy who unexpectedly conceived and whose fetuses progressively developed spinal malformations. The first woman's regimen included zidovudine, zalcitabine, and sulfamethoxazole/trimethoprim at the time of conception and throughout pregnancy. Folic acid 10 mg/day was added in an attempt to lessen the antifolate activity of sulfamethoxazole/trimethoprim. At 32 weeks' gestation, a fetal ultrasound revealed hemivertebrae in the lumbar spine. A viable infant was delivered at term by Cesarean section; a bony mass was present in the lumbar spine but no neurological abnormalities were noted. The second woman's medications included didanosine, stavudine, nevirapine, and sulfamethoxazole/trimethoprim at the time of conception and throughout pregnancy. A fetal ultrasound performed at 19 weeks gestation was significant for spina bifida and ventriculomegaly. The pregnancy was electively terminated (Richardson et al, 2000).
    B) OTHER NON-SPECIFIC
    1) ABACAVIR
    a) There have been sufficient numbers of first trimester exposures to abacavir to detect at least a 2-fold increase in the risk of birth defects. No increases in birth defects overall have been observed in data collected from the Antiretroviral Pregnancy Registry. Prevalence of birth defects associated with maternal first trimester abacavir use was 3% (25 of 823 births; 95% confidence interval, 2% to 4.5%) compared with the total United States population-based prevalence of 2.7% (Prod Info ZIAGEN(R) oral tablets, oral solution, 2015a; Prod Info EPZICOM(R) oral tablets, 2015a; Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    2) DIDANOSINE
    a) A slight increase in birth defects overall has been observed in data collected from the Antiretroviral Pregnancy Registry. Prevalence of birth defects associated with maternal first-trimester didanosine use was 4.6% (19 of 409 births; 95% confidence interval (CI), 2.8% to 7.2%) compared with the total United States population-based prevalence of 2.7%. No pattern of defects was discovered (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    b) After a single oral didanosine dose of 375 mg in 2 women, maternal blood levels were 295 and 629 nanograms (ng)/mL; corresponding fetal blood levels were 42 and 121 ng/mL. Amniotic fluid levels were less than 5 ng/mL and 135 ng/mL, respectively (Pons et al, 1991).
    3) EMTRICITABINE
    a) There have been sufficient numbers of first trimester exposure to emtricitabine to detect at least a 2-fold increase in risk of birth defects. No increases in birth defects overall have been observed in data collected from the Antiretroviral Pregnancy Registry. Prevalence of birth defects associated with maternal first-trimester emtricitabine use was 2.3% (21 of 899 births; 95% confidence interval, 1.4% to 3.5%), compared with the total United States population-based prevalence of 2.7% (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    4) LAMIVUDINE
    a) There have been sufficient numbers of first trimester exposures to lamivudine to detect at least a 1.5-fold increase in risk of birth defects. No increases in birth defects overall have been observed in data collected from the Antiretroviral Pregnancy Registry. Prevalence of birth defects associated with maternal first trimester lamivudine use was 3.1% (127 of 4088 births; 95% confidence interval, 2.6% to 3.7%) compared with the total United States population-based prevalence of 2.7% (Prod Info EPIVIR(R) oral tablets, oral solution, 2015b; Prod Info EPZICOM(R) oral tablets, 2015a; Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    5) STAVUDINE
    a) There have been sufficient numbers of first trimester exposure to stavudine to detect at least a 2-fold increase in risk of birth defects. No increases in birth defects overall with stavudine have been observed in data collected from the Antiretroviral Pregnancy Registry. Prevalence of birth defects associated with maternal first trimester stavudine use was 2.5% (20 of 801 births; 95% confidence interval, 1.5% to 3.8%) compared with the total United States population-based prevalence of 2.7% (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    6) TENOFOVIR
    a) In a prospective cohort study, maternal use of tenofovir disoproxil fumarate any time during pregnancy was not associated with increased risk for infant adverse birth outcomes of low birth weight (LBW) or small for gestational age (SGA) compared with tenofovir-unexposed infants; however, tenofovir-exposed infants had a slightly lower mean age-appropriate length and head circumference at 1 year. The incidence of tenofovir use was 21% (449/2029 HIV-infected mothers), with a median duration of 4.8 months; 13% had first trimester exposure. Based on adjusted odds ratios (aOR), tenofovir exposure was not associated with LBW (aOR, 0.87; p=0.4) or SGA (aOR, 1.04; p=0.88) at birth; further adjustment for gestational age did not affect the results. There was also no association with tenofovir exposure and newborn length-for-age z-scores (LAZ) and head circumference-for-age z-scores (HCAZ). However, at age 1 year, infants exposed in utero to tenofovir-containing combination antiretroviral therapy had slightly but statistically significantly lower adjusted mean LAZ (-0.17 vs -0.03; p=0.04) and HCAZ (0.17 vs 0.42; p=0.02) compared with those unexposed to tenofovir. These results correspond approximately to a mean 0.41 cm shorter length and a mean 0.32 cm smaller head circumference at 1 year in the tenofovir-exposed infants (Siberry et al, 2012).
    b) There have been sufficient numbers of first trimester exposures to tenofovir to detect at least a 2-fold increase in risk of birth defects. No increases in birth defects overall have been observed in data collected from the Antiretroviral Pregnancy Registry. Prevalence of birth defects associated with maternal first trimester tenofovir use was 2.3% (31 of 1370 births; 95% confidence interval, 1.5% to 3.2%) compared with the total United States population-based prevalence of 2.7% (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    7) ZIDOVUDINE
    a) There have been sufficient numbers of first trimester exposures to zidovudine to detect at least a 1.5-fold increase in risk of birth defects. No increases in birth defects overall have been observed in data collected from the Antiretroviral Pregnancy Registry. Prevalence of birth defects associated with maternal first trimester zidovudine use was 3.3% (124 of 3789 births; 95% confidence interval, 2.7% to 3.9%) compared with the total United States population-based prevalence of 2.7% (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    b) According to one study, there is a possible link between mitochondrial dysfunction and perinatal administration of prophylactic nucleoside analogues, specifically zidovudine. Eight children exposed to zidovudine in-utero had mitochondrial dysfunction. Of these, 5 presented with delayed neurological symptoms and 2 of these died, while 3 were symptom-free but had severe biological or neurological abnormalities (Blanche et al, 1999).
    c) In zidovudine-exposed, but HIV-uninfected infants, transient anemia and other blood abnormalities (ie, neutropenia, thrombocytopenia, and lymphopenia), as well as hyperlactatemia have been documented (Venhoff & Walker, 2006).
    C) LACK OF EFFECT
    1) EMTRICITABINE
    a) In human studies with emtricitabine, there was no difference in the risk of major birth defects compared to the background rate of birth defects (Prod Info ODEFSEY(R) oral tablets, 2016; Prod Info DESCOVY(R) oral tablets, 2016).
    2) TENOFOVIR
    a) In a retrospective analysis of pregnant women infected with highly viremic hepatitis B virus (HBV) (defined as HBV-DNA levels of 6 or more log(10) copies/mL prior to treatment) and who received oral tenofovir disoproxil fumarate 300 mg once daily beginning in the third trimester, 11 Asian women gave birth to 11 infants who did not have birth defects or congenital malformations. Maternal use of tenofovir was started at the medial gestational age of 29 weeks (range, 28 to 32 weeks) and the median duration of use before delivery was 10 weeks (range, 7 to 12 weeks). Serum HBV-DNA levels were significantly reduced in the mother at delivery compared with baseline (mean, 5.25 +/- 1.79 vs 8.87 +/- 0.45 log(10) copies/mL, respectively; p less than 0.01). At birth and subsequently, all infants received appropriate immunoprophylaxis with hepatitis B immunoglobulin and vaccination. At 28 to 36 weeks after birth, all infants were hepatitis B surface antigen negative. While these preliminary results suggest that tenofovir use in the third trimester was safe and effective in preventing vertical transmission from highly viremic HBV-infected mothers, larger, prospectively randomized and controlled studies are needed to confirm these findings (Pan et al, 2012).
    b) In a prospective cohort study of prevention of mother-to-child HIV transmission in HIV-infected pregnant women with intrapartum dosing of tenofovir disoproxil fumarate 600 mg, there were no significant neonatal adverse effects and the median ratio of tenofovir concentrations in cord blood to maternal plasma was 0.73 (range, 0.26 to 1.95). Women administered one dose of 600 mg tenofovir at the onset of active labor or up to 8 hours before a planned cesarean section (group 1, n=13 women and 10 infants) had infants with 76 ng/mL median tenofovir cord BCs, with a higher median cord BC with cesarean deliveries (94 ng/mL). The median BC in all group 1 infants fell to 12 ng/mL by 12 hours postpartum. In groups 2 (n=7 women and 7 infants) and 3 (n=8 women and 9 infants), women received one dose of tenofovir 900 mg intrapartum and infants received a tenofovir dose 4 mg/kg post delivery; in group 3, women and infants also received emtricitabine 600 mg and 3 mg/kg, respectively. The median tenofovir cord BC was lower than group 1 (68 ng/mL), but higher with cesarean sections (110 ng/mL). Infants had a median tenofovir BC of 29 ng/mL before postpartum dosing, and was 46 ng/mL and 30 ng/mL within 24 and 36 hours, respectively, of dosing (n=14) (Flynn et al, 2011).
    c) In a retrospective case series of 15 pregnant HIV-infected women in an urban clinic, tenofovir was well-tolerated without serious adverse events; however, long-term evaluation of tenofovir's effects on childhood growth and a larger prospective evaluation are warranted. Between 2001 and 2005, 15 women received tenofovir (standard dose) as a part of highly active antiretroviral therapy (HAART) for 16 pregnancies to prevent mother-to-infant transmission of HIV. The median nadir CD4+ count was 191 cells per microliter (range, 0 to 676). Two women were treated with tenofovir at the beginning of the pregnancy, 13 women started tenofovir at a median of 19 weeks gestation, and 3 women began treatment during the third trimester. Median exposure to tenofovir in utero was 127 days. All women received intravenous zidovudine during delivery. Complications were reported in 9 pregnancies, but they were not related to tenofovir treatment. Fifteen women had successful deliveries at a median of 36 weeks gestation, with a median birth weight of 3255 g. One neonate was born at 30 weeks and weighed 2500 g. Overall, in a cohort of heavily treatment-experienced women, tenofovir was an adequately tolerated component of HAART (Nurutdinova et al, 2008).
    D) ANIMAL STUDIES
    1) ABACAVIR
    a) Animal studies conducted with the individual components of the product suggest that abacavir and lamivudine are transferred to the fetus through the placenta. Fetal malformations and developmental toxicities were reported in animals administered abacavir at doses up to 35 times the human exposure. Embryonic and fetal toxicities as well as offspring toxicities were reported with abacavir at doses approximately half of the above mentioned dose. Administration of lamivudine at doses approximately 35 times the recommended adult dose showed evidence of early embryolethality. There were no reports of teratogenicity. Teratogenicity was not reported with zidovudine administration at doses up to 600 mg/kg/day, however, embryofetal toxicity was observed. Maternal toxicity and an increase in fetal malformations was reported with zidovudine doses approximately 350 times peak human plasma concentrations (Prod Info TRIZIVIR oral tablets, 2015).
    2) EMTRICITABINE
    a) In animal studies, no adverse effects were observed at emtricitabine doses up to 108 times the recommended daily dose for humans (Prod Info ODEFSEY(R) oral tablets, 2016; Prod Info DESCOVY(R) oral tablets, 2016).
    b) In embryofetal toxicity studies of emtricitabine in animals at exposures of up to approximately 120-fold higher than human exposure based on AUC at the recommended daily doses, there was no increase in fetal variations or malformations (Prod Info TRUVADA(R) oral tablets, 2012; Prod Info EMTRIVA(R) oral capsule, solution, 2012).
    3) LAMIVUDINE
    a) Animal studies conducted with the individual components of the product suggest that abacavir and lamivudine are transferred to the fetus through the placenta. Fetal malformations and developmental toxicities were reported in animals administered abacavir at doses up to 35 times the human exposure. Embryonic and fetal toxicities as well as offspring toxicities were reported with abacavir at doses approximately half of the above mentioned dose. Administration of lamivudine at doses approximately 35 times the recommended adult dose showed evidence of early embryolethality. There were no reports of teratogenicity. Teratogenicity was not reported with zidovudine administration at doses up to 600 mg/kg/day, however, embryofetal toxicity was observed. Maternal toxicity and an increase in fetal malformations was reported with zidovudine doses approximately 350 times peak human plasma concentrations (Prod Info TRIZIVIR oral tablets, 2015).
    b) Doses of lamivudine up to 130 and 60 times the adult dose in animals did not produce any teratogenic effects (Prod Info EPIVIR(R) oral tablets, solution, 2009).
    4) LAMIVUDINE/ZIDOVUDINE
    a) Prenatal exposure to combined maternal treatment with zidovudine and lamivudine was reported to result in significant, although minor, alterations in neurobehavioral development of animals. Early development of prenatally exposed animals was delayed, particularly affecting somatic growth and maturation of several sensorimotor reflexes by one or two days (Venerosi et al, 2001).
    5) STAVUDINE
    a) No evidence of teratogenicity was seen in animals with exposures up to 399 and 183 times, respectively, of that seen at human clinical dosage of 1 mg/kg/day (Prod Info ZERIT(R) oral capsules, solution, 2009).
    6) TENOFOVIR
    a) In animal reproduction studies in which animals received tenofovir doses up to 19 times the human dose based on body surface area, there was no evidence of impaired fertility or harm to the fetus (Prod Info VIREAD(R) oral tablets, powder, 2012; Prod Info TRUVADA(R) oral tablets, 2012).
    7) TENOFOVIR ALAFENAMIDE
    a) In animal studies, no adverse effects were observed at tenofovir alafenamide doses up to 53 times the recommended daily dose for humans (Prod Info ODEFSEY(R) oral tablets, 2016; Prod Info DESCOVY(R) oral tablets, 2016).
    8) TENOFOVIR ALAFENAMIDE/COBICISTAT/ELVITEGRAVIR/EMTRICITABINE
    a) During animal studies with single-agent administration, there were no reports of teratogenicity or adverse effects on reproductive function with elvitegravir at doses up to 23 times the recommended human dose RHD) or with cobicistat at doses up to 3.8 times the RHD. There was no reported increase in fetal variations or malformations with administration of emtricitabine at doses up to approximately 108 times the RHD. Similarly, there was no evidence of impaired fertility or fetal harm due to administration of tenofovir alafenamide at doses approximately 53 times greater than the RHD (Prod Info GENVOYA(R) oral tablets, 2015).
    9) ZIDOVUDINE
    a) Animal studies conducted with the individual components of the product suggest that abacavir and lamivudine are transferred to the fetus through the placenta. Fetal malformations and developmental toxicities were reported in animals administered abacavir at doses up to 35 times the human exposure. Embryonic and fetal toxicities as well as offspring toxicities were reported with abacavir at doses approximately half of the above mentioned dose. Administration of lamivudine at doses approximately 35 times the recommended adult dose showed evidence of early embryolethality. There were no reports of teratogenicity. Teratogenicity was not reported with zidovudine administration at doses up to 600 mg/kg/day, however, embryofetal toxicity was observed. Maternal toxicity and an increase in fetal malformations was reported with zidovudine doses approximately 350 times peak human plasma concentrations (Prod Info TRIZIVIR oral tablets, 2015).
    3.20.3) EFFECTS IN PREGNANCY
    A) PREGNANCY CATEGORY
    1) ABACAVIR
    a) Abacavir has been classified as FDA pregnancy category C (Prod Info ZIAGEN(R) oral tablets, solution, 2008a).
    2) ABACAVIR/DOLUTEGRAVIR/LAMIVUDINE
    a) Abacavir/dolutegravir/lamivudine combination therapy has been classified as FDA pregnancy category C (Prod Info TRIUMEQ(R) oral tablets, 2014).
    3) ABACAVIR/LAMIVUDINE
    a) Abacavir/lamivudine has been classified as FDA pregnancy category C (Prod Info EPZICOM(R) oral tablets, 2009).
    4) ABACAVIR/LAMIVUDINE/ZIDOVUDINE
    a) Abacavir/lamivudine/zidovudine has been classified as FDA pregnancy category C (Prod Info TRIZIVIR oral tablets, 2015).
    5) COBICISTAT/ELVITEGRAVIR/EMTRICITABINE/TENOFOVIR
    a) Cobicistat/elvitegravir/emtricitabine/tenofovir has been classified as FDA pregnancy category B (Prod Info STRIBILD(TM) oral tablets, 2012).
    6) DELAVIRDINE
    a) Delavirdine has been classified as FDA pregnancy category C (Prod Info RESCRIPTOR(R) oral tablets, 2008).
    7) DIDANOSINE
    a) Didanosine has been classified as FDA pregnancy category B (Prod Info VIDEX(R) pediatric powder for oral solution, 2010).
    8) EFAVIRENZ/EMTRICITABINE/TENOFOVIR
    a) Efavirenz/emtricitabine/tenofovir has been classified as FDA pregnancy category D (Prod Info ATRIPLA(R) oral tablets, 2010).
    9) EMTRICITABINE
    a) Emtricitabine has been classified as FDA pregnancy category B (Prod Info EMTRIVA(R) oral capsule, solution, 2012).
    10) EMTRICITABINE/RILPIVIRINE/TENOFOVIR
    a) Emtricitabine/rilpivirine/tenofovir has been classified as FDA pregnancy category B (Prod Info COMPLERA(TM) oral tablets, 2011).
    11) EMTRICITABINE/RILPIVIRINE/TENOFOVIR ALAFENAMIDE
    a) Although human studies with emtricitabine show no difference in the risk of major birth defects compared to the background rate of birth defects, there are not enough data to evaluate the use of emtricitabine/rilpivirine/tenofovir alafenamide during pregnancy (Prod Info ODEFSEY(R) oral tablets, 2016).
    b) Patients exposed to emtricitabine/rilpivirine/tenofovir alafenamide during pregnancy may register with the Antiretroviral Pregnancy Registry by calling 1-800-258-4263 (Prod Info ODEFSEY(R) oral tablets, 2016).
    12) EMTRICITABINE/TENOFOVIR
    a) Emtricitabine/tenofovir has been classified as FDA pregnancy category B(Prod Info TRUVADA(R) oral tablets, 2012).
    13) EMTRICITABINE/TENOFOVIR ALAFENAMIDE
    a) Administer emtricitabine/tenofovir alafenamide to a pregnant woman only if the potential benefit outweighs the potential risk to the fetus.(Prod Info DESCOVY(R) oral tablets, 2016).
    b) Patients exposed to emtricitabine/tenofovir alafenamide during pregnancy may register with the Antiretroviral Pregnancy Registry by calling 1-800-258-4263 (Prod Info DESCOVY(R) oral tablets, 2016).
    14) LAMIVUDINE
    a) Lamivudine has been classified as FDA pregnancy category C (Prod Info EPIVIR(R) oral tablets, solution, 2008).
    15) STAVUDINE
    a) Stavudine has been classified as FDA pregnancy category C (Prod Info ZERIT(R) oral capsules, solution, 2009).
    16) TENOFOVIR
    a) Tenofovir has been classified as FDA pregnancy category B (Prod Info VIREAD(R) oral tablets, powder, 2012).
    17) TENOFOVIR ALAFENAMIDE/COBICISTAT/ELVITEGRAVIR/EMTRICITABINE
