97-17854. U.S. Public Health Service Recommendations for Use of Antiretroviral Drugs During Pregnancy for Maternal Health and Reduction of Perinatal Transmission of Human Immunodeficiency Virus Type 1 in the United States; Request for Comment  

  • [Federal Register Volume 62, Number 131 (Wednesday, July 9, 1997)]
    [Notices]
    [Pages 36809-36823]
    From the Federal Register Online via the Government Publishing Office [www.gpo.gov]
    [FR Doc No: 97-17854]
    
    
    =======================================================================
    -----------------------------------------------------------------------
    
    DEPARTMENT OF HEALTH AND HUMAN SERVICES
    
    Office of Public Health and Science, HHS
    
    
    U.S. Public Health Service Recommendations for Use of 
    Antiretroviral Drugs During Pregnancy for Maternal Health and Reduction 
    of Perinatal Transmission of Human Immunodeficiency Virus Type 1 in the 
    United States; Request for Comment
    
    AGENCY: Office of Public Health and Science, HHS.
    
    ACTION: Notice.
    
    -----------------------------------------------------------------------
    
    SUMMARY: The Department of Health and Human Services, Office of Public 
    Health and Science is establishing guidelines for use of antiretroviral 
    drugs by HIV-1-infected pregnant women for maternal health indications 
    and reduction of perinatal HIV-1 transmission.
    
    DATES: Comments on the proposed guidelines must be received on or 
    before August 8, 1997 in order to ensure that NIH will be able to 
    consider the comments in preparing the final guidelines.
    
    ADDRESSES: Written comments to this notice should be submitted to: The 
    HIV/AIDS Treatment Information Service, P.O. Box 6303, Rockville, MD 
    20849-
    
    [[Page 36810]]
    
    6303. Only written comments will be accepted. After consideration of 
    the comments, the final document will be published in the Centers for 
    Disease Control and Prevention (CDC) ``Morbidity and Mortality Weekly 
    Report'' (MMWR).
    
    FOR FURTHER INFORMATION CONTACT: Copies of the ``U.S. Public 
    HealthService Recommendations for Use of Antiretroviral Drugs During 
    Pregnancy for Maternal Health and Reduction of Perinatal Transmission 
    of Human Immunodeficiency Virus Type 1 in the United States'' are 
    available from the National AIDS Clearinghouse (1-800-458-5231) and on 
    the Clearinghouse Web site (http://www.cdcnac.org) and from the HIV/
    AIDS Treatment Information Service (1-800-448-0440; Fax: 301-519-6616; 
    TTY: 1-800-243-7012) and on their Web site (http://www.hivatis.org).
    
    SUPPLEMENTARY INFORMATION: The U.S. Public Health Service Task Force 
    Recommendations for Use of Antiretroviral Drugs During Pregnancy for 
    Maternal Health and Reduction of Perinatal Transmission of Human 
    Immunodeficiency Virus Type 1 would update the 1994 guidelines 
    developed by the U.S. Public Health Service for use of zidovudine (ZDV) 
    to reduce the risk of perinatal human immunodeficiency virus (HIV) type 
    1 transmission.(MMWR 1994)
        On May 9, 1997 the U.S. Public Health Service convened a ``Workshop 
    on Antiretroviral Therapy to Reduce the Risk of Perinatal 
    Transmission'' to review information related to use of antiretroviral 
    drugs to reduce perinatal HIV transmission and for treatment of HIV 
    infection in women in the United States. The medical, scientific, 
    public health and bioethics communities and interested professional, 
    community and advocacy organizations were represented. These guidelines 
    represent a consensus of 35 expert consultants, including medical, 
    public health, and bioethics specialists, HIV-infected women and AIDS 
    advocacy organization representatives, who have reviewed and revised 
    the document twice since that meeting. The document has also been sent 
    for review by 22 representatives of professional and AIDS advocacy 
    organizations.
        In February 1994, the results of Pediatric AIDS Clinical Trials 
    Group (PACTG) Protocol 076 demonstrated that ZDV chemoprophylaxis could 
    reduce perinatal HIV-1 transmission by nearly 70%.(Connor 1994) Since 
    that time, epidemiologic data have confirmed the efficacy of ZDV for 
    reduction of perinatal transmission and extended this efficacy to 
    children of women with advanced disease, low CD4 lymphocyte count and 
    prior ZDV therapy. Additionally, there have been major advances in 
    understanding the pathogenesis of HIV-1 infection and in the treatment 
    and monitoring of HIV-1 disease. These advances have resulted in 
    changes in standard antiretroviral therapy recommendations for HIV-1-
    infected adults in the United States to more aggressive combination 
    drug regimens that maximally suppress viral replication. Although 
    considerations related to pregnancy may factor into decisions as to 
    timing and choice of therapy, pregnancy per se is not an adequate 
    reason to defer standard therapy. There are unique considerations 
    regarding use of antiretroviral drugs in pregnancy, including the 
    potential need to alter dosing due to physiologic changes associated 
    with pregnancy, the potential for adverse short- or long-term effects 
    on the fetus and newborn, and effectiveness for reducing the risk of 
    perinatal transmission. Data to address many of these considerations 
    are not yet available. Therefore, offering antiretroviral therapy to an 
    HIV-1-infected woman during pregnancy, whether primarily to treat her 
    HIV-1 infection, primarily to reduce perinatal transmission, or for 
    both purposes, should be accompanied by a discussion of the known and 
    unknown short- and long-term benefits and risks of such therapy for her 
    and her infant. Standard antiretroviral therapy should be discussed 
    with and offered to HIV-1-infected pregnant women. Additionally, to 
    prevent perinatal transmission, ZDV chemoprophylaxis should be 
    incorporated into whatever antiretroviral regimen is offered. This 
    document is intended to give the health care professional information 
    for discussion with the woman to enable her to make an informed 
    decision regarding use of antiretroviral drugs during pregnancy.
    
    Introduction
    
        In February 1994, PACTG Protocol 076 demonstrated that a 3-part 
    regimen of ZDV could reduce the risk of mother to child HIV-1 
    transmission by nearly 70%.(Connor 1994) The regimen includes oral ZDV 
    initiated at 14 to 34 weeks gestation and continuing throughout 
    pregnancy, followed by intravenous ZDV during labor and oral 
    administration of ZDV to the infant for 6 weeks after delivery (Table 
    1). In August 1994, a U.S. Public Health Service (USPHS) Task Force 
    issued recommendations for use of ZDV for reduction of perinatal HIV-1 
    transmission (MMWR 1994), and in July 1995, the USPHS issued 
    recommendations for universal prenatal HIV-1 counseling and HIV-1 
    testing with consent for all pregnant women in the U.S..(MMWR 1995) In 
    the three years since these results became available, epidemiologic 
    studies in the U.S. and France have demonstrated dramatic decreases in 
    perinatal transmission following incorporation of the PACTG 076 ZDV 
    regimen into general clinical practice. (Cooper 1996; Fiscus 1996; 
    Fiscus 1997; Thomas 1997; Blanche 1997;Simonds 1996)
        Since 1994 there have been major advances in understanding the 
    pathogenesis of HIV-1 infection and in the treatment and monitoring of 
    HIV-1 disease. It is now appreciated that the rapidity and magnitude of 
    viral turnover during all stages of HIV-1 infection is much greater 
    than previously recognized; plasma virions are estimated to have a mean 
    half-life of only 6 hours.(Perelson 1996) Thus, current therapeutic 
    interventions focus on early initiation of aggressive combination 
    antiretroviral regimens to maximally suppress viral replication, 
    preserve immune function, and reduce the development of 
    resistance.(Havlir 1996) New, potent antiretroviral drugs which inhibit 
    the protease enzyme of HIV-1 are now available. When a protease 
    inhibitor is used in combination with nucleoside analogue reverse 
    transcriptase inhibitors, plasma HIV-1 RNA levels may be reduced for 
    prolonged periods of time to undetectable levels using current assays. 
    Improved clinical outcome and survival have been observed in adults 
    receiving such regimens. Additionally, more direct quantitation of 
    viral load has become available through assays that measure HIV-1 RNA 
    copy number; these assays have provided powerful new tools to assess 
    disease stage and risk for progression as well as the effects of 
    therapy. These advances have led to major changes in the standard of 
    care for treatment and monitoring for HIV-1-infected adults in the 
    United States.
        There have also been advances in the understanding of the 
    pathogenesis of perinatal HIV-1 transmission. It is now recognized that 
    the majority of perinatal transmission likely occurs near to or during 
    delivery.(Mofenson 1997) Additional data and follow-up are now 
    available on infants and women enrolled in PACTG 076 demonstrating the 
    short-term safety of the ZDV regimen, but new data from animal studies 
    affirm the need for long-term follow-up of children with antiretroviral 
    exposure in utero.
    
    [[Page 36811]]
    
        These developments have important implications for maternal and 
    fetal health. Antiretroviral use in HIV-1 infected women during 
    pregnancy must take into account two separate but elated issues: (1) 
    Antiretroviral treatment of the woman's HIV infection, and (2) 
    Antiretroviral chemoprophylaxis to reduce the risk of perinatal HIV-1 
    transmission. While ZDV chemoprophylaxis alone has been shown to 
    significantly reduce the risk of perinatal transmission, antiretroviral 
    monotherapy is now considered to be suboptimal for treatment of HIV 
    infection, and combination drug therapy is the current standard of care 
    when considering treatment of the woman's HIV infection in the United 
    States. The USPHS Panel on Clinical Practices for Treatment of HIV 
    Infection will soon release guidelines for use of antiretrovirals in 
    infected adolescents and adults, including use of antiretrovirals for 
    treatment of infected women who are pregnant.(Panel 1997) The current 
    document will focus on antiretroviral chemoprophylaxis for reduction of 
    perinatal transmission, and will review the special considerations 
    regarding use of antiretroviral drugs in pregnant women; update the 
    results of PACTG 076 and related clinical trials and epidemiologic 
    studies; discuss use of HIV-1 RNA assays during pregnancy; and provide 
    updated recommendations on antiretroviral chemoprophylaxis for the 
    reduction of perinatal transmission.
        These recommendations have been developed for use in the United 
    States. Although perinatal HIV-1 transmission is an international 
    problem, alternative strategies may be appropriate in other countries. 
    The policy and practices in other countries regarding use of 
    antiretroviral drugs for reduction of perinatal HIV-1 transmission may 
    differ from these recommendations, and will depend on local 
    considerations, including availability and cost of ZDV, access to 
    facilities for safe intravenous infusions during labor, and alternative 
    interventions that may be under evaluation in that area.
    
    Special Considerations Regarding the use of Antiretroviral Drugs by 
    HIV-1-Infected Pregnant Women and Their Infants
    
        Treatment recommendations for HIV-1-infected pregnant women have 
    been based on the belief that therapies of known benefit to women 
    should not be withheld during pregnancy unless there are known adverse 
    effects on the mother, fetus or infant and these adverse effects 
    outweigh the benefit to the woman.(Minkoff 1997) Thus, given the 
    absence of demonstrated risk and compelling evidence of therapeutic 
    advantage, guidelines for optimal antiretroviral therapy in pregnant 
    HIV-1-infected women should be the same as those delineated for non-
    pregnant adults. However, it must be realized that the potential impact 
    of such therapy on the fetus and infant is unknown, and long-term 
    follow-up is needed for children who have had exposure to 
    antiretroviral drugs in utero. The decision to use any antiretroviral 
    drug during pregnancy should be made by the woman following discussion 
    with her health care provider regarding the known and unknown benefits 
    and risks to her and her fetus.
        Combination antiretroviral therapy, generally consisting of two 
    nucleoside analogue reverse transcriptase inhibitors and a protease 
    inhibitor, is the currently recommended standard treatment for non-
    pregnant HIV-1-infected adults with CD4 lymphocyte count <500 m\3\,="" hiv-1="" rna="" copy="" number="">10,000/mL, or clinical symptoms of HIV disease. 
    Pregnancy per se should not preclude use of optimal therapeutic 
    regimens. However, recommendations regarding the choice of 
    antiretroviral drugs for treatment of infected pregnant women are 
    subject to unique considerations, including potential changes in dosing 
    requirements due to the physiologic changes associated with pregnancy 
    and the potential effects of the antiretroviral drug on the fetus and 
    newborn.
        Physiologic changes that occur during pregnancy may affect the 
    kinetics of drug absorption, distribution, biotransformation and 
    elimination in the pregnant woman, thereby affecting drug dose 
    requirements. During pregnancy, gastrointestinal transit time becomes 
    prolonged; body water and fat increase over gestation accompanied by 
    increases in cardiac output, ventilation, and liver and renal blood 
    flow; plasma protein concentrations decrease; renal sodium reabsorption 
    increases; and there are changes in metabolic enzyme pathways in the 
    liver. Placental transport of drugs, compartmentalization of drugs in 
    the embryo/fetus and placenta, and biotransformation of drugs by the 
    fetus and placenta as well as elimination of drugs by the fetus can 
    also affect drug pharmacokinetics in the pregnant woman. Additional 
    important considerations regarding drug use in pregnancy are the 
    effects of the drug on the fetus and newborn, including the potential 
    for teratogenicity, mutagenicity, or carcinogenicity, and the 
    pharmacokinetics and toxicity of transplacentally-transferred drugs. 
    The potential harm to the fetus from maternal ingestion of a specific 
    drug depends not only on the drug itself, but the dose ingested, the 
    gestational age at exposure, duration of exposure, the interaction with 
    other agents to which the fetus is exposed, and to an unknown extent, 
    the genetic makeup of the mother and fetus.
        Information about the safety of drugs in pregnancy comes from 
    animal toxicity data, anecdotal experience, registry data and clinical 
    trials. There are currently minimal data available on the 
    pharmacokinetics and safety of antiretrovirals during pregnancy for 
    antiretrovirals other than ZDV. In the absence of data, drug choice 
    needs to be individualized based on discussion with the woman and 
    available data from preclinical and clinical testing of the individual 
    drugs.
        Preclinical data include in vitro and animal in vivo screening 
    tests for carcinogenicity, clastogenicity/mutagenicity, and 
    reproductive and teratogenic effects. It is important to recognize that 
    the predictive value of such tests for adverse effects in humans is 
    unknown. For example, of approximately 1,200 known animal teratogens, 
    only about 30 are known to be teratogenic in humans. (Mills 1995) In 
    addition to antiretroviral agents, many drugs commonly used to treat 
    the consequences of HIV-1 infection may have positive findings on one 
    or more of these screening tests. For example, acyclovir is positive on 
    some in vitro carcinogenicity and clastogenicity assays and is 
    associated with some fetal abnormalities in rats; however, data on 
    human experience from the Acyclovir in Pregnancy Registry indicate no 
    increased risk of birth defects in infants with in utero exposure to 
    acyclovir to date. (MMWR 1993) Table 2 shows the FDA Pregnancy Category 
    and available data regarding placental passage and long-term animal 
    carcinogenicity studies for currently approved antiretroviral drugs.
    
    Nucleoside Analogue Reverse Transcriptase Inhibitors
    
        Of the five currently approved nucleoside analogue antiretrovirals, 
    only ZDV and lamivudine (3TC) pharmacokinetics have been evaluated in 
    clinical trials in human pregnancy to date. ZDV is well-tolerated in 
    pregnancy at usual adult doses and in the full-term neonate at 2 mg per 
    kg body weight orally every 6 hours, as observed in PACTG 076. A small 
    phase I study in South Africa evaluated the safety and pharmacokinetics 
    of 3TC alone or in combination with ZDV in 20 infected pregnant women 
    starting at 38 weeks gestation through labor and given for 1 week 
    following birth to their infants.
    
    [[Page 36812]]
    
    (Johnson 1996, Moodley 1997) The drug was well-tolerated in the women 
    at the usual adult dose of 150 mg orally twice daily, had 
    pharmacokinetics similar to those observed in non-pregnant adults, and 
    no pharmacokinetic interaction with ZDV was observed. No data are 
    currently available regarding the pharmacokinetics of 3TC administered 
    earlier than 38 weeks gestation. The drug crossed the placenta, 
    achieving comparable serum concentrations in the woman, umbilical cord 
    and neonate, and no short-term adverse effects were observed in the 
    neonates. Oral clearance of 3TC in infants at 1 week of age was 
    prolonged compared to older pediatric populations (0.35 L per kg per 
    hour compared to 0.64-1.1 L per kg per hour, respectively). There are 
    currently no data on 3TC pharmacokinetics between 2-6 weeks of age, and 
    the exact age at which 3TC clearance begins to approximate that in 
    older children is not known. Based on these limited data, 3TC in a dose 
    of 150 mg administered orally twice daily in pregnant HIV-1-infected 
    women and 2 mg per kg body weight administered orally twice daily in 
    their neonates (half the dose recommended for older children) is being 
    evaluated in several phase I studies in combination with ZDV and other 
    drugs in the U.S., and in a phase III perinatal prevention trial in 
    Africa.
        In rodent studies, prolonged, continuous high doses of ZDV 
    administered to adult rodents have been associated with the development 
    of noninvasive squamous epithelial vaginal tumors in 3% to 12% of 
    females. (Ayers 1996) In humans, ZDV is extensively metabolized, and 
    the major form of ZDV excreted in the urine is the glucuronide, whereas 
    in mice, high concentrations of unmetabolized ZDV are excreted in the 
    urine. It is hypothesized by scientists at Glaxo-Wellcome, Inc., the 
    manufacturer of ZDV, that the vaginal tumors in mice may be a topical 
    effect of chronic local ZDV exposure of the vaginal epithelium, 
    resulting from reflux of urine containing highly concentrated ZDV from 
    the bladder into the vagina. Consistent with this hypothesis, in a 
    study conducted by Glaxo-Wellcome, Inc. in which 5 or 20 mg ZDV/mL 
    saline was administered intravaginally to female mice, vaginal squamous 
    cell carcinomas were observed in mice receiving the highest 
    concentration. (Ayers 1996) No increase in the incidence of tumors in 
    other organ sites has been seen in other studies of ZDV conducted in 
    adult mice and rats. High doses of zalcitabine (ddC) have been 
    associated with the development of thymic lymphomas in rodents. Long-
    term animal carcinogenicity screening studies in rodents administered 
    ddI or 3TC are negative; similar studies for stavudine (d4T) have not 
    been completed.
        Two rodent studies evaluating the potential for transplacental 
    carcinogenicity of ZDV have had differing results. In one ongoing study 
    carried out by scientists at the National Cancer Institute, two very 
    high daily doses of ZDV were administered during the last third of 
    gestation in mice. The doses chosen for this study were near the 
    maximum dose beyond which fetal toxicity would be observed and 
    approximately 25 and 50 times greater than the daily dose given to 
    humans, although the cumulative dose received by the pregnant mouse was 
    similar to the cumulative dose received by a pregnant woman taking 6 
    months of ZDV.
        In the offspring of ZDV-exposed pregnant mice at the highest dose 
    level followed for 12 months, a statistically significant increase in 
    lung, liver, and female reproductive organ tumors were observed; the 
    investigators also documented incorporation of ZDV into the DNA in a 
    variety of newborn mouse tissues, although this did not clearly 
    correlate with the presence of tumors. The second study was carried out 
    by scientists at Glaxo-Wellcome, Inc. In that study, pregnant mice were 
    given one of several regimens of ZDV; doses were based on 
    pharmacokinetic data in mice and humans and were intended to achieve 
    blood levels somewhat higher (approximately 3-fold) than those achieved 
    in clinical practice. The daily doses received by mice during gestation 
    ranged from one-twelfth to one-fiftieth the daily doses received by 
    mice in the previous study. Some of the offspring also received ZDV for 
    varying periods of time over their lifespan. No increase in the 
    incidence of tumors was observed in the offspring of these mice, except 
    in those offspring that had received additional lifetime ZDV exposure 
    in whom the previously noted vaginal tumors once again were noted.
        The relevance of these data to humans is unknown. An expert panel 
    convened by the National Institutes of Health in January 1997 to review 
    these data concluded that the proven benefit of ZDV in reducing the 
    risk of perinatal transmission outweighed the hypothetical concerns of 
    transplacental carcinogenesis raised by the rodent study. The panel 
    also concluded that the information regarding the theoretical risk of 
    transplacental carcinogenesis should be discussed with all HIV-infected 
    pregnant women in the course of counseling them on the benefits and 
    potential risks of antiretroviral therapy during pregnancy, and 
    emphasized the need for careful long-term follow-up of all children 
    exposed in utero to antiretroviral drugs. It is important to recognize 
    that transplacental carcinogenicity studies have not been performed for 
    any of the other available antiretroviral drugs, and no long-term or 
    transplacental animal carcinogenicity studies of combinations of 
    antiretroviral drugs have been performed.
        All of the nucleoside analogue antiretroviral drugs except 
    didanosine (ddI) are classified as FDA Pregnancy Category C (see 
    footnote to Table 2 for definitions); ddI is classified as Category B. 
    While all the nucleoside analogues cross the placenta in primates, in 
    primate and placental perfusion studies ddI and ddC undergo 
    significantly less placental transfer (fetal/maternal drug ratios of 
    0.3 to 0.5) than do ZDV, d4T and 3TC (fetal/maternal drug ratios >0.7).
    