    a) The combination substance of tenofovir alafenamide/cobicistat/elvitegravir/emtricitabine has been classified as FDA pregnancy category B (Prod Info GENVOYA(R) oral tablets, 2015).
    b) Use during pregnancy only if the potential maternal benefit outweighs the potential fetal risk. Patients exposed to cobicistat/elvitegravir/emtricitabine/tenofovir alafenamide during pregnancy may register with the Antiretroviral Pregnancy Registry by calling 1-800-258-4263 (Prod Info GENVOYA(R) oral tablets, 2015).
    18) ZALCITABINE
    a) Zalcitabine has been classified as FDA pregnancy category C (Prod Info HIVID(R), 2002).
    b) NOTE: Zalcitabine is no longer available in the United States (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2010).
    19) ZIDOVUDINE
    a) Zidovudine has been classified as FDA pregnancy category C (Prod Info RETROVIR(R) oral tablets, capsules, syrup, 2008).
    B) LACTIC ACIDOSIS
    1) The FDA has issued a warning concerning an increased risk of severe or fatal lactic acidosis in pregnant women who take the combination of HIV drugs, stavudine and didanosine, with other antiretroviral agents. Pancreatitis is also a well-documented complication of stavudine and didanosine (Prod Info Epivir(R), lamivudine, 2001; Prod Info Zerit(R),, 2002).
    a) Three cases of fatal lactic acidosis, with and without pancreatitis, were reported in pregnant women taking stavudine and didanosine in combination with other antiretroviral drugs. Several nonfatal cases in pregnant women (with and without pancreatitis) were also reported. Although it has been suggested that women may be at an increased risk of developing lactic acidosis and liver toxicity, it is unclear if pregnancy potentiates these known adverse events ((Anon, 2001)).
    C) MATERNAL HEPATOTOXICITY
    1) EFAVIRENZ, LAMIVUDINE, AND ZIDOVUDINE
    a) CASE REPORT: Hepatotoxicity was reported in a 28-year-old pregnant woman receiving antiretroviral treatment with zidovudine, lamivudine, and efavirenz. At week 18 of gestation and about 5 months after antiretroviral therapy initiation, she presented with sclera icterus and jaundice. Lab values showed elevated transaminases and bilirubin. Following discontinuation of her medications, transaminase levels declined, but hyperbilirubinemia persisted until post-delivery. Lab values returned to normal five months after delivery of a viable infant (Hill et al, 2001).
    D) MITOCHONDRIAL TOXICITY
    1) Nucleoside analogue drugs have been linked to mitochondrial toxicity, including symptomatic lactic acidosis and hepatic steatosis. Despite this possible association, there are clear benefits to nucleoside analogues in decreasing the transmission of HIV from mother to child. Combination therapy that includes didanosine and stavudine concomitantly should not be used during pregnancy, as maternal mortality due to lactic acidosis and hepatic failure has been reported with long-term use (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    E) PLACENTAL BARRIER
    1) All of the following NRTIs are reported to cross the placental barrier with newborn to maternal drug ratio reported as follows (Taylor & Low-Beer, 2001; Stek et al, 2012):
    1) ABACAVIR: Yes (rat)
    2) DIDANOSINE: 0.5 (human)
    3) EMTRICITABINE: 1.2 (human)
    4) LAMIVUDINE: 1
    5) STAVUDINE: 0.76
    6) ZIDOVUDINE: 0.85 (human)
    7) ZALCITABINE: 0.3 to 0.5 (rhesus monkey)
    F) LACK OF EFFECT
    1) Antiretroviral therapy was not associated with increased, unadjusted rates of premature labor and delivery in pregnant women infected with HIV-1, and there was no difference between treated and untreated groups in the adjusted, overall rates of adverse pregnancy events. In a meta-analysis of pregnant women with HIV-1 infection who were enrolled in 7 clinical studies (PACTG 076 and 185; PACTS, WITS, and 3 single-site studies), the unadjusted rates of premature delivery for women receiving any antiretroviral treatment (n=2123) were significantly lower than for women not treated (n=1143; p=0.02). There was no difference between women treated without protease inhibitor-based regimens compared with women receiving multi-agent regimens including protease inhibitors (Tuomala et al, 2002).
    2) ABACAVIR
    a) A meta-analysis of 2123 pregnant women infected with the human immunodeficiency virus did not show a difference in adjusted overall rates of adverse pregnancy events between treated and untreated groups. However, unadjusted rates of premature delivery were lower in treated women compared with those who were not treated; rates of premature and very premature delivery were similar between combination regimens that included and excluded protease inhibitors. The risk of low birth weight was lower among infants born to mothers receiving combination therapy without protease inhibitors (n=396) compared with infants born to women receiving monotherapy (n=1590; odds ratio (OR), 0.58; 95% confidence interval (CI), 0.41 to 0.84). The risk of low birth weight was greater for infants of mothers treated with combination regimens that included protease inhibitors (n=137) than for infants of mothers receiving combination regimens without protease inhibitors (OR, 2.03, 95% CI, 1.16 to 3.54) (Tuomala et al, 2002a).
    3) LAMIVUDINE/STAVUDINE
    a) CASE REPORT: An HIV-positive pregnant woman treated with stavudine 40 mg twice a day and lamivudine 150 mg twice a day delivered a healthy, normal child at term. The antiviral treatment was initiated at gestational week 20 and was well-tolerated. The pregnancy was uneventful and fetal growth considered normal. At nine months of age, the child was HIV-negative and developing normally (Ristola et al, 1999).
    4) TENOFOVIR
    a) In a retrospective analysis of pregnant women infected with highly viremic hepatitis B virus (HBV) (defined as HBV-DNA levels of 6 or more log(10) copies/mL prior to treatment) and who received oral tenofovir disoproxil fumarate 300 mg once daily beginning in the third trimester, 11 Asian women gave birth to 11 infants who did not have birth defects or congenital malformations. Maternal use of tenofovir was started at the median gestational age of 29 weeks (range, 28 to 32 weeks) and the median duration of use before delivery was 10 weeks (range, 7 to 12 weeks). Serum HBV-DNA levels were significantly reduced in the mother at delivery compared with baseline (mean, 5.25 +/- 1.79 vs 8.87 +/- 0.45 log(10) copies/mL, respectively; p less than 0.01). At birth and subsequently, all infants received appropriate immunoprophylaxis with hepatitis B immunoglobulin and vaccination. At 28 to 36 weeks after birth, all infants were hepatitis B surface antigen negative. While these preliminary results suggest that tenofovir use in the third trimester was safe and effective in preventing vertical transmission from highly viremic HBV-infected mothers, larger, prospectively randomized and controlled studies are needed to confirm these findings (Pan et al, 2012).
    b) In a retrospective case series of 15 pregnant HIV-infected women in an urban clinic, tenofovir was well-tolerated without serious adverse events; however, long-term evaluation of tenofovir's effects on childhood growth and a larger prospective evaluation are warranted. Between 2001 and 2005, 15 women received tenofovir (standard dose) as a part of highly active antiretroviral therapy (HAART) for 16 pregnancies to prevent mother-to-infant transmission of HIV. The median nadir CD4+ count was 191 cells/mcL (range, 0 to 676). Two women were treated with tenofovir at the beginning of the pregnancy, 13 women started tenofovir at a median of 19 weeks gestation, and 3 women began treatment during the third trimester. Median exposure to tenofovir in utero was 127 days. All women received intravenous zidovudine during delivery. Complications were reported in 9 pregnancies, but they were not related to tenofovir treatment. Fifteen women had successful deliveries at a median of 36 weeks gestation, with a median birth weight of 3255 g. One neonate was born at 30 weeks and weighed 2500 g. Overall, in a cohort of heavily treatment-experienced women, tenofovir was an adequately tolerated component of HAART (Nurutdinova et al, 2008).
    G) ANIMAL STUDIES
    1) ABACAVIR
    a) Animal studies conducted with the individual components of the product suggest that abacavir and lamivudine are transferred to the fetus through the placenta. Fetal malformations and developmental toxicities were reported in animals administered abacavir at doses up to 35 times the human exposure. Embryonic and fetal toxicities as well as offspring toxicities were reported with abacavir at doses approximately half of the above mentioned dose. Administration of lamivudine at doses approximately 35 times the recommended adult dose showed evidence of early embryolethality. There were no reports of teratogenicity. Teratogenicity was not reported with zidovudine administration at doses up to 600 mg/kg/day, however, embryofetal toxicity was observed. Maternal toxicity and an increase in fetal malformations was reported with zidovudine doses approximately 350 times peak human plasma concentrations (Prod Info TRIZIVIR oral tablets, 2015).
    2) LAMIVUDINE
    a) Animal studies conducted with the individual components of the product suggest that abacavir and lamivudine are transferred to the fetus through the placenta. Fetal malformations and developmental toxicities were reported in animals administered abacavir at doses up to 35 times the human exposure. Embryonic and fetal toxicities as well as offspring toxicities were reported with abacavir at doses approximately half of the above mentioned dose. Administration of lamivudine at doses approximately 35 times the recommended adult dose showed evidence of early embryolethality. There were no reports of teratogenicity. Teratogenicity was not reported with zidovudine administration at doses up to 600 mg/kg/day, however, embryofetal toxicity was observed. Maternal toxicity and an increase in fetal malformations was reported with zidovudine doses approximately 350 times peak human plasma concentrations (Prod Info TRIZIVIR oral tablets, 2015).
    b) There were indications of early embryolethality when lamivudine was given to animals at doses similar to the maximum recommended human dose (Prod Info EPIVIR(R) oral tablets, solution, 2009).
    3) TENOFOVIR
    a) In studies in animals, decreased fetal growth and reduced fetal bone porosity was noted within 2 months of exposure when maternal animals received tenofovir at doses 2-fold higher than those used for human therapeutic use (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2010).
    4) TENOFOVIR ALAFENAMIDE/COBICISTAT/ELVITEGRAVIR/EMTRICITABINE
    a) During animal studies with single-agent administration, there were no reports of teratogenicity or adverse effects on reproductive function with elvitegravir at doses up to 23 times the recommended human dose RHD) or with cobicistat at doses up to 3.8 times the RHD. There was no reported increase in fetal variations or malformations with administration of emtricitabine at doses up to approximately 108 times the RHD. Similarly, there was no evidence of impaired fertility or fetal harm due to administration of tenofovir alafenamide at doses approximately 53 times greater than the RHD (Prod Info GENVOYA(R) oral tablets, 2015).
    5) ZIDOVUDINE
    a) Animal studies conducted with the individual components of the product suggest that abacavir and lamivudine are transferred to the fetus through the placenta. Fetal malformations and developmental toxicities were reported in animals administered abacavir at doses up to 35 times the human exposure. Embryonic and fetal toxicities as well as offspring toxicities were reported with abacavir at doses approximately half of the above mentioned dose. Administration of lamivudine at doses approximately 35 times the recommended adult dose showed evidence of early embryolethality. There were no reports of teratogenicity. Teratogenicity was not reported with zidovudine administration at doses up to 600 mg/kg/day, however, embryofetal toxicity was observed. Maternal toxicity and an increase in fetal malformations was reported with zidovudine doses approximately 350 times peak human plasma concentrations (Prod Info TRIZIVIR oral tablets, 2015).
    3.20.4) EFFECTS DURING BREAST-FEEDING
    A) LACK OF INFORMATION
    1) ABACAVIR, DIDANOSINE, STAVUDINE, TENOFOVIR: At the time of this review, no data were available to assess the potential effects of exposure to this agent during lactation in humans (Prod Info ZIAGEN(R) oral solution, oral tablets, 2008; Prod Info VIDEX(R) pediatric powder for oral solution, 2010; Prod Info ZERIT(R) oral capsules, solution, 2009; Prod Info VIREAD(R) oral tablets, 2010).
    B) BREAST MILK
    1) TENOFOVIR ALAFENAMIDE/COBICISTAT/ELVITEGRAVIR/EMTRICITABINE
    a) Avoid breastfeeding while taking this drug (Prod Info GENVOYA(R) oral tablets, 2015).
    b) It is unknown whether cobicistat, elvitegravir, or tenofovir are excreted into human breast milk. Emtricitabine was detected samples of breast milk from 5 HIV-1 infected mothers. Breastfeeding infants exposed to emtricitabine may be at risk for developing a viral resistance to emtricitabine. Additional adverse effects are currently unknown (Prod Info GENVOYA(R) oral tablets, 2015).
    2) EMTRICITABINE/RILPIVIRINE/TENOFOVIR ALAFENAMIDE
    a) Mothers should not breastfeed while taking emtricitabine/rilpivirine/tenofovir alafenamide (Prod Info ODEFSEY(R) oral tablets, 2016).
    3) EMTRICITABINE/TENOFOVIR ALAFENAMIDE
    a) Advise women to avoid breastfeeding during therapy (Prod Info DESCOVY(R) oral tablets, 2016).
    4) ABACAVIR: Is excreted in breast milk of animals, but it is not known if it is excreted in human milk (Prod Info Ziagen(TM), abacavir, 1998).
    5) DIDANOSINE: It is not known whether didanosine appears in human milk. Animal studies indicate that didanosine and/or metabolites do appear in animal milk. Patients taking didanosine are advised to discontinue breast feeding (Prod Info Videx(R), didanosine, 1996).
    6) EMTRICITABINE: Samples of human breast milk suggest that emtricitabine is excreted into human breast milk (Prod Info ODEFSEY(R) oral tablets, 2016)at low levels. The impact on breastfed infants exposed to emtricitabine is unknown (Prod Info EMTRIVA(R) oral capsule, solution, 2012; Prod Info STRIBILD(TM) oral tablets, 2012).
    7) EMTRICITABINE/TENOFOVIR: In a pharmacokinetic analysis of breast milk samples of 5 HIV-infected mothers who exclusively breastfed their infants, median emtricitabine and tenofovir breast milk doses represented 2% and 0.03%, respectively, of the proposed oral infant doses. The women received a nevirapine 200 mg dose plus 2 tablets of emtricitabine 200 mg/tenofovir 300 mg at the start of labor, then 1 emtricitabine 200 mg/tenofovir 300 mg tablet once daily for 7 days postpartum. Among breast milk samples collected 10 minutes to 21 hours post maternal drug administration, median maximal concentrations of emtricitabine and tenofovir were 679 nanograms (ng)/mL (interquartile range (IQR), 658 to 743 ng/mL) and 14.1 ng/mL (IQR, 11.6 to 16.25 ng/mL), respectively; corresponding median minimal concentrations were 177 ng/mL (105 to 253.5 ng/mL) and 6.8 ng/mL (5.83 to 8.75 ng/mL). For a neonate weighing 3 kg and ingesting a mean volume of 500 mL/day of breast milk, these values would correspond to median neonatal emtricitabine and tenofovir doses of 126 mcg/day (range, 89 to 340 mcg/day) and 4.2 mcg/day (range, 3.4 to 7.1 mcg/day), respectively (Benaboud et al, 2011).
    8) LAMIVUDINE: Lamivudine is excreted into human milk, but in low concentrations (Johnson et al, 1999; Moodley et al, 1998a). In rat studies, concentrations of lamivudine in milk were slightly higher than in plasma (Prod Info Epivir(R), lamivudine, 2001).
    9) TENOFOVIR: Samples of human breast milk suggest that tenofovir is excreted into human breast milk at low levels. The impact on breastfed infants exposed to tenofovir is unknown. Tenofovir has also been reported in the milk of lactating rats (Prod Info VIREAD(R) oral tablets, powder, 2012; Prod Info STRIBILD(TM) oral tablets, 2012).
    10) STAVUDINE: It is not known whether stavudine is excreted in human milk. In animals studies stavudine was readily excreted into breast milk (Prod Info Zerit(R),, 2002).
    11) ZALCITABINE: It is not known whether zalcitabine is excreted in human milk (Prod Info Hivid(R), zalcitabine, 1996).
    12) ZIDOVUDINE: The mean concentration of zidovudine was similar in human milk and serum following single-dose administration (200 mg) to 13 HIV-infected women (Prod Info RETROVIR(R) oral tablets, capsules, syrup, 2010). However, the effects on the nursing infant have not been determined (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    13) Due to the risk of postnatal transmission of HIV, the Centers for Disease Control and Prevention does not recommend breastfeeding for HIV-infected mothers, including those who are receiving combination antiretroviral therapy or prophylaxis (Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission, 2012).
    C) ANIMAL STUDIES
    1) ABACAVIR
    a) Abacavir is excreted in the milk of lactating animals (Prod Info ZIAGEN(R) oral solution, oral tablets, 2008).
    2) DIDANOSINE
    a) Didanosine and its metabolites have been shown to be excreted in the milk of lactating animals following oral administration (Prod Info VIDEX(R) pediatric powder for oral solution, 2010).
    3) EMTRICITABINE
    a) In animal studies, no adverse effects were observed at emtricitabine lactation doses up to 60 times the recommended daily dose for humans (Prod Info ODEFSEY(R) oral tablets, 2016).
    4) STAVUDINE
    a) Stavudine is excreted into the milk of lactating animals (Prod Info ZERIT(R) oral capsules, solution, 2009)
    5) TENOFOVIR
    a) The drug is excreted into the milk of lactating animals (Prod Info VIREAD(R) oral tablets, powder, 2012; Prod Info STRIBILD(TM) oral tablets, 2012)
    6) TENOFOVIR ALAFENAMIDE
    a) In animal studies, no adverse effects were observed at tenofovir alafenamide lactation doses up to 14 times the recommended daily dose for humans (Prod Info ODEFSEY(R) oral tablets, 2016).
    3.20.5) FERTILITY
    A) ANIMAL STUDIES
    1) ABACAVIR
    a) Dosages that resulted in approximately 8 times the human exposure at the recommended dose (based on body surface area comparisons) had no adverse effects on the fertility of male and female animals (Prod Info ZIAGEN(R) oral solution, oral tablets, 2008).
    2) DELAVIRDINE
    a) Dosages up to 200 mg of delavirdine per kg of body weight per day did not impair fertility in male or female animals (Prod Info RESCRIPTOR(R) oral tablets, 2008).
    3) DIDANOSINE
    a) No evidence of impaired fertility has been found in animals receiving up to 14.2 times the estimated human dose of didanosine, based on plasma levels (Prod Info VIDEX(R) pediatric powder for oral solution, 2010).
    4) EMTRICITABINE
    a) In animal studies, fertility was unaffected at emtricitabine doses approximately 140 times the recommended daily dose for humans (Prod Info ODEFSEY(R) oral tablets, 2016).
    b) Normal fertility was seen in the offspring of animals exposed from before birth (in utero) through sexual maturity at daily exposures (AUC) of approximately 60-fold higher than human exposures at the recommended 200 mg daily dose (Prod Info EMTRIVA(R) oral capsule, solution, 2012).
    5) STAVUDINE
    a) No evidence of impaired fertility was seen in animals given stavudine at doses that resulted in peak serum concentrations that were up to 216 times those observed in humans who received a clinical dosage of 1 mg/kg/day (Prod Info ZERIT(R) oral capsules, solution, 2009).
    TENOFOVIR
    b) There were no effects on fertility, mating performance, or early embryonic development when tenofovir was administered at doses 10 times the human dose based on surface area comparisons to male animals for 28 days prior to mating and to female animals for 15 days prior to mating through day 7 of gestation (Prod Info VIREAD(R) oral tablets, powder, 2012). There was, however, an alteration of the estrous cycle in female animals (Prod Info TRUVADA(R) oral tablets, 2012).

    6) TENOFOVIR ALAFENAMIDE
    a) In animal studies, fertility was unaffected at tenofovir alafenamide doses approximately 62 times the recommended daily dose for humans (Prod Info ODEFSEY(R) oral tablets, 2016).

Carcinogenicity

    3.21.2) SUMMARY/HUMAN
    A) DIDANOSINE
    1) At the time of this review, no data were available to assess the carcinogenic potential of this agent.
    B) ZIDOVUDINE
    1) Extremely large doses have been associated with vaginal neoplasms in mice and rats; the significance in humans is not known.
    3.21.3) HUMAN STUDIES
    A) LACK OF INFORMATION
    1) DIDANOSINE
    a) At the time of this review, no data were available to assess the carcinogenic potential of this agent.
    B) NEOPLASM
    1) ZIDOVUDINE
    a) Doses far in excess of those administered to humans, have been associated with vaginal neoplasms in female mice and rats. The significance of these findings to humans is not known (Rachlis & Fanning, 1993).
    C) LYMPHOMA-LIKE DISORDER
    1) ZALCITABINE
    a) Thymic lymphomas have been induced in 3 of 50 mice administered zalcitabine 500 mg/kg/day for 13 weeks, and in 17 of 50 mice receiving 1000 mg/kg/day. This did not occur in mice from the vehicle control group (Sanders et al, 1995).
    3.21.4) ANIMAL STUDIES
    A) LIVER ADENOMAS
    1) TENOFOVIR
    a) In long-term carcinogenicity studies, increased incidence of liver adenomas were found in female mice at exposures of tenofovir 10 times those observed in humans at the therapeutic dose for HIV infection. In rats, there was no increase in tumor incidence at exposures up to 4 times that observed in humans at the therapeutic dose (Prod Info STRIBILD(TM) oral tablets, 2012).
    B) LACK OF EFFECT
    1) EMTRICITABINE
    a) No drug-related increases in tumor incidence were determined in long-term carcinogenicity studies of emtricitabine. Mice administered doses up to 750 mg/kg/day (23 times the human systemic exposure at the therapeutic dose of 200 mg/day) were not found to have drug-related increases in tumor incidence. Rats administered doses up to 60 mg/kg/day (28-fold the human systemic exposure at the therapeutic daily dose) were not found to have drug-related increases in tumor incidence (Prod Info STRIBILD(TM) oral tablets, 2012).

Genotoxicity

    A) DIDANOSINE
    1) Tests indicate that didanosine is not mutagenic at pharmacologic doses (Prod Info Videx(R), didanosine, 1996).
    B) EMTRICITABINE
    1) There was no evidence of genotoxicity in the reverse mutation bacterial test (Ames), or the mouse lymphoma or mouse micronucleus assays of emtricitabine(Prod Info STRIBILD(TM) oral tablets, 2012).
    C) TENOFOVIR
    1) There was no evidence of genotoxicity in the in vitro bacterial mutagenicity test (Ames) or the mouse micronucleus assay (for male mice only) of tenofovir. However, in the in vitro mouse lymphoma assay, tenofovir was mutagenic (Prod Info STRIBILD(TM) oral tablets, 2012).
    D) ZIDOVUDINE
    1) In vitro studies with zidovudine have demonstrated weak mutagenicity at high concentrations, and produced dose-related chromosomal abnormalities in human lymphocytes at concentrations of 3 mcg/mL and higher (Prod Info Retrovir(R), zidovudine, 1996).
    2) Zidovudine was negative in bacterial mutagenicity assay (Ayers, 1988).
    3) In mammalian cells, concentrations of 1000 to 5000 micrograms/mL were weakly mutagenic (Ayers, 1988).
    4) Aberrations in cultured human lymphocytes were seen at zidovudine concentrations of 3 micrograms/mL and higher (Ayers, 1988).
    5) Torres et al (2007) suggested that the mutagenicity produced by the nucleoside analogs zidovudine, abacavir, and lamivudine are driven by cumulative dose, and raises the concern of whether zidovudine-lamivudine has greater mutagenic effects than zidovudine alone in perinatally-exposed children (Torres et al, 2007).

Monitoring Parameters Levels

    4.1.1) SUMMARY
    A) Monitor serum electrolytes and hepatic enzymes.
    B) Monitor serum lipase in patients with abdominal pain or severe acidosis.
    C) Lactic acid concentration and serum pH should be monitored in acidotic patients.
    D) Cardiac failure, likely due to acidosis, has been reported; therefore, cardiac monitoring is recommended in the setting of acidosis or chest pain.
    E) Sources of infection should be sought in patients with neutropenia or significant acidosis.
    4.1.2) SERUM/BLOOD
    A) HEMATOLOGIC
    1) Complete blood counts (CBC's) should be monitored intensively in patients who overdose on nucleoside analogs, particularly neutrophil counts, platelets, WBC's, and hemoglobin.
    B) BLOOD/SERUM CHEMISTRY
    1) Serum electrolytes should be monitored in all patients who overdose on nucleoside analogs.
    2) Lactic acid concentration and serum pH should be monitored in acidotic patients.
    3) Serum potassium should be monitored in patients taking didanosine, particularly in those with pre-existing diarrhea or low baseline potassium (Katlama et al, 1991).
    4) These agents may cause hepatotoxicity. Monitor liver function tests in patients with significant exposures.
    5) These agents may infrequently cause kidney damage. Monitor serum creatinine and BUN in patients with significant exposures.
    6) Some of these agents (particularly didanosine and zalcitabine) have been shown to cause pancreatic injury. It may be advisable to monitor amylase and lipase levels in overdose cases.
    7) Monitor CPK, especially in patients with prolonged seizures.
    4.1.3) URINE
    A) OTHER
    1) Monitor urinary output in patients with significant exposures.
    2) Monitor urine myoglobin in patients with prolonged seizures.
    4.1.4) OTHER
    A) OTHER
    1) ECG
    a) Cardiac failure, likely due to acidosis, has been reported; therefore, cardiac monitoring is recommended in the setting of acidosis or chest pain.
    2) ELECTROPHYSIOLOGICAL TESTING
    a) Nerve conduction velocity tests may be advisable in patients exhibiting signs/symptoms of peripheral neuropathy.

Methods

    A) CHROMATOGRAPHY
    1) LAMIVUDINE: Harker et al (1994) described a high performance liquid chromatography (HPLC) technique for quantification of lamivudine in human serum. The process uses a serum sample size of 1 ml and is valid over a concentration range of 10-5000 ng/ml (Harker et al, 1994).
    2) Lamivudine was quantified in nonhuman primate plasma and CSF via a reverse-phase high-pressure liquid chromatography technique (Blaney et al, 1995).
    3) STAVUDINE: Janiszewski et al (1992) described a sensitive high-performance liquid chromatographic technique for the determination of stavudine in human plasma and urine (Janiszewski et al, 1992).
    B) IMMUNOASSAY
    1) Ferrua et al (1994) described a competitive ELISA method for assaying stavudine in human plasma. This method quantifies intracellular and unmetabolized drug in CEM and Molt 4 cell lines. Additionally, this method can be used for the indirect measurement of intracellular phosphorylated stavudine metabolites following reverse-phase HPLC separation (Ferrua et al, 1994).
    2) ZALCITABINE: A solid-phase extraction combined with radioimmunoassay (SPE-RIA) is described for zalcitabine that shows good linearity and precision for plasma concentrations of the drug observed clinically. A commercially available antiserum shows high specificity for the drug and its analogs and the test can be readily performed in laboratories (Roberts et al, 1994).
    3) Burger et al (1994) described a SPE-RIA procedure for quantifying zalcitabine in human plasma, urine and CSF (Burger et al, 1994).

Life Support

    A) Support respiratory and cardiovascular function.

Patient Disposition

    6.3.1) DISPOSITION/ORAL EXPOSURE
    6.3.1.1) ADMISSION CRITERIA/ORAL
    A) Patients with severe toxicity should be admitted. Patients with severe lactic acidosis, hepatic failure, or renal failure should be admitted to an intensive care setting.
    6.3.1.2) HOME CRITERIA/ORAL
    A) Suicidal patients and those with symptoms should be referred to a healthcare facility. Asymptomatic patients with inadvertent ingestion of NRTIs can be observed at home.
    6.3.1.3) CONSULT CRITERIA/ORAL
    A) Infectious disease should be consulted if a change to anti-retroviral therapy is indicated. Consult a medical toxicologist for patients with severe toxicity or in whom the diagnosis is not clear.
    6.3.1.5) OBSERVATION CRITERIA/ORAL
    A) Asymptomatic or mildly symptomatic patients should be observed for 4 to 6 hours, primarily monitoring signs of coingestant toxicity.