    Non-Nucleoside Analogue Reverse Transcriptase Inhibitors
    
        There are 2 FDA-approved non-nucleoside reverse transcriptase 
    inhibitors, nevirapine and delavirdine. A phase I study in the U.S. 
    evaluated the safety and pharmacokinetics of nevirapine in 7 HIV-1-
    infected pregnant women and their infants. Nevirapine was administered 
    as a single 200 mg oral dose at the onset of labor, and as a single 
    dose of 2 mg per kg body weight at 2-3 days of age to their infants. 
    (Mirochnick 1997) The drug was well-tolerated by the women, crossed the 
    placenta and achieved neonatal blood concentrations equivalent to that 
    in the mother. No short-term adverse effects were observed in mothers 
    or neonates. Elimination of nevirapine in the pregnant women in this 
    study was prolonged (mean half-life, 66 hours) compared to non-pregnant 
    individuals (mean half-life, 45 hours following a single dose). Data on 
    chronic dosing with nevirapine beginning at 38 weeks gestation is under 
    study but not yet available; no data are available regarding the safety 
    and pharmacokinetics of chronic dosing with nevirapine beginning 
    earlier in pregnancy. The half-life of nevirapine was prolonged in 
    neonates (median half-life, 36.8 hours) compared to what is observed in 
    older children (mean half-life, 24.8 hours following a single dose). A 
    single dose of nevirapine at 2-3 days of age in neonates whose mothers 
    received nevirapine during labor maintained levels associated with 
    antiviral activity for the first week of life. (Mirochnick 1997) Based 
    on these data, a phase III perinatal transmission
    
    [[Page 36813]]
    
    prevention clinical trial sponsored by the PACTG will evaluate 
    nevirapine administered as a 200 mg single dose to the woman during 
    active labor and a single dose to the newborn at 2-3 days of age in 
    combination with standard maternal antiretroviral therapy and ZDV 
    chemoprophylaxis.
        Delavirdine has not been studied in pregnant women. Delavirdine is 
    positive on at least one in vitro screening test for carcinogenic 
    potential. Long-term and transplacental animal carcinogenicity studies 
    are not available for either of these drugs at the present time. Both 
    drugs are associated with impaired fertility in rodents when 
    administered at high doses, and delavirdine is teratogenic in rodents 
    when very high doses are administered during pregnancy (ventricular 
    septal defects were observed at doses associated with severe maternal 
    toxicity). Both nevirapine and delavirdine are classified as FDA 
    Pregnancy Category C.
    
    Protease Inhibitors
    
        Although phase I studies of several protease inhibitors (indinavir, 
    ritonavir and nelfinavir in combination with ZDV and 3TC) in pregnant 
    infected women and their infants will soon start in the U.S., there are 
    currently no data available regarding drug dosage, safety and tolerance 
    of any of the protease inhibitors in pregnancy or in neonates. In mice, 
    indinavir and ritonavir both have significant placental passage; 
    however, in rabbits, indinavir shows little placental passage. Rodent 
    data are not available on placental passage for saquinavir and 
    nelfinavir, and transplacental passage of any of the protease 
    inhibitors in humans is unknown.
        Administration of indinavir to pregnant rodents has revealed no 
    evidence of teratogenicity. However, treatment-related increases in the 
    incidence of supernumerary and cervical ribs were observed in offspring 
    of pregnant rodents receiving indinavir at doses comparable to those 
    administered to humans. In pregnant rats receiving high doses of 
    ritonavir that were associated with maternal toxicity, some 
    developmental toxicity was observed in the offspring, including 
    decreased fetal weight, delayed skeletal ossification, wavy ribs, 
    enlarged fontanelles and cryptorchidism; however, in rabbits, only 
    decreased fetal weight and viability was observed at maternally toxic 
    doses. Rodent studies have not demonstrated embryotoxicity or 
    teratogenicity with saquinavir or nelfinavir.
        Indinavir is associated with infrequent side effects in adults 
    (hyperbilirubinemia and renal stones) that could be problematic for the 
    newborn if transplacental passage occurs and the drug is administered 
    near to delivery. Due to the immature hepatic metabolic enzymes in 
    neonates, the drug would likely have a prolonged half-life and possibly 
    exacerbate the physiologic hyperbilirubinemia observed in neonates. 
    Additionally, due to immature neonatal renal function and the inability 
    of the neonate to voluntarily ensure adequate hydration, high drug 
    concentrations and/or delayed elimination in the neonate could result 
    in a higher risk for drug crystallization and renal stone development 
    than observed in adults. These concerns are theoretical and such 
    effects have not been reported; because the half-life of indinavir in 
    adults is short, these concerns may only be relevant if drug is 
    administered near the time of delivery. Saquinavir, ritonavir and 
    nelfinavir are classified as FDA Pregnancy Category B; indinavir is 
    classified as Category C.
    
    Update on PACTG 076 Results and Other Studies Relevant to ZDV 
    Chemoprophylaxis of Perinatal HIV-1 Transmission
    
        Final results were reported in 1996 for all 419 infants enrolled in 
    PACTG 076. The results are the same as those initially reported in 
    1994; the Kaplan-Meier estimated transmission rate in infants who 
    received placebo was 22.6% compared to 7.6% within those who received 
    ZDV, a 66% reduction in transmission risk.(Sperling 1996)
        The mechanism by which ZDV reduced transmission in PACTG 076 has 
    not been fully defined. The effect of ZDV on maternal HIV-1 RNA did not 
    fully account for the observed efficacy of ZDV in reducing 
    transmission, raising the possibility that pre-exposure prophylaxis of 
    the fetus/infant is an important component of protection. If so, 
    transplacental passage of antiretroviral drugs would be important for 
    prevention of transmission. Additionally, in placental perfusion 
    studies, ZDV has been shown to be metabolized into the active tri-
    phosphate within the placenta (Sandberg 1995, Qian 1994), and this 
    could have provided additional protection against in utero 
    transmission. This phenomenon may be unique to ZDV, as metabolism to 
    the active tri-phosphate form within the placenta has not been observed 
    in the other nucleoside analogues that have been studied in this 
    fashion (ddI and ddC).(Dancis 1993, Sandberg 1994) Development of ZDV-
    resistant virus was not necessarily associated with failure to prevent 
    transmission. In a preliminary evaluation of genotypic resistance in 
    women in PACTG 076, ZDV-resistant virus was present at delivery in only 
    one of 7 transmitting women who had received ZDV and had evaluable 
    samples; this woman had ZDV resistant virus at study entry despite no 
    prior ZDV experience. (Eastman 1997) Additionally, the one woman in 
    whom virus developed ZDV genotypic resistance between entry and 
    delivery in this evaluation did not transmit HIV-1 to her infant.
        No increase in congenital abnormalities compared to the general 
    population was seen in PACTG 076 or observed in evaluation of data from 
    the Antiretroviral Pregnancy Registry.(AntiReg 1997) Follow-up data on 
    uninfected infants from PACTG 076 to a median age of 3.9 years has not 
    shown any differences in growth, neurodevelopment or immunologic status 
    between infants born to mothers who received ZDV compared to those born 
    to mothers who received placebo.(Connor1995) No malignancies have been 
    observed in short-term (up to 6 years of age) follow-up over 734 
    infants from PACTG 076 and natural history studies who had in utero ZDV 
    exposure.(Hanson 1997) However, follow-up is too limited at this time 
    to provide a definitive assessment of carcinogenic risk with human 
    exposure. Long-term follow-up continues to be recommended for all 
    infants with in utero ZDV exposure (or in utero exposure to any of the 
    antiretroviral drugs).
        The effect of temporary administration of ZDV during pregnancy to 
    reduce perinatal transmission on the induction of viral resistance to 
    ZDV and long-term maternal health requires further evaluation. 
    Preliminary data from an interim analysis of PACTG protocol 288 (a 
    study following women enrolled in PACTG 076 through 3 years postpartum) 
    indicate no significant differences at 18 months postpartum in CD4 
    lymphocyte count or clinical status between those women who received 
    ZDV compared to those who received placebo. (Bardeguez 1997) Limited 
    data on the development of genotypic ZDV resistance mutations (codons 
    70 and/or 215) in PACTG 076 are available from a subset of women 
    receiving ZDV, including the majority of those with infected infants. 
    (Eastman 1997) Virus from one of 36 ZDV-receiving women (3%) with 
    paired isolates from entry and delivery developed a ZDV genotypic 
    resistance mutation. However, the population of women in PACTG 076 had 
    very low HIV-1 RNA copy number, and while the
    
    [[Page 36814]]
    
    risk of inducing resistance with administration of ZDV chemoprophylaxis 
    alone for several months during pregnancy was low in this substudy, it 
    would likely be higher in a population of women with more advanced 
    disease and higher levels of viral replication.
        The efficacy of ZDV chemoprophylaxis for reducing transmission 
    among populations of infected women with characteristics unlike those 
    in PACTG 076 has been evaluated in another perinatal protocol (PACTG 
    185) as well as natural history studies. PACTG 185 evaluated the 3-part 
    ZDV regimen combined with passive immunization with hyperimmune HIV-1 
    immunoglobulin (HIVIG), an immunoglobulin containing high levels of 
    antibody to HIV-1, in infected pregnant women with advanced HIV-1 
    disease receiving antiretroviral therapy. Twenty-one percent of the 
    women in this trial had CD4 count <200 m\3\="" and="" 23%="" had="" received="" zdv="" prior="" to="" the="" current="" pregnancy,="" many="" for="" prolonged="" periods="" of="" time.="" all="" women="" and="" infants="" in="" this="" study="" received="" the="" 3-part="" zdv="" regimen,="" and="" were="" randomized="" to="" receive="" hivig="" vs="" standard="" intravenous="" immunoglobulin="" (ivig).="" because="" it="" was="" known="" that="" advanced="" disease="" and="" low="" cd4="" count="" were="" associated="" with="" high="" risk="" for="" perinatal="" transmission,="" it="" was="" hypothesized="" that="" even="" with="" zdv="" chemoprophylaxis,="" the="" perinatal="" transmission="" rate="" would="" be="" 11-15%.="" however,="" at="" the="" first="" interim="" analysis,="" the="" combined="" group="" transmission="" rate="" was="" only="" 4.8%,="" and="" did="" not="" significantly="" differ="" by="" duration="" of="" zdv="" use="" or="" treatment="" arm="" (hivig="" vs="" ivig).(execsum="" 1997)="" enrollment="" was="" halted="" because="" the="" unexpectedly="" low="" transmission="" rate="" resulted="" in="" an="" inability="" to="" answer="" the="" primary="" protocol="" question="" in="" a="" timely="" fashion.="" however,="" the="" results="" of="" the="" trial="" confirm="" the="" efficacy="" of="" zdv="" observed="" in="" pactg="" 076,="" and="" extend="" this="" efficacy="" to="" women="" with="" advanced="" disease,="" low="" cd4="" count="" and="" prior="" zdv="" therapy.="" these="" data="" are="" also="" consistent="" with="" epidemiologic="" data="" from="" several="" natural="" history="" studies.="" in="" a="" study="" in="" connecticut,="" 39%="" of="" women="" with="" cd4="" count=""><200>3 who did not receive ZDV therapy during 
    pregnancy had infected infants compared to 4% of women with similar CD4 
    counts who received ZDV. (Simpson 1997) In North Carolina, perinatal 
    HIV-1 transmission has declined over time from 21% in 1993 to 6% in 
    early 1996; only 3% of women who received all three components of the 
    ZDV regimen had infected infants. (Fiscus 1997) In a large U.S. 
    prospective multicenter natural history cohort of 556 mother-infant 
    pairs, perinatal transmission declined from 19% in infants born before 
    March 1994, before the results of PACTG 076 were available, to 8% in 
    infants born after March 1994; decline in transmission was observed 
    regardless of maternal CD4 lymphocyte count, duration of membrane 
    rupture, mode of delivery, gestational age, and illicit drug use. 
    (Cooper 1996) In another multicenter U.S. cohort, perinatal 
    transmission declined from 20% among 1,160 children born before March 
    1994 to 12% among 373 born afterwards. (Simonds 1996)
        At the present time, there are no clinical trials which demonstrate 
    that antiretroviral drugs other than ZDV are effective in reducing 
    perinatal transmission. Potent combination antiretroviral regimens have 
    been shown to significantly suppress viral replication and improve 
    clinical status in infected adults. However, the efficacy of ZDV 
    exceeds the magnitude of reduction in plasma HIV-1 RNA copy number 
    observed in PACTG 076. If pre-exposure prophylaxis of the infant is an 
    important mechanism of prevention, it is possible that any 
    antiretroviral drug with significant placental passage may be equally 
    effective, although if antiretroviral activity within the placenta is 
    important for protection, ZDV may be unique among the available 
    nucleoside analogue drugs. While there are advantages of combination 
    therapy for the woman's own health, further research is needed before 
    it can be determined if there is an additional advantage to combination 
    antiretroviral therapy for reducing perinatal transmission.
    
    Perinatal HIV-1 Transmission and Maternal HIV-1 RNA Copy Number
    
        The clear correlation of HIV-1 RNA levels with disease progression 
    risk in non-pregnant infected adults suggests that HIV-1 RNA should be 
    monitored during pregnancy at least as often as recommended for non-
    pregnant individuals (e.g., every 3 to 4 months or approximately once 
    each trimester). Whether increased frequency of testing is needed 
    during pregnancy is unclear and requires further study. Although there 
    is no convincing data that pregnancy accelerates HIV-1 disease 
    progression, longitudinal measurements of HIV-1 RNA levels during and 
    after pregnancy have been evaluated in only one prospective cohort to 
    date. In this cohort of 198 HIV-1-infected women, plasma HIV-1 RNA 
    levels were higher at 6 months post partum than ante partum in many 
    women; this increase was observed in women who had received and not 
    received ZDV during pregnancy, as well as in women who continued 
    therapy post partum. (Cao 1997)
        Data on the correlation of viral load with risk of perinatal 
    transmission have been conflicting, with some small studies suggesting 
    an absolute correlation between HIV-1 RNA copy number and transmission 
    risk. (Dickover 1996) However, in several larger studies while higher 
    HIV-1 RNA levels were observed in transmitting women, there was large 
    overlap in HIV-1 RNA copy number between transmitting and non-
    transmitting women, transmission was observed across the entire range 
    of HIV-1 RNA levels (including in women with undetectable HIV-1 RNA), 
    and the positive predictive value of RNA copy number for transmission 
    was relatively low. (Mayaux 1997, Burchett 1996, Cao 1997, Thea 1997) 
    In PACTG 076, there was a relationship between HIV-1 RNA copy number 
    and transmission in women receiving placebo, but in ZDV-receiving women 
    the relationship was markedly attenuated and no longer statistically 
    significant. (Sperling 1996) No HIV-1 RNA threshold below which there 
    was no risk of transmission was identified, and ZDV was effective in 
    reducing transmission regardless of maternal HIV-1 RNA copy number.
        While a general correlation between plasma and genital viral load 
    has been described, women with undetectable plasma HIV-1 RNA levels in 
    whom virus was detectable in the genital tract have been reported. 
    (Rasheed 1996) If exposure to virus in the maternal genital tract 
    during delivery is an important risk factor for perinatal transmission, 
    then plasma HIV-1 RNA levels may not be a fully accurate indicator of 
    risk.
        Whether lowering maternal HIV-1 RNA copy number during pregnancy 
    would reduce perinatal transmission risk requires more study. In a 
    virologic study in 44 infected pregnant women, ZDV was effective in 
    reducing transmission despite minimal effect on HIV-1 RNA levels, 
    similar to what was observed in PACTG 076. (Melvin 1997) However, it is 
    not known if a more potent antiretroviral regimen that more 
    significantly suppresses viral replication would be associated with 
    enhanced efficacy in reducing transmission risk over and above that 
    observed with ZDV alone. At the present time, determination of HIV-1 
    copy number is important for decisions related to treatment. However, 
    because ZDV benefit is observed regardless of maternal HIV-1 RNA level 
    and because transmission may occur when HIV-1 RNA is not detectable, 
    HIV-1 RNA
    
    [[Page 36815]]
    
    should not be the determining factor in decisions regarding use of ZDV 
    chemoprophylaxis against perinatal transmission.
    