Monitoring

    A) Monitor serum electrolytes and hepatic enzymes.
    B) Monitor serum lipase in patients with abdominal pain or severe acidosis.
    C) Lactic acid concentration and serum pH should be monitored in acidotic patients.
    D) Cardiac failure, likely due to acidosis, has been reported; therefore, cardiac monitoring is recommended in the setting of acidosis or chest pain.
    E) Sources of infection should be sought in patients with neutropenia or significant acidosis.

Oral Exposure

    6.5.1) PREVENTION OF ABSORPTION/PREHOSPITAL
    A) No prehospital decontamination is indicated. Prehospital care should focus on assessment of vital signs and general supportive care.
    6.5.2) PREVENTION OF ABSORPTION
    A) ACTIVATED CHARCOAL
    1) CHARCOAL ADMINISTRATION
    a) Consider administration of activated charcoal after a potentially toxic ingestion (Chyka et al, 2005). Administer charcoal as an aqueous slurry; most effective when administered within one hour of ingestion.
    2) CHARCOAL DOSE
    a) Use a minimum of 240 milliliters of water per 30 grams charcoal (FDA, 1985). Optimum dose not established; usual dose is 25 to 100 grams in adults and adolescents; 25 to 50 grams in children aged 1 to 12 years (or 0.5 to 1 gram/kilogram body weight) ; and 0.5 to 1 gram/kilogram in infants up to 1 year old (Chyka et al, 2005).
    1) Routine use of a cathartic with activated charcoal is NOT recommended as there is no evidence that cathartics reduce drug absorption and cathartics are known to cause adverse effects such as nausea, vomiting, abdominal cramps, electrolyte imbalances and occasionally hypotension (None Listed, 2004).
    b) ADVERSE EFFECTS/CONTRAINDICATIONS
    1) Complications: emesis, aspiration (Chyka et al, 2005). Aspiration may be complicated by acute respiratory failure, ARDS, bronchiolitis obliterans or chronic lung disease (Golej et al, 2001; Graff et al, 2002; Pollack et al, 1981; Harris & Filandrinos, 1993; Elliot et al, 1989; Rau et al, 1988; Golej et al, 2001; Graff et al, 2002). Refer to the ACTIVATED CHARCOAL/TREATMENT management for further information.
    2) Contraindications: unprotected airway (increases risk/severity of aspiration) , nonfunctioning gastrointestinal tract, uncontrolled vomiting, and ingestion of most hydrocarbons (Chyka et al, 2005).
    6.5.3) TREATMENT
    A) SUPPORT
    1) Treatment is symptomatic and supportive. Seizures may occur and should be treated aggressively. Cardiac failure has been reported and cardiac monitoring is recommended. Hepatic failure may occur and liver function should be monitored. Peripheral neuropathies, which are generally reversible on drug withdrawal, may occur and should be treated with pain management as needed.
    B) SEIZURE
    1) SUMMARY
    a) Attempt initial control with a benzodiazepine (eg, diazepam, lorazepam). If seizures persist or recur, administer phenobarbital or propofol.
    b) Monitor for respiratory depression, hypotension, and dysrhythmias. Endotracheal intubation should be performed in patients with persistent seizures.
    c) Evaluate for hypoxia, electrolyte disturbances, and hypoglycemia (or, if immediate bedside glucose testing is not available, treat with intravenous dextrose).
    2) DIAZEPAM
    a) ADULT DOSE: Initially 5 to 10 mg IV, OR 0.15 mg/kg IV up to 10 mg per dose up to a rate of 5 mg/minute; may be repeated every 5 to 20 minutes as needed (Brophy et al, 2012; Prod Info diazepam IM, IV injection, 2008; Manno, 2003).
    b) PEDIATRIC DOSE: 0.1 to 0.5 mg/kg IV over 2 to 5 minutes; up to a maximum of 10 mg/dose. May repeat dose every 5 to 10 minutes as needed (Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008).
    c) Monitor for hypotension, respiratory depression, and the need for endotracheal intubation. Consider a second agent if seizures persist or recur after repeated doses of diazepam .
    3) NO INTRAVENOUS ACCESS
    a) DIAZEPAM may be given rectally or intramuscularly (Manno, 2003). RECTAL DOSE: CHILD: Greater than 12 years: 0.2 mg/kg; 6 to 11 years: 0.3 mg/kg; 2 to 5 years: 0.5 mg/kg (Brophy et al, 2012).
    b) MIDAZOLAM has been used intramuscularly and intranasally, particularly in children when intravenous access has not been established. ADULT DOSE: 0.2 mg/kg IM, up to a maximum dose of 10 mg (Brophy et al, 2012). PEDIATRIC DOSE: INTRAMUSCULAR: 0.2 mg/kg IM, up to a maximum dose of 7 mg (Chamberlain et al, 1997) OR 10 mg IM (weight greater than 40 kg); 5 mg IM (weight 13 to 40 kg); INTRANASAL: 0.2 to 0.5 mg/kg up to a maximum of 10 mg/dose (Loddenkemper & Goodkin, 2011; Brophy et al, 2012). BUCCAL midazolam, 10 mg, has been used in adolescents and older children (5-years-old or more) to control seizures when intravenous access was not established (Scott et al, 1999).
    4) LORAZEPAM
    a) MAXIMUM RATE: The rate of intravenous administration of lorazepam should not exceed 2 mg/min (Brophy et al, 2012; Prod Info lorazepam IM, IV injection, 2008).
    b) ADULT DOSE: 2 to 4 mg IV initially; repeat every 5 to 10 minutes as needed, if seizures persist (Manno, 2003; Brophy et al, 2012).
    c) PEDIATRIC DOSE: 0.05 to 0.1 mg/kg IV over 2 to 5 minutes, up to a maximum of 4 mg/dose; may repeat in 5 to 15 minutes as needed, if seizures continue (Brophy et al, 2012; Loddenkemper & Goodkin, 2011; Hegenbarth & American Academy of Pediatrics Committee on Drugs, 2008; Sreenath et al, 2009; Chin et al, 2008).
    5) PHENOBARBITAL
    a) ADULT LOADING DOSE: 20 mg/kg IV at an infusion rate of 50 to 100 mg/minute IV. An additional 5 to 10 mg/kg dose may be given 10 minutes after loading infusion if seizures persist or recur (Brophy et al, 2012).
    b) Patients receiving high doses will require endotracheal intubation and may require vasopressor support (Brophy et al, 2012).
    c) PEDIATRIC LOADING DOSE: 20 mg/kg may be given as single or divided application (2 mg/kg/minute in children weighing less than 40 kg up to 100 mg/min in children weighing greater than 40 kg). A plasma concentration of about 20 mg/L will be achieved by this dose (Loddenkemper & Goodkin, 2011).
    d) REPEAT PEDIATRIC DOSE: Repeat doses of 5 to 20 mg/kg may be given every 15 to 20 minutes if seizures persist, with cardiorespiratory monitoring (Loddenkemper & Goodkin, 2011).
    e) MONITOR: For hypotension, respiratory depression, and the need for endotracheal intubation (Loddenkemper & Goodkin, 2011; Manno, 2003).
    f) SERUM CONCENTRATION MONITORING: Monitor serum concentrations over the next 12 to 24 hours. Therapeutic serum concentrations of phenobarbital range from 10 to 40 mcg/mL, although the optimal plasma concentration for some individuals may vary outside this range (Hvidberg & Dam, 1976; Choonara & Rane, 1990; AMA Department of Drugs, 1992).
    6) OTHER AGENTS
    a) If seizures persist after phenobarbital, propofol or pentobarbital infusion, or neuromuscular paralysis with general anesthesia (isoflurane) and continuous EEG monitoring should be considered (Manno, 2003). Other anticonvulsants can be considered (eg, valproate sodium, levetiracetam, lacosamide, topiramate) if seizures persist or recur; however, there is very little data regarding their use in toxin induced seizures, controlled trials are not available to define the optimal dosage ranges for these agents in status epilepticus (Brophy et al, 2012):
    1) VALPROATE SODIUM: ADULT DOSE: An initial dose of 20 to 40 mg/kg IV, at a rate of 3 to 6 mg/kg/minute; may give an additional dose of 20 mg/kg 10 minutes after loading infusion. PEDIATRIC DOSE: 1.5 to 3 mg/kg/minute (Brophy et al, 2012).
    2) LEVETIRACETAM: ADULT DOSE: 1000 to 3000 mg IV, at a rate of 2 to 5 mg/kg/min IV. PEDIATRIC DOSE: 20 to 60 mg/kg IV (Brophy et al, 2012; Loddenkemper & Goodkin, 2011).
    3) LACOSAMIDE: ADULT DOSE: 200 to 400 mg IV; 200 mg IV over 15 minutes (Brophy et al, 2012). PEDIATRIC DOSE: In one study, median starting doses of 1.3 mg/kg/day and maintenance doses of 4.7 mg/kg/day were used in children 8 years and older (Loddenkemper & Goodkin, 2011).
    4) TOPIRAMATE: ADULT DOSE: 200 to 400 mg nasogastric/orally OR 300 to 1600 mg/day orally divided in 2 to 4 times daily (Brophy et al, 2012).
    C) MONITORING OF PATIENT
    1) Monitor serum electrolytes and hepatic enzymes.
    2) Monitor serum lipase in patients with abdominal pain or severe acidosis.
    3) Lactic acid concentration and serum pH should be monitored in acidotic patients.
    4) Cardiac failure, likely due to acidosis, has been reported; therefore, cardiac monitoring is recommended in the setting of acidosis or chest pain.
    5) Sources of infection should be sought in patients with neutropenia or significant acidosis.
    D) TRANSFUSION
    1) In the presence of bone marrow suppression, transfusions and protective measures for granulocytopenia may be needed until recovery.
    2) There is little data on the use of hematopoietic colony stimulating factors to treat neutropenia after drug overdose or idiosyncratic reactions. These agents have been shown to shorten the duration of severe neutropenia in patients receiving cancer chemotherapy (Hartman et al, 1997; Stull et al, 2005). They have also been used to treat agranulocytosis induced by nonchemotherapy drugs (Beauchesne & Shalansky, 1999). They may be considered in patients with severe neutropenia who have or are at significant risk for developing febrile neutropenia.
    a) Filgrastim: The usual starting dose in adults is 5 micrograms/kilogram/day by intravenous infusion or subcutaneous injection (Prod Info NEUPOGEN(R) injection, 2006).
    b) Sargramostim: Usual dose is 250 micrograms/square meter/day infused IV over 4 hours (Prod Info LEUKINE(R) injection, 2006).
    c) Monitor CBC with differential.
    E) ACIDOSIS
    1) Untreated patients with lactic acidosis may develop confusion, hypotension, coma, and circulatory collapse.
    2) METABOLIC ACIDOSIS: Treat severe metabolic acidosis (pH less than 7.1) with sodium bicarbonate, 1 to 2 mEq/kg is a reasonable starting dose(Kraut & Madias, 2010). Monitor serum electrolytes and arterial or venous blood gases to guide further therapy.
    3) Monitor serum sodium to avoid overload. Increase minute ventilation in intubated patients.
    4) RIBOFLAVIN: Fouty et al (1998) have proposed a riboflavin deficiency in AIDS patients taking these drugs and developing lactic acidosis and hepatic steatosis (Fouty et al, 1998). These authors have treated 3 patients with this syndrome with riboflavin 50 mg and reported return of serum lactate levels to normal. Other authors have reported resistant lactic acidosis which improved after treatment with riboflavin 50 mg/day orally or intravenously (Shiber, 2005; Luzzati et al, 1999).
    5) L-CARNITINE: Preliminary data from a pilot study of 6 patients suggested that L-carnitine may be helpful for patients with symptomatic nucleoside-analog-related lactic acidosis (Claessens et al, 2003). The dose used by these authors was L-carnitine 50 milligrams/kilogram/day as a 2 hour infusion 3 times/day in patients not receiving dialysis. For patients receiving continuous hemodiafiltration the dose was 100 mg/kg/day by continuous infusion.
    F) CONSULTATION
    1) HOTLINE: Health and Human Services (HHS) has launched the National Clinicians' Post-Exposure Prophylaxis Hotline (PEPline), a national toll-free hotline for assisting health care providers counsel and treat health care workers with job-related exposure to blood-borne diseases and infections, including hepatitis and HIV infection. The toll free number is: 1-888-448-4911 ((Anon, 1997)).

Enhanced Elimination

    A) HEMODIALYSIS
    1) Hemodialysis and whole bowel irrigation have no role in the management of NRTI overdose.

Summary

    A) TOXICITY: A full month supply of many of these agents has been ingested in overdose without clinical effects, though toxicity can occur at therapeutic doses with nucleoside reverse transcriptase inhibitors (NRTIs). LAMIVUDINE: No clinical signs or symptoms developed in an adult ingesting 6 grams of lamivudine. STAVUDINE: No acute toxicity was reported in patients treated with 12 to 24 times the recommended daily dosage. ZALCITABINE: Pediatric: Overdoses of 1.5 mg/kg have been reported; no sequelae developed.
    B) THERAPEUTIC DOSE: ABACAVIR: ADULT: 300 mg orally twice daily or 600 mg once daily. PEDIATRIC: 8 mg/kg orally twice daily. EMTRICITABINE: ADULT: 200 mg/day capsule, 240 mg/day oral solution. PEDIATRIC: 0 to 3 months of age: 3 mg/kg orally once daily; 3 months to 17 years of age: 6 mg/kg once daily oral solution, up to a maximum of 240 mg; children weighing more than 33 kg and can swallow whole capsule: 200 mg once daily. LAMIVUDINE: ADULT: 150 mg orally twice a day or 300 mg once daily. PEDIATRIC: 0 to 28-days-old: 2 mg/kg orally twice daily; 28 days or older: 4 mg/kg orally twice daily, maximum 150 mg twice daily. STAVUDINE: ADULT: less than 60 kg: 30 mg orally every 12 hours; 60 kg or more: 40 mg orally every 12 hours. PEDIATRIC: 0 to 13-days-old: 0.5 mg/kg/dose orally every 12 hours; 14-days-old and less than 30 kg: 1 mg/kg/dose orally every 12 hours ; 30 kg to less than 60 kg: 30 mg orally every 12 hours; 60 kg or more: 40 mg orally every 12 hours. TENOFOVIR: ADULT: 300 mg orally once daily. PEDIATRIC: at least 12-year-old and 35 kg or more: 300 mg orally once daily.

Therapeutic Dose

    7.2.1) ADULT
    A) SPECIFIC SUBSTANCE
    1) ABACAVIR
    a) 300 mg twice daily or 600 mg once daily in combination with other antiretroviral agents (Prod Info ZIAGEN(R) oral tablets, oral solution, 2015).
    2) ABACAVIR/LAMIVUDINE
    a) 1 tablet (abacavir sulfate 600 mg/lamivudine 300 mg) orally once daily in combination with other antiretroviral agents (Prod Info EPZICOM(R) oral tablets, 2015)
    3) ABACAVIR/DOLUTEGRAVIR/LAMIVUDINE
    a) The recommended dose is one fixed-dose tablet of abacavir 600 mg, dolutegravir 50 mg, and lamivudine 300 mg orally once daily (Prod Info TRIUMEQ(R) oral tablets, 2014)
    4) ADEFOVIR
    a) Type B viral hepatitis, chronic: 10 mg orally once daily (Prod Info HEPSERA(R) oral tablets, 2006).
    5) ELVITEGRAVIR/COBICISTAT/EMTRICITABINE/TENOFOVIR
    1) The recommended dose is 1 tablet (elvitegravir 150 mg/cobicistat 150 mg/emtricitabine 200 mg/tenofovir disoproxil fumarate 300 mg) orally once a day with food (Prod Info STRIBILD(TM) oral tablets, 2012).
    6) ELVITEGRAVIR/COBICISTAT/EMTRICITABINE/TENOFOVIR ALAFENAMIDE
    a) The recommended dose is 1 tablet (elvitegravir 150 mg/cobicistat 150 mg/emtricitabine 200 mg/tenofovir alafenamide 10 mg) orally once daily with food (Prod Info GENVOYA(R) oral tablets, 2015)
    7) EMTRICITABINE
    a) A recommended dose is a 200-mg capsule orally once daily, or 240 mg (24 mL) as an oral solution administered once daily (Prod Info EMTRIVA(R) capsules, oral solution, 2008).
    b) An oral dose of 200 mg once daily as monotherapy has been administered in HIV infection. In chronic hepatitis B, 100 to 300 mg once daily has been given ((Anon, 1999); p 773; Kahn et al, 1998; Murphy, 1999).
    8) EMTRICITABINE/RILPIVIRINE/TENOFOVIR DISOPROXIL FUMARATE
    a) Recommended adult dose is 1 tablet (emtricitabine 200 mg/rilpivirine 25 mg/tenofovir disoproxil fumarate 300 mg) taken orally once daily (Prod Info COMPLERA(R) oral tablets, 2016).
    9) EMTRICITABINE/TENOFOVIR ALAFENAMIDE
    a) HIV infection, 35 kg or more and CrCl of 30 mL/min or greater: Recommended dose is emtricitabine 200 mg/tenofovir alafenamide 25 mg (1 tablet) orally once daily (Prod Info DESCOVY(R) oral tablets, 2016).
    10) EMTRICITABINE/TENOFOVIR DISOPROXIL FUMARATE
    a) HIV prophylaxis: Emtricitabine 200 mg/tenofovir disoproxil fumarate 300 mg (1 tablet) orally once daily (Prod Info TRUVADA(R) oral tablets, 2016)
    b) HIV infection, 35 kg or more: Emtricitabine 200 mg/tenofovir disoproxil fumarate 300 mg (1 tablet) orally once daily, with or without food (Prod Info TRUVADA(R) oral tablets, 2016)
    11) LAMIVUDINE
    a) : The recommended dose is 150 mg orally twice daily or 300 mg orally once daily in combination with other antiretroviral agents for treatment of HIV (Prod Info EPIVIR(R) oral tablets, oral solution, 2015a).
    12) STAVUDINE
    a) 60 kg or more: 40 mg orally every 12 hours (Prod Info ZERIT(R) oral capsules, solution, 2009).
    b) Less than 60 kg: 30 mg orally every 12 hours (Prod Info ZERIT(R) oral capsules, solution, 2009).
    13) TENOFOVIR
    a) The recommended oral dose of tenofovir disoproxil fumarate for HIV-1 or chronic hepatitis B is 300 mg once daily with or without food (Prod Info VIREAD(R) oral tablets, oral powder, 2012).
    b) Intravenous tenofovir 1 to 3 mg/kg daily has been administered in HIV infection (Deeks et al, 1998).
    14) TENOFOVIR ALAFENAMIDE/EMTRICITABINE/RILPIVIRINE
    a) The recommended dose is 1 tablet (emtricitabine 200 mg/rilpivirine 25 mg/tenofovir alafenamide 25 mg) taken ORALLY once daily (Prod Info ODEFSEY(R) oral tablets, 2016).
    7.2.2) PEDIATRIC
    A) SPECIFIC SUBSTANCE
    1) ABACAVIR
    a) 3 MONTHS AND OLDER (ORAL SOLUTION): 8 mg/kg twice daily or 16 mg/kg once daily in combination with other antiretroviral agents; MAXIMUM 600 mg daily (Prod Info ZIAGEN(R) oral tablets, oral solution, 2015).
    b) PEDIATRIC PATIENTS WEIGHING 14 KG OR GREATER (ORAL TABLETS): If able to swallow tablets, dose according to weight: (Prod Info ZIAGEN(R) oral tablets, oral solution, 2015)
    1) 14 kg to less than 20 kg: 150 mg in the morning and 150 mg in the evening
    2) 20 kg to less than 25 kg: 150 mg in the morning and 300 mg in the evening
    3) 25 kg and greater: 300 mg in the morning and 300 mg in the evening
    2) ABACAVIR/LAMIVUDINE
    a) WEIGHING 25 KG OR MORE: 1 tablet (abacavir sulfate 600 mg/lamivudine 300 mg) orally once daily in combination with other antiretroviral agents (Prod Info EPZICOM(R) oral tablets, 2015).
    3) ABACAVIR/DOLUTEGRAVIR/LAMIVUDINE
    a) Safety and efficacy in the pediatric or adolescent population have not been established (Prod Info TRIUMEQ(R) oral tablets, 2014)
    4) ELVITEGRAVIR/COBICISTAT/EMTRICITABINE/TENOFOVIR
    a) YOUNGER THAN 18 YEARS OF AGE: Safety and efficacy have not been established in pediatric patients (Prod Info STRIBILD(TM) oral tablets, 2012).
    5) ELVITEGRAVIR/COBICISTAT/EMTRICITABINE/TENOFOVIR ALAFENAMIDE
    a) 12 YEARS OR OLDER, WEIGHING AT LEAST 35 KG: The recommended dose is 1 tablet (elvitegravir 150 mg/cobicistat 150 mg/emtricitabine 200 mg/tenofovir alafenamide 10 mg) orally once daily with food (Prod Info GENVOYA(R) oral tablets, 2015)
    b) LESS THAN 12 YEARS: Safety and effectiveness have not been established in patients less than 12 years of age (Prod Info GENVOYA(R) oral tablets, 2015).
    6) EMTRICITABINE
    a) 0 to 3 MONTHS OF AGE: 3 mg/kg orally once daily (oral solution) (Prod Info EMTRIVA(R) capsules, oral solution, 2008).
    b) 3 MONTHS TO 17 YEARS OF AGE: 6 mg/kg administered once daily via an oral solution, up to a MAXIMUM daily amount of 240 mg (24 mL) (Prod Info EMTRIVA(R) capsules, oral solution, 2008).
    c) WEIGHING MORE THAN 33 KG: Children who can swallow an intact capsule, 200 mg orally once daily (Prod Info EMTRIVA(R) capsules, oral solution, 2008).
    7) EMTRICITABINE/TENOFOVIR ALAFENAMIDE
    a) 12 YEARS AND OLDER, WEIGHING AT LEAST 35 KG AND CRCL OF 30 ML/MIN OR GREATER: Recommended dose is emtricitabine 200 mg/tenofovir alafenamide 25 mg (1 tablet) orally once daily (Prod Info DESCOVY(R) oral tablets, 2016).
    b) YOUNGER THAN 12 YEARS OF AGE OR WEIGHING LESS THAN 35 KG: Safety and efficacy have not been established (Prod Info DESCOVY(R) oral tablets, 2016).
    8) EMTRICITABINE/RILPIVIRINE/TENOFOVIR DISOPROXIL FUMARATE
    a) 12 YEARS AND OLDER AND WEIGHING AT LEAST 35 KG: Recommended dose is 1 tablet (emtricitabine 200 mg/rilpivirine 25 mg/tenofovir disoproxil fumarate 300 mg) taken orally once daily (Prod Info COMPLERA(R) oral tablets, 2016).
    b) YOUNGER THAN 12 YEARS OF AGE OR WEIGHING LESS THAN 35 KG: Safety and efficacy have not been established (Prod Info COMPLERA(R) oral tablets, 2016).
    9) EMTRICITABINE/TENOFOVIR DISOPROXIL FUMARATE
    a) HIV INFECTION, 12 YEARS AND OLDER WEIGHING 35 KG OR MORE: Emtricitabine 200 mg/tenofovir disoproxil fumarate 300 mg (1 tablet) orally once daily (Prod Info TRUVADA(R) oral tablets, 2016)
    b) HIV INFECTION, CHILDREN WEIGHING 17 TO 35 KG (Prod Info TRUVADA(R) oral tablets, 2016):
    1) 17 KG TO LESS THAN 22 KG: Emtricitabine 100 mg/tenofovir disoproxil fumarate 150 mg (1 tablet) orally once daily
    2) 22 KG TO LESS THAN 28 KG: Emtricitabine 133 mg/tenofovir disoproxil fumarate 200 mg (1 tablet) orally once daily
    3) 28 KG TO LESS THAN 35 KG: Emtricitabine 167 mg/tenofovir disoproxil fumarate 250 mg (1 tablet) orally once daily
    10) LAMIVUDINE
    a) CHRONIC HEPATITIS B INFECTION: 2 YEARS OF AGE AND OLDER: 3 mg/kg orally once daily; MAXIMUM 100 mg/day (Prod Info EPIVIR-HBV(R) oral tablets, solution, 2007; Jonas et al, 2002).
    b) HIV INFECTION:
    1) 29 DAYS TO 16 YEARS OF AGE: 4 mg/kg orally every 12 hours; MAXIMUM 150 mg/dose (Working Group on Antiretroviral Therapy and Medical Management of HIV-Infected Children, 2009; Prod Info EPIVIR(R) oral tablets, solution, 2009).
    2) GREATER THAN 16 YEARS OF AGE:
    a) LESS THAN 50 KG: 4 mg/kg orally every 12 hours; MAXIMUM 150 mg/dose (Working Group on Antiretroviral Therapy and Medical Management of HIV-Infected Children, 2009; Prod Info EPIVIR(R) oral tablets, solution, 2009).
    b) 50 KG OR GREATER: 150 mg orally twice daily for 300 mg orally once daily (Working Group on Antiretroviral Therapy and Medical Management of HIV-Infected Children, 2009; Prod Info EPIVIR(R) oral tablets, solution, 2009).
    c) CHILDREN 3 MONTHS AND OLDER (ORAL SOLUTION): According to the manufacturer, the recommended dosing, in HIV-1 infected pediatric patients, is 4 mg/kg twice daily OR 8 mg/kg once daily; MAX dose 300 mg daily (Prod Info EPIVIR(R) oral tablets, oral solution, 2015a)
    d) CHILDREN 3 MONTHS AND OLDER (ORAL TABLET) : According to the manufacturer, the recommended dosing in HIV-1 infected pediatric patients, based on the patient's weight, is as follows (Prod Info EPIVIR(R) oral tablets, oral solution, 2015a):
    1) 14 TO LESS THAN 20 KG: 75 mg twice daily
    2) 20 TO LESS THAN 25 KG: 75 mg in the morning and 150 mg in the evening
    3) 25 KG OR GREATER: 150 mg twice daily OR 300 mg once daily
    e) A dose of 2 mg/kg twice daily is recommended for . A dose of 4 mg/kg twice daily is recommended for infants 28 days of age and older (Tremoulet et al, 2007; AIDS Info, 2008; Moodley et al, 1998).
    11) STAVUDINE
    a) BIRTH TO 13 DAYS OLD: 0.5 mg/kg/dose administered orally every 12 hours (Prod Info ZERIT(R) oral capsules, solution, 2009).
    b) 14 DAYS OLD AND LESS THAN 30 KG: 1 mg/kg/dose administered orally every 12 hours (Prod Info ZERIT(R) oral capsules, solution, 2009).
    c) WEIGHING 30 KG TO LESS THAN 60 KG: 30 mg orally every 12 hours (Prod Info ZERIT(R) oral capsules, solution, 2009).
    d) WEIGHING GREATER THAN 60 KG: 40 mg orally every 12 hours (Prod Info ZERIT(R) oral capsules, solution, 2009).
    B) TENOFOVIR
    1) 2 YEARS TO 18 YEARS OF AGE FOR THE TREATMENT OF HIV-1:
    a) TABLETS: The recommended dose is one tablet based on body weight (150, 200, 250, or 300 mg) once daily without regard for food, in patients that weigh at least 17 kg and are able to swallow intact tablets. MAXIMUM DOSE: 300 mg (Prod Info VIREAD(R) oral tablets, powder, 2012).
    b) ORAL POWDER: The recommended dose is 8 mg/kg once daily with food. MAXIMUM DOSE: 300 mg (Prod Info VIREAD(R) oral tablets, powder, 2012).
    2) 12 YEARS OR OLDER (WEIGHING 35 KG OR MORE) FOR THE TREATMENT OF CHRONIC HEPATITIS B:
    a) TABLETS: The recommended dose is 300 mg ORALLY once daily (Prod Info VIREAD(R) oral tablets, oral powder, 2012).
    C) TENOFOVIR ALAFENAMIDE/EMTRICITABINE/RILPIVIRINE
    1) 12 YEARS AND OLDER WEIGHING AT LEAST 35 KG: The recommended dose is 1 tablet (emtricitabine 200 mg/rilpivirine 25 mg/tenofovir alafenamide 25 mg) taken ORALLY once daily (Prod Info ODEFSEY(R) oral tablets, 2016).
    2) LESS THAN 12 YEARS OF AGE OR WEIGHING LESS THAN 35 KG: Safety and efficacy have not been established (Prod Info ODEFSEY(R) oral tablets, 2016) .

Maximum Tolerated Exposure

    A) SPECIFIC SUBSTANCE
    1) LAMIVUDINE
    a) NO TOXICITY/MILD TOXICITY
    1) No clinical signs or symptoms were reported in an adult ingesting 6 grams of lamivudine. Hematologic tests remained normal (Prod Info EPIVIR-HBV(R) oral tablets, oral solution, 2004).
    2) CASE REPORT: A 17-year-old woman ingested 3.9 g lamivudine and 15.6 g efavirenz. She developed mild agitation and somnolence, described feeling "high, alienated with artificial visual impressions" without hallucinations, had a clumsy gait, and developed abdominal pain and vomiting. The following day she had diarrhea, abdominal cramps and a diffuse pruritic maculopapular rash. Wild bizarre dreams persisted for 5 days. The neurologic effects were likely secondary to the efavirenz (Boscacci et al, 2006).
    2) STAVUDINE
    a) NO TOXICITY/MILD TOXICITY
    1) No acute toxicity was reported in patients treated with 12 to 24 times the recommended daily dosage (Prod Info ZERIT(R) oral capsules, oral solution, 2006).
    3) ZALCITABINE
    a) CHRONIC OVERDOSE
    1) Peripheral neuropathy occurred in all patients receiving 6 times the recommended daily dosage by week 10. A patient receiving 25 times the recommended daily dosage experienced rash and fever after 1 and 1/2 weeks necessitating discontinuation of the drug (Prod Info HIVID(R) oral tablets, 2002).

Pharmacologic Mechanism

    A) The major mechanism of action of the nucleoside antivirals is as competitive inhibitors of the RNA/DNA polymerase reverse transcriptase of HIV, causing chain termination in the growing viral DNA chain (Moyle, 2000a; Jeffries, 1989; Yarchoan et al, 1989). To be pharmacologically effective, these agents must first be phosphorylated to an active 5'-triphosphate moiety via cellular kinases of the host. Nucleosides which lack the 5'-triphosphate group are not substrates for DNA polymerase (Broder, 1990).
    B) Resistance generally occurs within 6 to 12 months and is a result of one mutation in the nucleotide chain (Larder, 1995).
    1) Cross-resistance exists between zalcitabine, didanosine, and lamivudine at codon 184, and also occurs at codon 65 or 74 between zalcitabine and didanosine. Mutations at codon 75 of the stavudine resistant strain confers cross-resistance to zalcitabine and didanosine (Larder, 1995). Cross-resistance between protease inhibitors and reverse transcriptase inhibitors is unlikely since different targeted enzymes are involved (Prod Info Invirase(R), 1996; Prod Info Crixivan(R), 1996; Prod Info Norvir(R), 1996).
    2) Cross-resistance has not been observed between zidovudine and nucleoside analogues lacking the 3'-azido moiety, including zalcitabine, didanosine, lamivudine, stavudine, and non-nucleoside reverse transcriptase inhibitors (i.e., nevirapine, delavirdine) (Larder et al, 1990) Richman, 1990; (Larder, 1995).
    C) IN VITRO TESTING: The utility of in vitro resistance testing and its interpretation are difficult, considering the extent of in vitro findings translating into in vivo situations is unknown. Phosphorylation of nucleoside analogues differs between cell lines as well as between the mitotic phases of those cells; a drug demonstrating toxicity in a cell line where it is efficiently phosphorylated may be significantly less toxic in a different cell line. To date, none of the in vitro studies have tested all the available nucleoside analogues using the same assay system and across multiple cell lines. Drugs have been evaluated on an individual basis rather than in the combinations in which they are used in clinical practice (Moyle, 2000).