    General Principles Regarding Use of Antiretrovirals in Pregnancy
    
        Care of the HIV-1-infected pregnant woman should involve a 
    collaboration between the HIV-specialist caring for the woman when she 
    is not pregnant, her obstetrician, and the woman herself. Decisions 
    regarding use of antiretroviral drugs during pregnancy should be made 
    by the woman following discussion with her health care provider of the 
    known and unknown benefits and risks of therapy. Initial evaluation of 
    an infected pregnant woman should include an assessment of HIV-1 
    disease status and recommendations regarding antiretroviral treatment 
    or alteration of her current antiretroviral regimen. This assessment 
    should include evaluation of the degree of existing immunodeficiency 
    determined by CD4 count; risk of disease progression determined by the 
    level of plasma RNA; history of prior or current antiretroviral 
    therapy; and gestational age. For those women not currently receiving 
    antiretroviral therapy, decision-making regarding initiation of therapy 
    should be the same as for non-pregnant individuals, with the additional 
    consideration of the potential impact of such therapy on the fetus and 
    infant. (PanelRec 1997) Similarly, for women currently receiving 
    antiretrovirals, decisions regarding alterations in therapy should use 
    the same parameters as for non-pregnant individuals. Additionally, use 
    of the 3-part ZDV chemoprophylaxis regimen, alone or in combination 
    with other antiretrovirals, should be discussed with and offered to all 
    infected pregnant women for the purpose of reducing perinatal 
    transmission risk.
        Decisions regarding the use and choice of antiretroviral drugs 
    during pregnancy are complex and must balance a number of competing 
    factors influencing risk and benefit. Discussion regarding use of 
    antiretroviral drugs during pregnancy should include what is known and 
    not known about the effects of such drugs on the fetus and newborn, 
    including lack of long-term outcome data on use of any of the available 
    antiretroviral drugs in pregnancy; what would be recommended in terms 
    of treatment for her own health; and the efficacy of ZDV for reduction 
    of perinatal transmission. These discussions should include what is 
    known from preclinical and animal studies and available clinical 
    information about use of the various antiretroviral agents during 
    pregnancy. It is important to place the hypothetical risks of these 
    drugs during pregnancy in perspective to the proven benefit of 
    antiretroviral therapy for her own health and ZDV chemoprophylaxis for 
    reducing the risk of HIV-1 transmission to her infant.
        Discussion of treatment options should be noncoercive, and the 
    final decision regarding the use of antiretroviral drugs is the 
    responsibility of the woman. Decisions regarding use and choice of 
    antiretroviral drugs in non-pregnant individuals are becoming 
    increasingly complicated, as the standard of care moves toward 
    simultaneous use of multiple antiretroviral drugs to suppress viral 
    replication below detectable limits. These decisions are further 
    complicated in pregnancy, as the long-term consequences of in utero 
    exposure to antiretroviral drugs, alone or in combination, for the 
    infant are unknown. A decision to not accept treatment with ZDV or 
    other drugs should not result in punitive action or denial of care, nor 
    should use of ZDV be denied to a woman who wishes to minimize exposure 
    of the fetus to other antiretroviral drugs and therefore chooses to 
    receive only ZDV during pregnancy to reduce the risk of perinatal 
    transmission after receiving appropriate counseling.
        A long-term treatment plan should be developed with the patient and 
    the importance of adherence to any prescribed antiretroviral regimen 
    discussed with her. Depending on individual circumstances, provision of 
    support services, drug treatment, and coordination of services between 
    the criminal justice system, drug treatment programs and prenatal care 
    providers may each play an important role in assisting women with 
    adherence to antiretroviral regimens.
        Public Health Service recommendations for infected women in the 
    U.S. to refrain from breastfeeding to avoid postnatal transmission of 
    HIV-1 to their infants through breast milk should not be altered for 
    women receiving antiretroviral therapy. (CDC 1985, CDC 1995) Passage of 
    antiretroviral drugs into breast milk has been evaluated for only a few 
    antiretroviral drugs: ZDV, 3TC and nevirapine can be detected in the 
    breast milk of women receiving the drugs, and ddI, d4T, and indinavir 
    can be detected in the breast milk of lactating rats receiving therapy. 
    The efficacy of antiretroviral therapy for prevention of postnatal 
    transmission of HIV-1 through breast milk and the toxicity of chronic 
    antiretroviral exposure of the infant via breast milk are unknown.
        It is strongly recommended that health care providers who are 
    treating HIV-1-infected pregnant women report cases of prenatal 
    exposure to ZDV, ddI, ddC, d4T, 3TC, saquinavir or indinavir alone or 
    in combination to the Antiretroviral Pregnancy Registry. The registry 
    is an epidemiologic project to collect observational, non-experimental 
    data on antiretroviral exposure during pregnancy for the purpose of 
    assessing potential teratogenicity of these drugs in pregnancy. 
    Registry data will be used to supplement animal toxicology studies and 
    assist clinicians in weighing the potential risks and benefits of 
    treatment for individual patients.
        The registry is a collaborative project jointly managed by Glaxo 
    Wellcome, Hoffmann-LaRoche Inc., Bristol-Myers Squibb Co., and Merck & 
    Co. Inc., with an advisory committee of practitioners and CDC and NIH 
    staff; it is anticipated that additional antiretroviral drugs will be 
    added to the registry in the future. The registry does not use patient 
    names, and birth outcome follow-up is obtained by registry staff from 
    the reporting physician. Referrals should be directed to Antiretroviral 
    Pregnancy Registry, Post Office Box 13398, Research Triangle Park, NC 
    27709-3398; telephone (919) 483-9437 or (800) 722-9292, ext. 39437; fax 
    919-315-8981.
    
    Recommendations for Antiretroviral Chemoprophylaxis to Reduce Perinatal 
    HIV Transmission
    
        The following recommendations for use of antiretroviral 
    chemoprophylaxis to reduce the risk of perinatal transmission are based 
    upon various circumstances that may be commonly encountered in clinical 
    practice (Table 3), with relevant considerations highlighted in the 
    subsequent discussion section. These scenarios present only 
    recommendations and flexibility should be exercised according to the 
    circumstances of the individual patient. In the 1994 recommendations, 6 
    clinical scenarios were delineated based on maternal CD4 count, 
    gestational age and prior antiretroviral use. Because current data 
    indicate that the PACTG 076 ZDV regimen is also effective women with 
    advanced disease, low CD4 count and prior ZDV therapy, clinical 
    scenarios by CD4 count and prior ZDV use are not presented. 
    Additionally, because current data indicate most transmission occurs 
    near to or during delivery, it was felt that ZDV chemoprophylaxis 
    should be recommended regardless of gestational age; thus, clinical 
    scenarios by gestational age are also not presented.
    
    [[Page 36816]]
    
        Table 1 shows the ZDV dosage and regimen used in PACTG 076. The 
    antenatal dosing regimen in PACTG 076 (100 mg orally five times daily) 
    was selected based on standard ZDV dosage for adults at the time of the 
    study. Recent reports from several laboratories have demonstrated that 
    administration of ZDV three times a day will maintain intracellular ZDV 
    tri-phosphate at levels comparable to that observed with more frequent 
    dosing. (Rodman 1996; Barry 1996; Gambertoglio 1996) Additionally, 
    comparable clinical response with twice daily dosing has been observed 
    in some clinical trials. (Mulder 1994, Mannucci 1994, Cooper 1993) 
    Thus, the current standard adult ZDV dosing regimen is 200 mg three 
    times daily or 300 mg twice daily. Because the mechanism by which ZDV 
    reduces perinatal transmission is not known, it cannot be known with 
    certainty that these dosing regimens will have equivalent efficacy to 
    that observed in PACTG 076. However, it would be anticipated that a two 
    or three times daily regimen might be associated with enhanced maternal 
    adherence over a five times daily regimen.
        The recommended ZDV dosage for infants was derived from 
    pharmacokinetic studies performed in term infants. (Boucher 1993) ZDV 
    is primarily cleared through hepatic glucuronidation to an inactive 
    metabolite. The glucuronidation metabolic enzyme system is immature in 
    neonates, leading to prolonged ZDV half-life and clearance compared to 
    older infants (ZDV half-life, 3.1 hours vs 1.9 hours, and clearance, 
    10.9 vs 19.0 mL per minute per kg body weight, respectively). Because 
    premature infants have even greater immaturity in hepatic metabolic 
    function than term infants, further prolongation in clearance may be 
    expected. In a small pharmacokinetic study of 7 premature infants who 
    were 28 to 33 weeks gestation and received a variety of ZDV dosing 
    regimens, mean ZDV half-life was 6.3 hours and mean clearance was 2.8 
    mL per minute per kg body weight during the first 10 days of life. 
    (Capparelli 1996) Appropriate ZDV dosing for premature infants has not 
    been defined, but is being evaluated in a phase I clinical trial in 
    premature infants less than 34 weeks gestation. The dosing regimen 
    being studied is 1.5 mg per kg body weight orally or intravenously 
    every 12 hours for the first 2 weeks of life; from 2 to 6 weeks of age, 
    the dose is increased to 2 mg per kg body weight every 8 hours.
        Because subtherapeutic dosing of antiretroviral drugs may be 
    associated with enhancing the likelihood for the development of drug 
    resistance, women who must temporarily discontinue therapy due to 
    pregnancy-related hyperemesis should not reinstitute therapy until 
    sufficient time has elapsed to assure that the drugs will be tolerated. 
    In order to reduce the potential for emergence of resistance, if 
    therapy requires temporary discontinuation for any reason during 
    pregnancy, all drugs should be stopped and reintroduced simultaneously.
    
    Clinical Scenarios
    
    Scenario #1
    
    HIV-Infected Pregnant Women Without Prior Antiretroviral Therapy
        Recommendation: HIV-1 infected pregnant women must receive standard 
    clinical, immunologic and virologic evaluation, and recommendations for 
    initiation and choice of antiretroviral therapy should be based on the 
    same parameters used in non-pregnant individuals, with consideration 
    and discussion of the known and unknown risks and benefits of such 
    therapy during pregnancy.
        The 3-part ZDV chemoprophylaxis regimen should be recommended for 
    all HIV-infected pregnant women to reduce the risk of perinatal 
    transmission. If the woman's clinical, immunologic and virologic status 
    indicates that more aggressive therapy is recommended to treat her 
    infection (Panelrec, 1997), other antiretroviral drugs should be 
    recommended in addition to ZDV. If the woman's status is such that 
    therapy would be considered optional, the use of additional 
    antiretrovirals may be offered, although whether this will provide 
    additional benefit to the woman or her child is not known. Women who 
    are in the first trimester of pregnancy may wish to consider delaying 
    initiation of therapy at least until after 10 to 12 weeks gestation.
        Discussion: The only drug that has been shown to reduce the risk of 
    perinatal HIV-1 transmission is ZDV when administered in the 3-part 
    PACTG 076 regimen; this regimen was shown to reduce transmission risk 
    by approximately 70%. The mechanism by which ZDV reduced transmission 
    is not known, and there are insufficient data available at present to 
    justify the substitution of any antiretroviral drug other than ZDV for 
    the purpose of reducing perinatal transmission. Therefore, if 
    combination antiretroviral therapy is initiated during pregnancy, it is 
    recommended that ZDV be included as a component of antenatal therapy 
    and the intrapartum and newborn ZDV parts of the chemoprophylactic 
    regimen should be recommended for the specific purpose of reducing 
    perinatal transmission.
        Women should be counseled that combination therapy may have 
    significant benefit for their own health but is of unknown benefit to 
    the fetus. Potent combination antiretroviral regimens may be shown in 
    the future to provide enhanced protection against perinatal 
    transmission, but this benefit is not yet proven. Decisions regarding 
    the use and choice of an antiretroviral regimen will need to be 
    individualized based on discussion with the woman about her risk for 
    disease progression and the risks and benefits of delaying initiation 
    of therapy; potential drug toxicities and interactions with other 
    drugs; the need for adherence to the prescribed drug schedule; and 
    preclinical, animal and clinical data relevant to use of the currently 
    available antiretrovirals during pregnancy.
        Because the period of organogenesis when the embryo is most 
    susceptible to potential teratogenic effects of drugs is the first 10 
    weeks of gestation and the risks of antiretroviral therapy during that 
    period are unknown, women who are in the first trimester of pregnancy 
    may wish to consider delaying initiation of therapy until after 10 to 
    12 weeks gestation. This decision should be carefully considered and 
    discussed between the health care provider and the patient, including 
    an assessment of the woman's health status and the benefits and risks 
    of delaying initiation of therapy for several weeks.
        Women for whom initiation of antiretroviral therapy for the 
    treatment of their HIV infection would be considered optional (eg. high 
    CD4 count and low or undetectable RNA copy number) should have the 
    potential benefits of standard combination therapy discussed with them 
    and standard therapy, including the 3-part ZDV chemoprophylaxis 
    regimen, offered to them. Some women may wish to restrict their 
    exposure to antiretroviral drugs during pregnancy but still wish to 
    reduce the risk of transmitting HIV-1 to their infant; the 3-part ZDV 
    chemoprophylaxis regimen should be recommended in this situation. In 
    these circumstances, the development of resistance should be minimized 
    by the limited viral replication in the patient and the time-limited 
    exposure to ZDV.
        Because ZDV alone does not suppress HIV replication to undetectable 
    levels, there are theoretical concerns that use of ZDV chemoprophylaxis 
    alone might select for ZDV resistant viral variants which might limit 
    future ability to favorable response to combination antiretroviral 
    regimens that include
    
    [[Page 36817]]
    
    ZDV. There are currently insufficient data to determine if such use 
    would have adverse consequences for the woman postpartum. In some adult 
    combination antiretroviral clinical trials, patients with previous ZDV 
    therapy experienced less benefit from combination therapy than those 
    who were antiretroviral naive. (Delta 1996, Hammer 1996, Saravolatz 
    1996) However, the median duration of prior ZDV in these studies was 12 
    to 20 months and enrolled patients had more advanced disease and lower 
    CD4 counts than the population of women enrolled in PACTG 076 or for 
    whom initiation of therapy would be considered optional. In one study, 
    patients with less than 12 months of ZDV responded as favorably to 
    combination therapy as did those without prior ZDV therapy.(Saravolatz 
    1996) In PACTG 076, the median duration of ZDV therapy was 11 weeks, 
    and the maximal duration of ZDV begun at 14 weeks gestation would be 
    6.5 months for a full-term pregnancy.
        However, for women initiating therapy who have more advanced 
    disease, concerns about development of resistance with use of ZDV alone 
    as chemoprophylaxis during pregnancy would be greater. Factors that 
    predict more rapid development of ZDV resistance include more advanced 
    HIV-1 disease, low CD4 count, high HIV-1 RNA copy number, and possibly 
    syncytium-inducing viral phenotype.(Kuritzkes 1996, Japour 1995) 
    Therefore, women with advanced disease, low CD4 count or high RNA copy 
    number should be counseled that therapy with a combination 
    antiretroviral regimen that includes ZDV for reducing transmission risk 
    would be more optimal for their own health than use of ZDV 
    chemoprophylaxis alone.
    
    Scenario #2
    
    HIV-Infected Women Receiving Antiretroviral Therapy During the Current 
    Pregnancy
        Recommendation: HIV-1 infected women receiving antiretroviral 
    therapy in whom pregnancy is identified after the first trimester 
    should continue therapy. For women receiving antiretroviral therapy in 
    whom pregnancy is recognized during the first trimester, the woman 
    should be counseled regarding the benefits and potential risks of 
    antiretroviral administration during this period, and continuation of 
    therapy should be considered. If therapy is discontinued during the 
    first trimester, all drugs should be stopped and reintroduced 
    simultaneously to avoid the development of resistance. If the current 
    therapeutic regimen does not contain ZDV, the addition of ZDV or 
    substitution of ZDV for another nucleoside analogue antiretroviral is 
    recommended after 14 weeks gestation. Intrapartum and newborn ZDV 
    administration is recommended regardless of the antepartum 
    antiretroviral regimen.
        Discussion: Women who require antiretroviral treatment for their 
    HIV infection should continue treatment during pregnancy. 
    Discontinuation of therapy could lead to rebound in viral load, which 
    theoretically could result in decline in immune status and/or disease 
    progression, all of which might have adverse consequences for the fetus 
    as well as the woman. Because the efficacy of non-ZDV containing 
    antiretroviral regimens for reduction of perinatal transmission is 
    unknown, it is recommended that ZDV be a component of the antenatal 
    antiretroviral treatment regimen after 14 weeks gestation, and that 
    intrapartum and newborn ZDV be administered. If a woman does not 
    receive ZDV as a component of her antepartum antiretroviral regimen 
    (eg. because of prior history of ZDV-related severe toxicity or 
    personal choice), intrapartum and newborn ZDV should continue to be 
    recommended.
        Some women receiving antiretroviral therapy may recognize their 
    pregnancy early in gestation, and concern for potential teratogenicity 
    may lead some to consider temporarily stopping antiretroviral treatment 
    until after the first trimester. There are insufficient data to support 
    or refute the teratogenic risk of antiretroviral drugs when 
    administered during the first 10 weeks of gestation. The decision to 
    discontinue therapy during the first trimester should be carefully 
    considered and discussed between the clinician and the woman. 
    Considerations include gestational age of the pregnancy, the woman's 
    clinical, immunologic and virologic status, and what is known and not 
    known about the potential effects of the antiretroviral drugs on the 
    fetus. If antiretroviral therapy is discontinued during the first 
    trimester, all agents should be stopped and restarted in the second 
    trimester simultaneously to avoid the development of resistance. There 
    are currently no data to address whether transient discontinuation of 
    therapy in this manner would be harmful for the woman and/or fetus.
        The impact of prior antiretroviral exposure on the efficacy of ZDV 
    chemoprophylaxis is unclear. Data from PACTG 185 indicate that duration 
    of prior ZDV therapy in women with advanced HIV-1 disease, many of whom 
    received prolonged ZDV prior to pregnancy, did not appear to be 
    associated with diminished ZDV efficacy for reduction of transmission: 
    perinatal transmission rates were similar among women who first 
    initiated ZDV during pregnancy and women who had received ZDV prior to 
    pregnancy. Thus at the present time, a history of ZDV therapy prior to 
    the current pregnancy should not limit recommendations for 
    administration of ZDV chemoprophylaxis to reduce perinatal 
    transmission.
        Some experts might consider administration of ZDV in combination 
    with other antiretroviral drugs to newborns of women with a history of 
    prior antiretroviral therapy, particularly in situations where the 
    woman is infected with HIV-1 with documented high-level ZDV resistance, 
    had disease progression while receiving ZDV, or had extensive prior ZDV 
    monotherapy. However, the efficacy of this approach is not known. The 
    appropriate dose and short and long-term safety for most antiretroviral 
    agents other than ZDV are not defined for neonates. Because of immature 
    liver metabolism and renal function, the half-life of many drugs 
    (including ZDV, 3TC and nevirapine) is prolonged during the neonatal 
    period, requiring specific dosing adjustments. Phase I studies of a 
    number of other antiretroviral drugs in neonates are ongoing, but data 
    are not yet available. The infected woman should be counseled regarding 
    the postulated benefit of combination antiretroviral drugs in the 
    neonate and the potential risks, what is known about appropriate dosing 
    of the drugs in newborn infants, and that use of additional 
    antiretroviral drugs for newborn prophylaxis is of unknown efficacy for 
    reducing perinatal transmission risk.
    
    Scenario #3
    
    HIV-Infected Women in Labor Who Have Had no Prior Therapy
        Recommendation: Administration of intrapartum intravenous ZDV 
    should be recommended along with the 6 week newborn ZDV regimen. In the 
    immediate postpartum period, the woman should have appropriate 
    assessments (eg., CD4 count, HIV-1 RNA copy number) to determine if 
    antiretroviral therapy is recommended for her own health.
        Discussion: Intrapartum ZDV will not prevent the portion of 
    perinatal transmission that occurs prior to labor. Therefore, the 
    efficacy of an intrapartum/newborn antiretroviral regimen in reducing 
    perinatal transmission is likely to be less than the
    
    [[Page 36818]]
    
    efficacy observed in PACTG 076. However, increasing data indicate that 
    a majority of perinatal transmission occurs near to or during birth. 
    Additionally, the efficacy of ZDV in reducing perinatal transmission is 
    not primarily related to treatment-induced reduction in maternal HIV-1 
    RNA copy number. This implies that the presence of systemic 
    antiretroviral drug levels in the neonate just prior to, during and for 
    a period following birth may be a critical component for reducing 
    transmission.
        There are minimal data to address the efficacy of a regimen that 
    lacks the antenatal ZDV component. An epidemiologic study from North 
    Carolina compared perinatal transmission rates from mother-infant pairs 
    who received different parts of the ZDV chemoprophylactic regimen. 
    (Fiscus 1997) Among those who received all 3 components, 6 of 188 
    infants were infected (3%). While the numbers were small, only one of 
    16 infants (6%) were infected among those who received intrapartum and 
    newborn ZDV.
        ZDV readily crosses the placenta. Administration of the intravenous 
    ZDV loading dose followed by continuous ZDV infusion during labor to 
    the woman will provide ZDV levels in the newborn during passage through 
    the birth canal that are nearly equivalent to maternal ZDV levels. The 
    initial intravenous ZDV loading dose assures rapid attainment of 
    virucidal ZDV levels in the woman and her infant, and the continuous 
    ZDV infusion assures stable drug levels in the infant during the birth 
    process regardless of the duration of labor. A study is currently 
    ongoing in the U.S. to evaluate if oral dosing of ZDV during labor in a 
    regimen of 300 mg orally every 3 hours would provide equivalent infant 
    drug exposure to intravenous ZDV administration. Until this data is 
    available, oral intrapartum administration of ZDV cannot be assumed to 
    be equivalent to the intravenous intrapartum ZDV.
        ZDV administered both during the intrapartum period and to the 
    newborn provides both pre-and post-exposure prophylaxis to the infant. 
    Some clinicians might consider administration of ZDV in combination 
    with other antiretroviral drugs to the newborn, analogous to 
    recommendations for post-exposure prophylaxis of nosocomial HIV-1 
    exposure. (CDC 1996) Any decision to use combination antiretroviral 
    prophylaxis in the newborn must be accompanied by a discussion with the 
    woman of potential benefits and risks and that there currently are no 
    data to address the efficacy and safety of this approach.
    