Toxicologic Mechanism

    A) It is thought that the neurotoxicity of these agents is related to inhibition of mitochondrial DNA polymerase which alters mitochondrial function in nerves (Lipsky, 1993; Sandberg et al, 1995). Mitochondrial DNA synthesis inhibition may also be responsible for lactic acidosis, hepatic steatosis, myopathy, and peripheral neuropathy (Moyle, 2000a; Fouty et al, 1998).
    1) However, familial mitochondrial diseases do not appear to have identical presentations to nucleoside analogue toxicities. For instance, stavudine and, to a lesser extent, zidovudine are poor substrates for mitochondrial thymidine kinase type 2, the major form in cells that is not actively mitotic such as neurons, myocytes and adipocytes. These are the cell types where proposed mitochondrial toxicities of neuropathy, myopathy and lipoatrophy are observed (Moyle, 2000).
    B) Fouty et al (1998) have proposed that lactic acidosis and hepatic steatosis may result from impaired mitochondrial DNA replication due to nucleoside reverse transcriptase inhibitors (NRTI)-mediated inhibition of DNA polymerase gamma, which may be combined with decreased riboflavin activity due to dietary deficiency, drug-induced inhibition of flavokinase, or both. Thus, riboflavin is proposed as a therapy for lactic acidosis in these cases (Fouty et al, 1998).
    1) Inhibition of DNA polymerase-gamma, an essential enzyme for the replication of mitochondrial DNA (mtDNA), results in depletion of mtDNA, successively leading to depletion of mtDNA-encoded proteins and to mitochondrial dysfunction. Lipodystrophy as well as lactic acidosis and hepatic steatosis are believed to be induced via this mechanism (Brinkman, 2000; Carr et al, 2000; ter Hofstede et al, 2000).
    2) Additionally, Walker et al (2000) suggest that NRTI therapy may also interfere with mitochondrial RNA formation. No repair mechanisms have been established for mitochondrial RNA, unlike DNA. Thus, NRTIs may be excised from mitochondrial DNA but remain incorporated in RNA strands, prematurely terminating synthesis of mitochondrial messenger and transfer RNA (Walker et al, 2000).

Physical Characteristics

    A) ABACAVIR SULFATE is a white to off-white solid that is soluble in water (Prod Info ZIAGEN(R) oral tablets oral solution, 2015).
    B) ADEFOVIR DIPIVOXIL is a white to off-white crystalline powder with an aqueous solubility of 19 mg/mL at pH 2 and 0.4 mg/mL at pH 7.2. The octanol/aqueous phosphate buffer (pH 7) partition coefficient (log p) is 1.91 (Prod Info HEPSERA(R) oral tablets, 2012).
    C) EMTRICITABINE is a white to off-white powder with a solubility of approximately 112 mg/mL in water at 25 degrees C (Prod Info GENVOYA(R) oral tablets, 2015).
    D) LAMIVUDINE is a white to off-white crystalline solid with a solubility of approximately 70 mg/mL in water at 20 degrees C (Prod Info EPIVIR(R) oral tablets, oral solution, 2015).
    E) STAVUDINE is a white to off-white crystalline powder with a solubility at 23 degrees C of approximately 83 mg/mL in water and 30 mg/mL in propylene glycol. The n-octanol/water partition coefficient at 23 degrees C is 0.144 (Prod Info stavudine oral capsules, 2015).
    F) TENOFOVIR ALAFENAMIDE FUMARATE is a white to off-white or tan powder with a solubility of 4.7 mg/mL in water at 20 degrees C (Prod Info ODEFSEY(R) oral tablets, 2016).
    G) TENOFOVIR DISOPROXIL FUMARATE is a white to off-white crystalline powder with a solubility of 13.4 mg/mL in distilled water at 25 degrees C. The octanol/phosphate buffer (pH 6.5) partition coefficient (log p) is 1.25 at 25 degrees C (Prod Info VIREAD(R) oral tablets, oral powder, 2016).

Molecular Weight

    A) ABACAVIR SULFATE: 670.76 g/mol (Prod Info ZIAGEN(R) oral tablets oral solution, 2015)
    B) ADEFOVIR DIPIVOXIL: 501.48 (Prod Info HEPSERA(R) oral tablets, 2012)
    C) EMTRICITABINE: 247.24 (Prod Info GENVOYA(R) oral tablets, 2015)
    D) LAMIVUDINE: 229.3 g/mol (Prod Info EPIVIR(R) oral tablets, oral solution, 2015)
    E) STAVUDINE: 224.21 (Prod Info stavudine oral capsules, 2015)
    F) TENOFOVIR ALAFENAMIDE FUMARATE: 534.5 (Prod Info ODEFSEY(R) oral tablets, 2016)
    G) TENOFOVIR DISOPROXIL FUMARATE: 635.52 (Prod Info VIREAD(R) oral tablets, oral powder, 2016)

General Bibliography

    1) AIDS Info: Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection. AIDS Info. Rockville, MD. 2008. Available from URL: http://aidsinfo.nih.gov/contentfiles/PediatricGuidelines.pdf.
    2) AMA Department of DrugsAMA Department of Drugs: AMA Evaluations Subscription, American Medical Association, Chicago, IL, 1992.
    3) Agarwala R , Mohan S , Herlitz LC , et al: The case: 41-year-old HIV patient with proteinuria and progressive renal dysfunction. Tenofovir toxicity. Kidney Int 2010; 77(5):475-476.
    4) Anon: "Compassionate" release of DDI. Lancet 1989; 2:1079-1080.
    5) Anon: FDA Talk Paper: FDA/Bristol Myers Squibb issues caution for HIV combination therapy with Zerit and Videx in pregnant women. U.S. Food and Drug Administration. Rockville, MD, USA. 2001. Available from URL: http://www.fda.gov/bbs/topics/ANSWERS/ANS01063.html. As accessed Accessed January 8, 2001.
    6) Anon: HHS launches national toll-free hotline for clinicians reporting exposure to blood-borne pathogens. United States Department of Health and Human Services. Washington, DC, USA. 1997. Available from URL: http://www.dhhs.gov. As accessed Accessed December 5, 1997.
    7) Anon: Important drug warning: Fatal hypersensitivity reactions, respiratory symptoms, and Ziagen(R) (abacavir sulfate), Glaxo Wellcome, Inc, Research Triangle Park, NC, 2000.
    8) Anon: Triangle Pharmaceuticals reports data from expanded study of anti-HBV activity of Coviracil(TM) (emtricitabine). PR Newswire United Business Media. New York, NY, USA. 1999. Available from URL: http://www.prnewswire.com.
    9) Aponte-Cipriani SL, Teplitz C, & Yancovitz S: Pancreatitis possibly related to 2',3'-dideoxycytidine (letter). Ann Int Med 1993; 119:539-540.
    10) Beauchesne MF & Shalansky SJ: Nonchemotherapy drug-induced agranulocytosis: a review of 118 patients treated with colony-stimulating factors. Pharmacotherapy 1999; 19(3):299-305.
    11) Benaboud S, Pruvost A, Coffie PA, et al: Concentrations of tenofovir and emtricitabine in breast milk of HIV-1-infected women in Abidjan, Cote d'Ivoire, in the ANRS 12109 TEmAA Study, Step 2. Antimicrob Agents Chemother 2011; 55(3):1315-1317.
    12) Berger AR, Arezzo JC, & Schaumburg HH: 2',3'-dideoxycytidine (ddC) toxic neuropathy: a study of 52 patients. Neurology 1993; 43:358-362.
    13) Blanche S, Tardiu M, Rustin P, et al: Persistent mitochondrial dysfunction and perinatal exposure to antiretroviral nucleoside analogues.. Lancet 1999; 354:1084-9.
    14) Blaney SM, Daniel MJ, & Harker AJ: Pharmacokinetics of lamivudine and BCH-189 in plasma and cerebrospinal fluid of nonhuman primates. Antimicrob Agents Chemo 1995; 39:2779-2782.
    15) Bleeker-Rovers CP, Kadir SW, & van Leusen R: Hepatic steatosis and lactic acidosis caused by stavudine in an HIV-infected patient. Netherlands J Med 2000; 57:190-193.
    16) Boscacci RT, Buclin T, Furrer H, et al: A case of voluntary intoxication with efavirenz and lamivudine. AIDS (London, England) 2006; 20(9):1352-1354.
    17) Brinkman K: Editorial response: hyperlactatemia and hepatic steatosis as features of mitochondrial toxicity of nucleoside analogue reverse transcriptase inhibitors. Clin Infect Dis 2000; 31:167-169.
    18) Brivet FG, Nion I, & Megarbane B: Fatal lactic acidosis and liver steatosis associated with didanosine and stavudine treatment: a respiratory chain dysfunction? (letter). J Hepatol 2000; 32:364-365.
    19) Broder S: Pharmacodynamics of 2',3'-dideoxycytidine: an inhibitor of human immunodeficiency virus. Am J Med 1990; 88(Suppl5B):2S-7S.
    20) Brophy GM, Bell R, Claassen J, et al: Guidelines for the evaluation and management of status epilepticus. Neurocrit Care 2012; 17(1):3-23.
    21) Browne MJ, Mayer KH, & Chafee SBD: 2',3'-Didehydro-2',3'-dideoxythymidine and (d4T) in patients with AIDS or AIDS-related complex: phase I trial. J Infect Dis 1993; 167:21-29.
    22) Burger DM, Rosing H, & Napel CHH: Application of a radioimmunoassay for determination of levels of zalcitabine (ddC) in human plasma, urine, and cerebrospinal fluid. Antimicrob Agents & Chemo 1994; 38:2763-2767.
    23) Carey P: Peripheral neuropathy: zalcitabine reassessed (review). Intl J STD AIDS 2000; 11:417-423.
    24) Carr A & Cooper DA: Adverse effects of antiretroviral therapy. Lancet 2000; 356:1423-1430.
    25) Carr A, Miller J, & Law M: A syndrome of lipoatrophy, lactic acidaemia and liver dysfunction associated with HIV nucleoside analogue therapy: contribution to protease inhibitor-related lipodystrophy syndrome. AIDS 2000; 14:F25-F32.
    26) Carr A, Morey AM, & Mallon P: Fatal portal hypertension, liver failure, and mitochondrial dysfunction after HIV-1 nucleoside analogue-induced hepatitis and lactic acidaemia. Lancet 2001; 357:1412-1414.
    27) Chamberlain JM, Altieri MA, & Futterman C: A prospective, randomized study comparing intramuscular midazolam with intravenous diazepam for the treatment of seizures in children. Ped Emerg Care 1997; 13:92-94.
    28) Chin RF , Neville BG , Peckham C , et al: Treatment of community-onset, childhood convulsive status epilepticus: a prospective, population-based study. Lancet Neurol 2008; 7(8):696-703.
    29) Choonara IA & Rane A: Therapeutic drug monitoring of anticonvulsants state of the art. Clin Pharmacokinet 1990; 18:318-328.
    30) Chyka PA, Seger D, Krenzelok EP, et al: Position paper: Single-dose activated charcoal. Clin Toxicol (Phila) 2005; 43(2):61-87.
    31) Claessens YE, Cariou A, Monchi M, et al: Detecting life-threatening lactic acidosis related to nucleoside-analog treatment of human immunodeficiency virus-infected patients, and treatment with L-carnitine. Crit Care Med 2003; 31:1042-1047.
    32) Clark SJ, Creighton S, & Portmann B: Acute liver failure associated with antiretroviral treatment for HIV: a report of six cases. J Hepatol 2002; 36:295-301.
    33) Coghlan ME, Sommadossi JP, & Jhala NC: Symptomatic lactic acidosis in hospitalized antiretroviral-treated patients with human immunodeficiency virus infection: a report of 12 cases. Clin Infect Dis 2001; 33:1914-1921.
    34) Comereski CR, Kelly WA, & Davidson TJ: Acute cardiotoxicity of nucleoside analogs FddA and Fddl in rats. Fund Appl Toxicol 1993; 20:360-364.
    35) Cooley TP, Kunches LM, & Saunders CA: Once-daily administration of 2',3'-dideoxyinosine (ddI) in patients with acquired immunodeficiency syndrome or AIDS-related complex: results of phase I trial. New Engl J Med 1990; 322:1340-1345.
    36) Cundy KC, Safrin S, Coleman R et al: Oral PMPA prodrug: relationship between clinical pharmacokinetics, safety and anti-HIV activity (abstract). 12th World AIDS Conference; June 28, 1998; 53, June 28, 1998.
    37) Cupler EJ & Dalakas MC: Exacerbation of peripheral neuropathy by lamivudine (letter). Lancet 1995; 345:460-461.
    38) Deeks SG, Barditch-Crovo P, & Lietman PS: Safety, pharmacokinetics, and antiretroviral activity of intravenous 9-(2-(R)-(phosphonomethoxy)propyl)adenine, a novel anti-human immunodeficiency virus (HIV) therapy, in HIV-infected adults. Antimicrob Agents Chemother 1998; 42(9):2380-2384.
    39) Deeks SG, Barditch-Crovo P, Lietman PS, et al: Safety, pharmacokinetics, and antiretroviral activity of intravenous 9-(2-(R)-(phosphonomethoxy)propyl)adenine, a novel anti-human immunodeficiency virus (HIV) therapy, in HIV-infected adults. Antimicrob Agents Chemother 1998a; 42(9):2380-2384.
    40) Dobkin JF: Abacavir enters the clinic. Infect Med 1999; 16:7.
    41) Donzanti BA, Kelley JA, & Tomaszewski JE: Acute cardiotoxicity of the anti-HIV dideoxynucleoside, F-ddA, in the rat. Fund Appl Toxicol 1995; 27:167-176.
    42) Dudley MN, Graham KK, & Kaul S: Pharmacokinetics of stavudine in patients with AIDS or AIDS-related complex. J Infect Dis 1992; 166:480-485.
    43) Dunkle LA, Cross R, & Gugliotti R: Dose-escalating study of safety and efficacy of dideoxy-didehydrothymidine (D4T) for HIV infection. Antiviral Res 1990; 1(Suppl):116.
    44) Elliot CG, Colby TV, & Kelly TM: Charcoal lung. Bronchiolitis obliterans after aspiration of activated charcoal. Chest 1989; 96:672-674.
    45) FDA: Poison treatment drug product for over-the-counter human use; tentative final monograph. FDA: Fed Register 1985; 50:2244-2262.
    46) Falco V, Rodriguez D, & Ribera E: Severe nucleoside-associated lactic acidosis in human immunodeficiency virus-infected patients: report of 12 cases and review of the literature. Clin Infect Dis 2002; 34:838-846.
    47) Falco V, Rodriguez D, Ribera E, et al: Severe nucleoside-associated lactic acidosis in human immunodeficiency virus-infected patients: report of 12 cases and review of the literature. Clin Infect Dis 2002a; 34:838-846.
    48) Ferrua B, Tran TT, & Quaranta JF: Measurement of the anti-HIV agent 2',3'-didehydro-2',3'-dideoxythymidine (D4T) by competitive ELISA. J Immunolog Methods 1994; 176:103-110.
    49) Fichtenbaum CJ, Clifford DB, & Powderly WG: Risk factors for dideoxynucleoside- induced toxic neuropathy in patients with the human immunodeficiency virus infection. J Acquir Immun Defic Syn Hum Retrovirol 1995; 10:169-174.
    50) Flynn PM , Mirochnick M , Shapiro DE , et al: Pharmacokinetics and safety of single-dose tenofovir disoproxil fumarate and emtricitabine in HIV-1-infected pregnant women and their infants. Antimicrob Agents Chemother 2011; 55(12):5914-5922.
    51) Fong IW: Hair loss associated with lamivudine (letter). Lancet 1994; 344:1702.
    52) Foster RH & Faulds D: Abacavir. Drugs 1998; 55:729-736.
    53) Fouty B, Frerman F, & Reves R: Riboflavin to treat nucleoside analogue-induced lactic acidosis. Lancet 1998; 352:291-292.
    54) Gilden D: T-20 and adefovir for salvage therapy - expect no miracles. GMHC Treat Issues 1998; 12:6-7.
    55) Golej J, Boigner H, Burda G, et al: Severe respiratory failure following charcoal application in a toddler. Resuscitation 2001; 49:315-318.
    56) Graff GR, Stark J, & Berkenbosch JW: Chronic lung disease after activated charcoal aspiration. Pediatrics 2002; 109:959-961.
    57) Gustavson LE, Fukuda EK, & Rubio FA: A pilot study of the bioavailability and pharmacokinetics of 2',3'-dideoxycytidine in patients with AIDS or AIDS- related complex. J Acquir Immune Defic Syndr 1990; 3:28-31.
    58) Harker AJ, Evans GL, & Hawley AE: High-performance liquid chromatographic assay for 2'-deoxy-3'-thiacytidine in human serum. J Chromato 1994; 657:227-232.
    59) Harris CR & Filandrinos D: Accidental administration of activated charcoal into the lung: aspiration by proxy. Ann Emerg Med 1993; 22:1470-1473.
    60) Hartman LC, Tschetter LK, Habermann TM, et al: Granulocyte colony-stimulating factor in severe chemotherapy-induced afebrile neutropenia.. N Engl J Med 1997; 336:1776-1780.
    61) Hegenbarth MA & American Academy of Pediatrics Committee on Drugs: Preparing for pediatric emergencies: drugs to consider. Pediatrics 2008; 121(2):433-443.
    62) Henry K, Acosta EP, & Jochimsen E: Hepatotoxicity and rash associated with zidovudine and zalcitabine chemoprophylaxis (letter). Ann Int Med 1996; 124:855.
    63) Hill JB, Sheffield JS, & Zeeman GG: Hepatotoxicity with antiretroviral treatment of pregnant women. Obstet Gynecol 2001; 98:909-911.
    64) Ho HT & Hitchcock MJM: Cellular pharmacology of 2',3'-dideoxy-2',3'- didehydrothymidine, a nucleoside analog active against human immunodeficiency virus. Antimicrob Agents Chemother 1989; 33:844-849.
    65) Honkoop P, de Man RA, & Heijtink RA: Hepatitis B reactivation after lamivudine (letter). Lancet 1995; 346:1156-1157.
    66) Horton CM, Dudley MN, & Kaul S: Population pharmacokinetics of stavudine (d4T) in patients with AIDS or advanced AIDS-related complex. Antimicrob Agent Chemo 1995; 39:2309-2315.
    67) Hvidberg EF & Dam M: Clinical pharmacokinetics of anticonvulsants. Clin Pharmacokinet 1976; 1:161.
    68) Indorf AS & Pegram PS: Esophageal ulceration related to zalcitabine (ddC). Ann Int Med 1992; 117:133-134.
    69) JEF Reynolds : Martindale: The Extra Pharmacopoeia (electronic version). The Pharmaceutical Press. London, UK (Internet Version). Edition expires 2000; provided by Truven Health Analytics Inc., Greenwood Village, CO.
    70) Janiszewski JS, Mulvana DE, Kaul S, et al: High-performance liquid chromatographic determination of 2',3'-didehydro-3'-deoxythymidine, a new anti-human immunodeficiency virus agent, in human plasma and urine. J Chromatogr 1992; 577:151-156.
    71) Jeffries DJ: The antiviral activity of dideoxycytidine. J Antimicrob Chemother 1989; 23(Suppl A):29-34.
    72) Johnson M, Moore K, Yuen G, et al: Clinical pharmacokinetics of lamivudine. Clin Pharmacokinet 1999; 36(1):41-66.
    73) Jonas MM, Mizerski J, Badia IB, et al: Clinical trial of lamivudine in children with chronic hepatitis B. N Engl J Med 2002; 346(22):1706-1713.
    74) Kahn J, Thompson M, & Mildvan D: Selection of FTC dose based on viral kinetics and pharmacokinetics in an accelerated clinical trial design (abstract), 12th World AIDS Conference, Geneva, Switzerland, 1998.
    75) Katlama C, Tubiana R, & Rosenheim M: Dideoxyinosine-associated hypokalaemia (letter). Lancet 1991; 337:183.
    76) Khawcharoenporn T, Shikuma CM, & Williams AE: Lamivudine-associated macrocytosis in HIV-infected patients. Int J STD AIDS 2007; 18(1):39-40.
    77) Klecker RW, Collins JM, & Yarchoan RC: Pharmacokinetics of 2',3'-dideoxycytidine in patients with AIDS and related disorders. J Clin Pharmacol 1988; 28:837-842.
    78) Kline MW, Dunkle LM, Church JA, et al: A phase I/II evaluation of stavudine (d4T) in children with human immunodeficiency virus infection. Pediatrics 1995; 96:247-252.
    79) Kraut JA & Madias NE: Metabolic acidosis: pathophysiology, diagnosis and management. Nat Rev Nephrol 2010; 6(5):274-285.
    80) Lambert JS, Siedlin M, & Reichman RC: 2',3'-dideoxyinosine (ddI) in patients with the acquired immunodeficiency syndrome or AIDS-related complex: a phase I trial. New Engl J Med 1990; 322:1333-1340.
    81) Larder BA, Chesebro B, & Richman DD: Susceptibilities of zidovudine- susceptible and -resistant human immunodeficiency virus isolates to antiviral agents determined by using a quantitative plaque reduction assay. Antimicrob Agents Chemother 1990; 34:436-441.
    82) Larder BA: Viral resistance and the selection of antiretroviral combinations. J AIDS 1995; 10(Suppl 1):S28-S33.
    83) Lewis LL, Venzon D, Church J, et al: Lamivudine in children with human immunodeficiency virus infection: a phase I/II study. J Infect Dis 1996; 174:16-25.
    84) Lipsky J: Zalcitabine and didanosine. Drug profiles. Lancet 1993; 341:30-32.
    85) Loddenkemper T & Goodkin HP: Treatment of Pediatric Status Epilepticus. Curr Treat Options Neurol 2011; Epub:Epub.
    86) Lonergan JT, Behling C, & Pfander H: Hyperlactatemia and hepatic abnormalities in 10 human immunodeficiency virus-infected patients receiving nucleoside analogue combination regimens. Clin Infect Dis 2000; 31:162-166.
    87) Luther J & Glesby MJ: Dermatologic adverse effects of antiretroviral therapy: recognition and management. Am J Clin Dermatol 2007; 8(4):221-233.
    88) Luzhansky JZ, Pierce AB, & Hoy JF: Leber's hereditary optic neuropathy in the setting of nucleoside analogue toxicity. AIDS 2001; 15:1588-1589.
    89) Luzzati R, Bravo PD, & Perri GD: Riboflavine and severe lactic acidosis (letter). Lancet 1999; 353:901-902.
    90) Manno EM: New management strategies in the treatment of status epilepticus. Mayo Clin Proc 2003; 78(4):508-518.
    91) Martin JC, Hitchcock JM, & Fridland A: Comparative studies of 2',3'- didehydro-2',3'-dideoxythymidine (D4T) with other pyrimidine nucleoside analogues. Ann Acad Sci 1990; 616:22-28.
    92) Martinez OP & French MAH: Acoustic neuropathy associated with zalcitabine- induced peripheral neuropathy. AIDS 1993; 7:901-902.
    93) McNeely MC, Yarchoan R, & Broder S: Dermatologic complications associated with administration of 2',3'-dideoxycytidine in patients with human immunodeficiency virus infection. J Am Acad Dermatol 1989; 21:1213-1217.
    94) Megarbane B, Brivet F, & Baud F: Severe lactic acidosis with multiorgan failure secondary to antiretroviral therapy (abstract). J Toxicol-Clin Toxicol 2001; 39:243.
    95) Merigan TC & Skowron G: Study Group of the AIDS Clinical Trials Group of the National Institute of Allergy and Infectious Diseases: Safety and tolerance of dideoxycytidine as a single agent: results of early-phase studies in patients with acquired immunodeficiency syndrome (AIDS) or advanced AIDS-related complex. Am J Med 1990 1990; 88 (Suppl 5B):11S-15S.
    96) Merigan TC, Skowron G, & Bozzette SA: Circulating p24 antigen levels and responses to dideoxycytidine in human immunodeficiency virus (HIV) infections: a phase I and II study. Ann Inter Med 1989; 110:189-194.
    97) Miller KD, Cameron M, & Wood LV: Lactic acidosis and hepatic steatosis associated with use of stavudine: report of four cases. Ann Intern Med 2000; 133:192-196.
    98) Mokrzycki MH, Harris C, & May H: Lactic acidosis associated with stavudine administration: a report of five cases. Clin Infect Dis 2000; 30:198-200.
    99) Moodley J, Moddley D, Pillay K, et al: Pharmacokinetics and antiretroviral activity of lamivudine alone or when coadministered with zidovudine in human immunodeficiency virus type 1-infected pregnant women and their offspring. J Infect Dis 1998; 178:1327-1333.
    100) Moodley J, Moddley D, Pillay K, et al: Pharmacokinetics and antiretroviral activity of lamivudine alone or when coadministered with zidovudine in human immunodeficiency virus type 1-infected pregnant women and their offspring. J Infect Dis 1998a; 178:1327-1.
    101) Moore RD, Wong WME, & Keruly JC: Incidence of neuropathy in HIV-infected patients on monotherapy versus those on combination therapy with didanosine, stavudine and hydroxyurea. AIDS 2000; 14:273-278.
    102) Moyle G: Clinical manifestations and management of antiretroviral nucleoside analog-related mitochondrial toxicity. Clin Ther 2000a; 22:911-936.
    103) Moyle G: Toxicity of antiretroviral nucleoside and nucleotide analogues. Is mitochondrial toxicity the only mechanism?. Drug Safety 2000; 23:467-481.
    104) Moyle GJ & Sadler M: Peripheral neuropathy with nucleoside antiretrovirals. Drug Safety 1998; 19:481-494.
    105) Murphy RL: New antiretroviral drugs, Part II; NRTIs, NNRTIs, and beyond. AIDS Clinical Cure 1999; 11(6):47-53.