    Scenario #4
    
    Infants Born to Mothers Who Have Received No Antiretroviral Therapy 
    During Pregnancy or Intrapartum
        Recommendation: The 6 week neonatal ZDV component of the ZDV 
    chemoprophylactic regimen should be discussed with the mother and 
    offered for the newborn; ZDV should be initiated as soon as possible 
    after birth, preferably within 12-24 hours after birth. Some clinicians 
    may choose to use ZDV in combination with other antiretroviral drugs, 
    particularly if the mother has known or suspected ZDV-resistant virus. 
    However, the efficacy of this approach is unknown and appropriate 
    dosing regimens for neonates are incompletely defined. In the immediate 
    postpartum period, the woman should undergo appropriate assessments 
    (e.g., CD4 count, HIV-1 RNA copy number) to determine if antiretroviral 
    therapy is required for her own health.
        Discussion: Definitive data are not available to address whether 
    ZDV administered solely during the neonatal period would reduce the 
    risk of perinatal transmission. However, data from a case-control study 
    of post-exposure prophylaxis of health care workers who had nosocomial 
    percutaneous exposure to blood from HIV-1-infected individuals indicate 
    that ZDV administration was associated with a 79% reduction in the risk 
    for HIV-1 seroconversion following exposure. (CDC 1995) Post-exposure 
    prophylaxis has also been shown to prevent retroviral infection in some 
    animal studies. (Van Rompay 1995, Tsai 1995, Bottiger 1997)
        The interval for which benefit may be gained from post-exposure 
    prophylaxis is undefined, but data from animal studies indicate that 
    the longer the delay in institution of prophylaxis, the less likely 
    prevention will be observed. In most animal studies, antiretroviral 
    prophylaxis initiated after 24-36 hours is usually not effective for 
    preventing infection, although later administration has been associated 
    with decreased viremia in ultimately infected animals in some cases. 
    (VanRompay 1995, Bottiger 1997, Tsai 1995) In the feline leukemia virus 
    cat model, ZDV treatment initiated within the first 4 days after viral 
    challenge afforded protection, while treatment initiated one week 
    postexposure did not prevent infection. (Mathes 1992) The relevance of 
    the animal studies to prevention of perinatal transmission in humans is 
    unknown. HIV-1 infection is established in the majority of infected 
    infants by 1 to 2 weeks of age. In a study of 271 infected infants, 
    HIV-1 DNA polymerase chain reaction (PCR) was positive in 38% of 
    infected infants tested within 48 hours of birth. No major change in 
    diagnostic sensitivity was observed over the first week of life, but 
    detection rose rapidly during the second week of life, reaching 93% by 
    14 days of age. (Dunn 1995) Therefore, it would be unlikely that 
    initiation of post-exposure prophylaxis after 14 days of age would have 
    efficacy in preventing transmission, as infection would already be 
    established in most children.
        Recommendations have been made for antiretroviral post-exposure 
    prophylaxis of nosocomial HIV-1 exposure. It was recommended that ZDV 
    be administered as soon after exposure as possible, and the addition of 
    3TC was recommended in most cases to provide increased antiretroviral 
    activity and presumed activity against ZDV-resistant HIV-1 strains. 
    (CDC 1996) The addition of a protease inhibitor was recommended for 
    particularly high-risk exposures. There are no data to address whether 
    the addition of other antiretroviral drugs to ZDV increase the 
    effectiveness of post-exposure prophylaxis. However, some clinicians 
    may wish to provide ZDV in combination with one or more other 
    antiretroviral agents in situations in which only post-exposure newborn 
    prophylaxis is administered. Such a decision must be accompanied by a 
    discussion with the woman of potential benefits and risks of this 
    approach.
    
    Recommendations for Monitoring of Women and Their Infants
    
    Pregnant Woman and Fetus
    
        HIV-1-infected pregnant women should be monitored in the same 
    fashion that nonpregnant individuals are monitored. This should include 
    measurement of CD4 lymphocyte count and HIV-1 RNA levels approximately 
    every trimester (every 3 to 4 months) to determine need for 
    antiretroviral therapy of maternal HIV-1 disease or alterations in such 
    therapy, and/or initiation of prophylaxis against Pneumocystis carinii 
    pneumonia. Some studies have found that changes in absolute CD4 count 
    during pregnancy may reflect the physiologic changes of pregnancy on 
    hemodynamic parameters and blood volume as opposed to a longterm 
    influence of pregnancy upon CD4 count; CD4 percent appears to be more 
    stable and may be a more accurate reflection of immune status during 
    pregnancy. (Miotti 1992, Tuomala 1997)
    
    [[Page 36819]]
    
    Long-range plans should be developed with the woman regarding 
    continuity of medical care and antiretroviral therapy for her own 
    health after she delivers her infant.
        Monitoring for potential complications of antiretroviral 
    administration during pregnancy should take into account what is known 
    about the side effects of the drugs the woman is receiving. For 
    example, routine hematologic and liver chemistry monitoring is 
    recommended for women receiving ZDV. Because there is less experience 
    with use of combination antiretroviral regimens during pregnancy, more 
    intensive monitoring may be warranted for women receiving drugs other 
    than or in addition to ZDV.
        Antepartum fetal monitoring for women who receive only ZDV 
    chemoprophylaxis should be performed as clinically indicated, as the 
    available data do not indicate that ZDV use in pregnancy is associated 
    with increased risk for fetal complications. However, much less is 
    known about the effect of combination antiretroviral therapy during 
    pregnancy on the fetus. More intensive monitoring should be considered, 
    including assessment of fetal anatomy with a level II ultrasound and 
    continued assessment of fetal growth and well-being during the third 
    trimester.
    
    Neonate
    
        A complete blood count and differential should be performed as a 
    baseline evaluation prior to administration of ZDV. Anemia has been the 
    primary complication of the 6 week ZDV regimen in the neonate, thus at 
    a minimum, repeat measurement of hemoglobin is required at the 
    completion of the 6 week ZDV regimen; repeat measurement may be 
    performed at 12 weeks of age, by which any ZDV-related hematologic 
    toxicity should be resolved. Infants who have anemia at birth or who 
    are premature warrant more intensive monitoring.
        There is little experience with potential toxicities in infants 
    whose mothers have received combination antiretroviral therapy. More 
    intensive monitoring of hematologic and chemistry measurements during 
    the first few weeks of life would be advised in these infants.
        All infants born to HIV-1-infected women should be placed on 
    prophylaxis to prevent Pneumocystis carinii pneumonia at 6 weeks of 
    age, following completion of the ZDV prophylaxis regimen. (CDC 1995) 
    Monitoring and diagnostic evaluation of HIV-1-exposed infants should 
    follow current standards of care. The available data do not indicate 
    any delay in HIV-1 diagnosis in infants who have received the ZDV 
    regimen. (Connor 1994, Kovacs 1995) However, the effect of combination 
    antiretroviral therapy in the mother and/or newborn on the sensitivity 
    of infant virologic diagnostic testing is unknown. Infants with 
    negative virologic tests during the first 6 weeks of life should have 
    diagnostic evaluation repeated after completion of the neonatal 
    antiretroviral prophylaxis regimen.
    
    Postpartum Follow-Up of Women
    
        Comprehensive care and support services are required for women 
    infected with HIV-1 and their families. Components of comprehensive 
    care include the full range of medical care services including family 
    planning and drug treatment; coordination of care for the woman, her 
    children and other family members; support services such as case 
    management and childcare; assistance with basic life needs such as 
    housing, food, and transportation; and legal and advocacy services. 
    This care should begin prior to pregnancy, with continuity of care 
    ensured throughout pregnancy and postpartum.
        Maternal medical services during the postpartum period must be 
    coordinated between obstetric and HIV-specialist health care providers. 
    Continuity of antiretroviral treatment when therapy is required for 
    treatment of the woman's HIV infection is especially critical and must 
    be assured. All women should have linkage with comprehensive health 
    care services for her own medical care and for assistance with family 
    planning and contraception.
        Data from PACTG Protocols 076 and 288 do not indicate adverse 
    effects through 18 months postpartum among women who received ZDV 
    during pregnancy; however, continued clinical, immunologic and 
    virologic follow-up of these women is ongoing. Women who have received 
    only ZDV chemoprophylaxis during pregnancy should receive appropriate 
    evaluation to determine the need for antiretroviral therapy in the 
    postpartum period.
    