    106) Murray HW, Squires KE, & Weiss W: Stavudine in patients with AIDS-Related complex: AIDS clinical trials group 089. J Infect Dis 1995; 171(Suppl2):S123-S130.
    107) None Listed: Position paper: cathartics. J Toxicol Clin Toxicol 2004; 42(3):243-253.
    108) Nurutdinova D, Onen NF, Hayes E, et al: Adverse effects of tenofovir use in HIV-infected pregnant women and their infants. Ann Pharmacother 2008; 42(11):1581-1585.
    109) Pan CQ, Mi LJ, Bunchorntavakul C, et al: Tenofovir disoproxil fumarate for prevention of vertical transmission of hepatitis B virus infection by highly viremic pregnant women: a case series. Dig Dis Sci 2012; 57(9):2423-2429.
    110) Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission: Recommendations for Use of Antiretroviral Drugs in Pregnant HIV-1-Infected Women for Maternal Health and Interventions to Reduce Perinatal HIV Transmission in the United States. Antiretroviral Pregnancy Registry. Wilmington, NC. 2010. Available from URL: http://aidsinfo.nih.gov/contentfiles/PerinatalGL.pdf. As accessed 2010-05-24.
    111) Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission: Recommendations for use of antiretroviral drugs in pregnant HIV-1-infected women for maternal health and interventions to reduce perinatal HIV transmission in the United States. AIDSinfo, U.S. Department of Health and Human Services. Rockville, MD. 2012. Available from URL: http://aidsinfo.nih.gov/guidelines/html/3/perinatal-guidelines/0/. As accessed 2012-08-02.
    112) Petersen EA, Ramirez-Ronda CH, & Hardy WD: Dose-related activity of stavudine in patients infected with human immunodeficiency virus. J Infect Dis 1995; 171(Suppl 2):S131-S139.
    113) Peyriere H, Reynes J, Rouanet I, et al: Renal tubular dysfunction associated with tenofovir therapy. Report of 7 cases. J Acquir Immune Defic Syndr 2004; 35:269-273.
    114) Pluda JM, Cooley TP, & Montaner JSG: A phase I/II study of 2'-deoxy-3'-thiacytidine (lamivudine) in patients with advanced human immunodeficiency virus infection. J Infect Dis 1995; 171:1438-1447.
    115) Pollack MM, Dunbar BS, & Holbrook PR: Aspiration of activated charcoal and gastric contents. Ann Emerg Med 1981; 10:528-529.
    116) Pons JC, Boubon MC, & Tauret AM: Fetoplacental passage of 2,3-dideoxyinosine (letter). Lancet 1991; 337:732.
    117) Pottage J, Thompson M & Kahn J et al: Potent antiretroviral efficacy of low dose FTC: initial results from a phase I/II clinical trial (abstract). 5th Conference on Retroviruses and Opportunistic Infections, 1998.
    118) Powderly WG, Klebert MK, & Clifford DB: Ototoxicity associated with dideoxycytidine (letter). Lancet 1990; 335:1106.
    119) Product Information: ATRIPLA(R) oral tablets, efavirenz/emtricitabine/tenofovir disoproxil fumarate oral tablets. Gilead Sciences and Bristol-Myers Squibb, LLC, Foster City, CA, 2010.
    120) Product Information: COMBIVIR(R) oral tablets, lamivudine, zidovudine oral tablets. GlaxoSmithKline, Research Triangle Park, NC, 2009.
    121) Product Information: COMPLERA(R) oral tablets, emtricitabine, rilpivirine, tenofovir disoproxil fumarate oral tablets. Gilead Sciences, Inc.(per manufacturer), Foster City, CA, 2016.
    122) Product Information: COMPLERA(TM) oral tablets, emtricitabine/rilpivirine/tenofovir disoproxil fumarate oral tablets. Gilead Sciences, Inc. (per Manufacturer), Foster City, CA, 2011.
    123) Product Information: DESCOVY(R) oral tablets, emtricitabine, tenofovir alafenamide oral tablets. Gilead Sciences Inc (per manufacturer), Foster City, CA, 2016.
    124) Product Information: EMTRIVA(R) capsules, oral solution, emtricitabine capsules, oral solution. Gilead Sciences, Foster City, CA, 2008.
    125) Product Information: EMTRIVA(R) oral capsule, solution, emtricitabine oral capsule, solution. Gilead Sciences, Inc. (per FDA), Foster City, CA, 2012.
    126) Product Information: EPIVIR(R) oral tablets, solution, lamivudine oral tablets, solution. GlaxoSmithKline, Research Triangle Park, NC, 2009.
    127) Product Information: EPIVIR(R) oral tablets, oral solution, lamivudine oral tablets, oral solution. ViiV Healthcare (per FDA), Research Triangle Park, NC, 2015.
    128) Product Information: EPIVIR(R) oral tablets, oral solution, lamivudine oral tablets, oral solution. ViiV Healthcare (per FDA), Research Triangle Park, NC, 2015a.
    129) Product Information: EPIVIR(R) oral tablets, oral solution, lamivudine oral tablets, oral solution. ViiV Healthcare (per FDA), Research Triangle Park, NC, 2015b.
    130) Product Information: EPIVIR(R) oral tablets, solution, lamivudine oral tablets, solution. GlaxoSmithKline, Research Triangle Park, NC, 2008.
    131) Product Information: EPIVIR-HBV(R) oral tablets, oral solution, lamivudine oral tablets, oral solution. GlaxoSmithKline, Research Triangle Park, NC, 2004.
    132) Product Information: EPIVIR-HBV(R) oral tablets, solution, lamivudine oral tablets, solution. GlaxoSmithKline, Research Triangle Park, NC, 2007.
    133) Product Information: EPZICOM(R) oral tablets, abacavir sulfate and lamivudine oral tablets. GlaxoSmithKline (per manufacturer), Research Triangle Park, NC, 2015.
    134) Product Information: EPZICOM(R) oral tablets, abacavir sulfate and lamivudine oral tablets. GlaxoSmithKline, Research Triangle Park, NC, 2009.
    135) Product Information: EPZICOM(R) oral tablets, abacavir sulfate lamivudine oral tablets. ViiV Healthcare (per FDA), Research Triangle Park, NC, 2015a.
    136) Product Information: Epivir(R), lamivudine. Glaxo Wellcome Inc, Research Triangle Park, NC, 2001.
    137) Product Information: GENVOYA(R) oral tablets, elvitegravir, cobicistat, emtricitabine, tenofovir alafenamide oral tablets. Gilead Sciences, Inc. (per manufacturer), Foster City, CA, 2015.
    138) Product Information: HEPSERA(R) oral tablets, adefovir dipivoxil oral tablets. Gilead Sciences,Inc, Foster City, CA, 2006.
    139) Product Information: HEPSERA(R) oral tablets, adefovir dipivoxil oral tablets. Gilead Sciences, Inc. (per FDA), Foster City, CA, 2012.
    140) Product Information: HIVID(R) oral tablets, zalcitabine oral tablets. Roche Laboratories Inc, Nutley, NJ, 2002.
    141) Product Information: HIVID(R), zalcitabine tablets. Roche Laboratories Inc., Nutley, NJ, 2002.
    142) Product Information: Hivid(R), zalcitabine. Roche Laboratories, Nutley, NJ, 1996.
    143) Product Information: LEUKINE(R) injection, sargramostim injection. Berlex, Seattle, WA, 2006.
    144) Product Information: NEUPOGEN(R) injection, filgrastim injection. Amgen,Inc, Thousand Oaks, CA, 2006.
    145) Product Information: ODEFSEY(R) oral tablets, emtricitabine, rilpivirine, tenofovir alafenamide oral tablets. Gilead Sciences Inc (per manufacturer), Foster City, CA, 2016.
    146) Product Information: RESCRIPTOR(R) oral tablets, delavirdine mesylate oral tablets. Pharmacia & Upjohn Company, New York, NY, 2008.
    147) Product Information: RETROVIR(R) oral tablets, capsules, syrup, zidovudine oral tablets, capsules, syrup. GlaxoSmithKline, Research Triangle Park, NC, 2008.
    148) Product Information: RETROVIR(R) oral tablets, capsules, syrup, zidovudine oral tablets, capsules, syrup. GlaxoSmithKline, Research Triangle Park, NC, 2010.
    149) Product Information: Retrovir(R), zidovudine. Glaxo Wellcome Inc, Research Triangle Park, NC, 1996.
    150) Product Information: STRIBILD(TM) oral tablets, elvitegravir cobicistat emtricitabine tenofovir disoproxil fumarate oral tablets. Gilead Sciences, Inc. (per manufacturer), Foster City, CA, 2012.
    151) Product Information: TRIUMEQ(R) oral tablets, abacavir dolutegravir lamivudine oral tablets. ViiV Healthcare (per manufacturer), Research Triangle Park, NC, 2014.
    152) Product Information: TRIZIVIR oral tablets, abacavir lamivudine zidovudine oral tablets. GlaxoSmithKline (per manufacturer), Research Triangle Park, NC, 2015.
    153) Product Information: TRIZIVIR(R) oral tablets, abacavir sulfate, lamivudine, and zidovudine oral tablets. GlaxoSmithKline, Research Triangle Park, NC, 2009.
    154) Product Information: TRUVADA(R) oral tablets, emtricitabine tenofovir disoproxil fumarate oral tablets. Gilead Sciences, Inc, Foster City, CA, 2012.
    155) Product Information: TRUVADA(R) oral tablets, emtricitabine, tenofovir disoproxil fumarate oral tablets. Gilead Sciences (per manufacturer), Foster City, CA, 2016.
    156) Product Information: TRUVADA(R) oral tablets, emtricitabine/tenofovir disoproxil fumarate oral tablets. Gilead Sciences Inc, Foster City, CA, 2009.
    157) Product Information: VIDEX(R) pediatric powder for oral solution, didanosine pediatric powder for oral solution. Bristol Myers Squibb, New York, NY, 2010.
    158) Product Information: VIREAD(R) oral tablets, oral powder, tenofovir disoproxil fumarate oral tablets, oral powder. Gilead Sciences, Inc. (per FDA), Foster City, CA, 2012.
    159) Product Information: VIREAD(R) oral tablets, oral powder, tenofovir disoproxil fumarate oral tablets, oral powder. Gilead Sciences, Inc. (per FDA), Foster City, CA, 2016.
    160) Product Information: VIREAD(R) oral tablets, powder, tenofovir disoproxil fumarate oral tablets, powder. Gilead Sciences, Inc. (per manufacturer), Foster City, CA, 2012.
    161) Product Information: VIREAD(R) oral tablets, tenofovir disoproxil fumarate oral tablets. Gilead Sciences, Inc., 2010.
    162) Product Information: Videx(R), didanosine. Bristol-Myers Squibb, Princeton, NJ, 1996.
    163) Product Information: ZERIT(R) oral capsules, oral solution, stavudine oral capsules, oral solution. Bristol-Myers Squibb Company, Princeton, NJ, 2006.
    164) Product Information: ZERIT(R) oral capsules, solution, stavudine oral capsules, solution. Bristol-Myers Squibb, Princeton, NJ, 2009.
    165) Product Information: ZIAGEN(R) oral solution, oral tablets, abacavir sulfate oral solution, oral tablets. GlaxoSmithKline, Research Triangle Park, NC, 2008.
    166) Product Information: ZIAGEN(R) oral tablets oral solution, abacavir oral tablets oral solution. ViiV Healthcare (per manufacturer), Research Triangle Park, North Carolina, 2015.
    167) Product Information: ZIAGEN(R) oral tablets, oral solution, abacavir sulfate oral tablets, oral solution. ViiV Healthcare (per FDA), Research Triangle Park, NC, 2015a.
    168) Product Information: ZIAGEN(R) oral tablets, oral solution, abacavir sulfate oral tablets, oral solution. ViiV Healthcare (per Manufacturer), Research Triangle Park, NC, 2015.
    169) Product Information: ZIAGEN(R) oral tablets, solution, abacavir sulfate oral tablets, solution. GlaxoSmithKline, Research Triangle Park, NC, 2008a.
    170) Product Information: ZIAGEN(R) oral tablets, solution, abacavir sulfate oral tablets, solution. GlaxoSmithKline, Research Triangle Park, NC, 2008.
    171) Product Information: Zerit(R),, stavudine,. Bristol-Myers Squibb,, Princeton,, NJ,, 2002.
    172) Product Information: Ziagen(TM), abacavir. Glaxo Wellcome Inc, Research Triangle Park, NC, 1998.
    173) Product Information: diazepam IM, IV injection, diazepam IM, IV injection. Hospira, Inc (per Manufacturer), Lake Forest, IL, 2008.
    174) Product Information: lorazepam IM, IV injection, lorazepam IM, IV injection. Akorn, Inc, Lake Forest, IL, 2008.
    175) Product Information: stavudine oral capsules, stavudine oral capsules. Mylan Pharmaceuticals Inc. (per DailyMed), Morgantown, WV, 2015.
    176) Rachlis A & Fanning MM: Zidovudine toxicity. Clinical features and management. Drug Safety 1993; 8:312-320.
    177) Rau NR, Nagaraj MV, Prakash PS, et al: Fatal pulmonary aspiration of oral activated charcoal. Br Med J 1988; 297:918-919.
    178) Richardson M, Osrin D, Donaghy S, et al: Spinal malformations in the fetuses of HIV infected women receiving combination antiretroviral therapy and co-trimoxazole. Eu J Obstet Gynecol Reprod Biol 2000; 93:215-217.
    179) Ristola M, Salo E, Ammala P, et al: Combined stavudine and lamivudine during pregnancy [letter]. AIDS 1999; 13(2):285.
    180) Roberts WL, Buckley TJ, & Rainey PM: Solid-phase extraction combined with radioimmunoassay for measurement of zalcitabine (2',3'-dideoxycytidine) in plasma and serum. Clin Chem 1994; 40:211-215.
    181) Sandberg JA, Binienda Z, & Lipe G: Placental transfer and fetal disposition of 2',3'-dideoxycytidine and 2'3'-dideoxyinosine in the rhesus monkey. Drug Metab Dispos 1995; 23:881-884.
    182) Sanders VM, Elwell MR, & Heath JE: Induction of thymic lymphoma in mice administered the dideoxynucleoside ddC. Fund Appl Toxicol 1995; 27:263-269.
    183) Sandstrom E & Oberg B: Antiviral therapy in human immunodeficiency virus infections. Current status (Part I). Drugs 1993; 45:488-508.
    184) Scott R, Besag FMC, & Neville BGR: Buccal midazolam and rectal diazepam for treatment of prolonged seizures in childhood and adolescence: a randomized trial. Lancet 1999; 353:623-626.
    185) Sheng WH, Hsieh SM, Lee SC, et al: Fatal lactic acidosis associated with highly active antiretroviral therapy in patients with advanced human immunodeficiency virus infection in Taiwan. Int J STD AIDS 2004; 15:249-253.
    186) Shiber JR: Lactic acidosis caused by nucleoside analogues. Am J Emerg Med 2005; 23(4):582-583.
    187) Siberry GK, Williams PL, Mendez H, et al: Safety of tenofovir use during pregnancy: early growth outcomes in HIV-exposed uninfected infants. AIDS 2012; Epub:Epub.
    188) Simdon J, Watters D, & Bartlett S: Ototoxicity associated with use of nucleoside analog reverse transcriptase inhibitors: a report of 3 possible cases and review of the literature. Clin Infect Dis 2001; 32:1623-1627.
    189) Simpson DM & Tagliati M: Nucleoside analogue-associated peripheral neuropathy in human immunodeficiency virus infection. J Acquir Immun Defic Syn Hum Retrovir 1995; 9:153-161.
    190) Skowron G: Biologic effects and safety of stavudine: overview of phase I and II clinical trials. J Infect Dis 1995; 171(Suppl 2):S113-S117.
    191) Sreenath TG, Gupta P, Sharma KK, et al: Lorazepam versus diazepam-phenytoin combination in the treatment of convulsive status epilepticus in children: A randomized controlled trial. Eur J Paediatr Neurol 2009; Epub:Epub.
    192) Stek AM , Best BM , Luo W , et al: Effect of pregnancy on emtricitabine pharmacokinetics. HIV Med 2012; 13(4):226-235.
    193) Stekler J, Maenza J, Stevens C, et al: Abacavir hypersensitivity reaction in primary HIV infection. AIDS 2006; 20(9):1269-1274.
    194) Stull DM, Bilmes R, Kim H, et al: Comparison of sargramostim and filgrastim in the treatment of chemotherapy-induced neutropenia. Am J Health Syst Pharm 2005; 62(1):83-87.
    195) Tal A & Dall L: Didanosine-induced hypertriglyceridemia. Am J Med 1993; 95:247.
    196) Tancrede-Bohin E, Grange F, & Bournerias I: Hypersensitivity syndrome associated with zalcitabine therapy. Lancet 1996; 347:971-972.
    197) Taylor GP & Low-Beer N: Antiretroviral therapy in pregnancy. A focus on safety. Drug Safety 2001; 24:683-702.
    198) Torres SM, Walker DM, Carter MM, et al: Mutagenicity of zidovudine, lamivudine, and abacavir following in vitro exposure of human lymphoblastoid cells or in utero exposure of CD-1 mice to single agents or drug combinations. Environ Mol Mutagen. 2007; 48(3-4):224-238.
    199) Tremoulet AH, Capparelli EV, Patel P, et al: Population pharmacokinetics of lamivudine in human immunodeficiency virus-exposed and -infected infants. Antimicrob Agents Chemother 2007; 51(12):4297-4302.
    200) Tuomala RE, Shapiro DE, Mofenson LM, et al: Antiretroviral therapy during pregnacy and the risk of an adverse outcome. N Engl J Med 2002a; 346(24):1863-1870.
    201) Tuomala RE, Shapiro DE, Mofenson LM, et al: Antiretroviral therapy during pregnancy and the risk of an adverse outcome. N Engl J Med 2002; 346(24):1863-1870.
    202) Venerosi A, Valanzano A, & Alleva E: Prenatal exposure to anti-HIV drugs: neurobehavioral effects of zidovudine (AZT) + lamivudine (3TC) treatment in mice. Teratology 2001; 63:26-37.
    203) Venhoff N & Walker UA: Mitochondrial disease in the offspring as a result of antiretroviral therapy. Expert Opin Drug Saf 2006; 5(3):373-381.
    204) Verma A, Schein RM, Jayaweera DT, et al: Fulminant neuropathy and lactic acidosis associated with nucleoside analog therapy. Neurology 1999; 53(6):1365-1367.
    205) Walker UA, Setzer B, & Volksbeck SIL: Toxicity of nucleoside-analogue reverse- transcriptase inhibitors (letter). Lancet 2000; 355:1096.
    206) Wallace MR, Miller LK, & Briqnac H: Clinical experience with stavudine as salvage therapy in patients with advanced HIV disease (letter). J Acquir Immune Defic Syn Hum Retrovir 1996; 13:96-97.
    207) Wohl DA, Pilcher CD, Evans S, et al: Absence of sustained hyperlactatemia in HIV-infected patients with risk factors for mitochondrial toxicity. J Acquir Immune Defic Syndr 2004; 35:274-278.
    208) Woodward CL, Hall AM, Williams IG, et al: Tenofovir-associated renal and bone toxicity. HIV Med 2009; 10(8):482-487.
    209) Working Group on Antiretroviral Therapy and Medical Management of HIV-Infected Children: Guidelines for the Use of Antiretroviral Agents in Pediatric HIV Infection. AIDSinfo. Rockville, MD. 2009. Available from URL: http://aidsinfo.nih.gov/contentfiles/PediatricGuidelines.pdf. As accessed 2009-02-25.
    210) Yarchoan R, Mitsuya H, & Meyers CE: Clinical pharmacology of 3'-azido-2',3'-dideoxythymidine (zidovudine) and related dideoxynucleosides. N Engl J Med 1989; 321:726-738.
    211) ter Hofstede HJ, de Marie S, & Foudraine NA: Clinical features and risk factors of lactic acidosis following long-term antiretroviral therapy: 4 fatal cases. Intl J STD AIDS 2000; 11:611-616.
    212) van Leeuwen R, Katlama C, & Kitchen V: Evaluation of safety and efficacy of 3TC (lamivudine) in patients with asymptomatic or mildly symptomatic human immunodeficiency virus infection: a phase I/II study. J Infect Dis 1995; 171:1166-1171.
    213) van Leeuwen R, Katlama C, & Kitchen V: The safety and pharmacokinetics of a reverse transcriptase inhibitor, 3TC, in patients with HIV infection: a Phase I study. AIDS 1992; 6:1471-1475.
    214) van Leeuwen R, Lange JM, Hussey EK, et al: The safety and pharmacokinetics of a reverse transcriptase inhibitor, 3TC, in patients with HIV infection: a phase I study. AIDS 1992a; 6(12):1471-1475.