    Long-Term Follow-Up of Infants
    
        Data remain insufficient to address the effect that exposure to ZDV 
    or other antiretroviral agents in utero might have on long-term risk 
    for neoplasia or organ system toxicities in children. Data from follow-
    up of PACTG 076 infants through 18 to 36 months of age do not indicate 
    any differences in immunologic, neurologic and growth parameters 
    between infants who were exposed to the ZDV regimen compared to 
    placebo; continued intensive follow-up through PACTG 219 is ongoing. 
    PACTG 219 will also provide intensive follow-up for infants born to 
    women who receive other antiretroviral drugs as part of PACTG perinatal 
    protocols, so some data regarding follow-up of exposure to other 
    antiretroviral agents alone or in combination will be available in the 
    future.
        Innovative methods are needed to provide follow-up to infants with 
    in utero exposure to ZDV or any other antiretrovirals outside of PACTG 
    protocols. Information regarding such exposure should be part of the 
    ongoing medical record of the child, particularly for uninfected 
    children. Follow-up of children with antiretroviral exposure should 
    continue into adulthood because of the theoretical concerns regarding 
    potential for carcinogenicity of the nucleoside analogue antiretroviral 
    drugs. Long-term follow-up should include at least yearly physical 
    examination of all antiretroviral-exposed children, and for older 
    adolescent females, gynecologic evaluation with pap smears.
        On a population basis, HIV-1 surveillance databases from states 
    that require HIV-1 reporting provide an opportunity to collect 
    information on in utero antiretroviral exposure. To the extent 
    permitted by federal law and regulations, these confidential registries 
    can be used to compare to birth defect and cancer registries to look 
    for potential adverse outcomes.
    
    Future Research Needs
    
        An increasing number of HIV-1-infected women will be receiving 
    antiretroviral therapy for their own health during pregnancy. 
    Preclinical evaluations of antiretroviral drugs for potential 
    pregnancy- and fetal-related toxicities should be completed for all 
    current and new antiretroviral drugs. More data are needed regarding 
    the safety and pharmacokinetics of antiretroviral drugs during 
    pregnancy and in the neonate, particularly when used in combination 
    regimens. Results from a number of phase I studies will be available in 
    the next year which will assist in delineating appropriate dosing and 
    provide data on short-term safety of these drugs in pregnant women and 
    infants. However, the long-term consequences of in utero antiretroviral 
    exposure for the infant is unknown, and mechanisms must be developed to 
    gather information about the long-term outcome for exposed infants. 
    Innovative methods are needed to enable identification and follow-up of 
    populations of children with in utero antiretroviral exposure.
    
    [[Page 36820]]
    
        Additional studies are needed to determine the long-term 
    consequences of transient use of ZDV chemoprophylaxis during pregnancy 
    for women who do not desire to receive combination therapy antenatally, 
    including the risk for development of ZDV-resistance.
        While there are theoretical reasons to believe that more potent 
    antiretroviral combination regimens that dramatically diminish viral 
    load may also prevent perinatal transmission, there are currently no 
    data to address this hypothesis. The efficacy of combination 
    antiretroviral therapy specifically to decrease the risk of perinatal 
    HIV-1 transmission needs to be evaluated in ongoing and future 
    perinatal clinical trials. Additionally, epidemiologic studies and 
    clinical trials are needed to delineate the relative efficacy of the 
    various components of the 3-part ZDV chemoprophylactic regimen. 
    Improved understanding of the factors associated with perinatal 
    transmission despite ZDV chemoprophylaxis is needed in order to develop 
    alternative effective regimens. Because of the dramatic decline in 
    perinatal HIV-1 transmission with widespread implementation of ZDV 
    chemoprophylaxis, the conduct of such epidemiologic studies and 
    clinical trials requires an international collaborative effort.
        Additionally, regimens that are more feasible for implementation in 
    the developing world are urgently needed. The 3-part ZDV 
    chemoprophylactic regimen is complex and may not be a feasible option 
    for many developing countries: most pregnant women show up in health 
    care systems only around the time of delivery; widespread safe 
    administration of intravenous ZDV infusions during labor may not be 
    possible; and the cost of the regimen may be prohibitive and many times 
    greater than the per capita health expenditures for the country. There 
    are several ongoing studies in developing countries that are evaluating 
    the efficacy of more practical, abbreviated modifications of the ZDV 
    regimen. Additionally, a number of non-antiretroviral interventions are 
    also under study. Results of these studies will be available in the 
    next few years.
    
    References
    
    Centers for Disease Control. Public Health Service Task Force on Use 
    of Zidovudine to Reduce Perinatal Transmission of Human 
    Immunodeficiency Virus. MMWR; 1994;43 (RR-11):1-21.
    Connor EM, Sperling RS, Gelber R, et al. Reduction of maternal-
    infant transmission of human immunodeficiency virus type 1 with 
    zidovudine treatment. N Engl J Med 1994; 331:1173-80.
    Centers for Disease Control. U.S. Public Health Service 
    recommendations for human immunodeficiency virus counseling and 
    voluntary testing for pregnant women. MMWR 1995; 44 (RR-7):1-14.
    Cooper ER, Nugent RP, Diaz C, et al. After AIDS Clinical Trial 076: 
    the changing pattern of zidovudine use during pregnancy, and the 
    subsequent reduction in vertical transmission of human 
    immunodeficiency virus in a cohort of infected women and their 
    infants. J Infect Dis 1996; 174:1207-11.
    Fiscus SA, Adimora AA, Schoenbach VJ, et al. Perinatal HIV infection 
    and the effect of zidovudine therapy on transmission in rural and 
    urban counties. JAMA 1996; 275:1483-8.
    Fiscus SA, Adimora AA, Schoenbach VJ, et al. Importance of maternal 
    ZDV therapy in the reduction of perinatal transmission of HIV. 
    Proceedings from the Fourth Conference on Retroviruses and 
    Opportunistic Infections, Washington. D.C.; January 22-26, 1997: 176 
    (Abstract 379).
    Thomas P, Singh T, Bornschlegel K, et al. Use of ZDV to prevent 
    perinatal HIV in New York City (NYC). Proceedings from the Fourth 
    Conference on Retroviruses and Opportunistic Infections, Washington. 
    D.C.; January 22-26, 1997: 176 (Abstract 381).
    Blanche S, Mayaux MJ, Mandelbrot L, et al. Acceptability and impact 
    of zidovudine prevention on mother-to-child transmission in France. 
    Proceedings from the Fourth Conference on Retroviruses and 
    Opportunistic Infections, Washington. D.C.; January 22-26, 1997:176 
    (Abstract 380).
    Simonds RJ, Nesheim S, Matheson P, et al. Declining mother-to-child 
    HIV transmission following perinatal zidovudine recommendations, 
    United States. Proceedings from the XI International Conference on 
    AIDS, Vancouver, Canada; July 7-12, 1996; Volume I: 248 (Abstract 
    Tu.C.440).
    Perelson AS, Neumann AU, Markowitz M, et al. HIV-1 dynamics in vivo: 
    virion clearance rate, infected cell life-span, and viral generation 
    time. Science 1996; 271:1582-6.
    Havlir DV, Richman DD. Viral dynamics of HIV: implications for drug 
    development and therapeutic strategies. Ann Intern Med 1996; 
    124:984-94.
    Mofenson LM. Mother-child HIV-1 transmission: timing and 
    determinants. Obstet Gynecol Clinics NA 1997: in press.
    Panel on Clinical Practices for Treatment of HIV Infection. 
    Guidelines for the use of antiretroviral agents in HIV-infected 
    adults and adolescents. U.S. Public Health Service, Washington, D.C. 
    1997, in press.
    Minkoff H, Augenbraun M. Antiretroviral therapy for pregnant women. 
    Am J Obstet Gynecol 1997; 176:478-89.
    Mills JL. Protecting the embryo from X-rated drugs. N Engl J Med 
    1995; 333:124-5.
    Centers for Disease Control. Pregnancy outcomes following systemic 
    prenatal acyclovir exposure--June 1, 1984-June 30, 1993. MMWR 1993; 
    42:806-9.
    Johnson MA, Goodwin C, Yuen GJ, et al. The pharmacokinetics of 3TC 
    administered to HIV-1 infected women (pre-partum, during labour and 
    post-partum) and their offspring. Proceedings from the XI 
    International Conference on AIDS, Vancouver, Canada; July 7-12, 
    1996; Volume I: 249-50 (Abstract Tu.C.445).
    Moodley J, Moodley D, Pillay K, et al. Antiviral effect of 
    lamivudine alone and in combination with zidovudine in HIV-infected 
    pregnant women. Proceedings from the Fourth Conference on 
    Retroviruses and Opportunistic Infections, Washington. D.C.; January 
    22-26, 1997: 176 (Abstract 607).
    Ayers KM, Clive D, Tucker WE Jr, Hajian G, de Miranda P. Nonclinical 
    toxicology studies with zidovudine: genetic toxicity tests and 
    carcinogenicity bioassays in mice and rats. Fundam Appl Toxicol 
    1996; 32:148-58.
    Olivero OA, Anderson LM, Diwan BA, et al. AZT is a genotoxic 
    transplacental carcinogen in animal models. JAIDS 1997:14:A29 
    (Abstract 52).
    Mirochnick M, Sullivan J, Gagnier P, et al. Safety and 
    pharmacokinetics of nevirapine in neonates born to HIV-1 infected 
    women. Proceedings from the Fourth Conference on Retroviruses and 
    Opportunistic Infections, Washington. D.C.; January 22-26, 1997: 176 
    (Abstract 723).
    Sperling RS, Shapiro DE, Coombs RW, et al. Maternal viral load, 
    zidovudine treatment, and the risk of transmission of human 
    immunodeficiency virus from mother to infant. N Engl J Med 1996; 
    335:1621-9.
    Sandberg JA, Slikker W Jr. Developmental pharmacology and toxicology 
    of anti-HIV therapeutic agents: dideoxynucleosides. FASEB J 
    1995;9:1157-63.
    Qian M, Bui T, Ho RJY, Unadkat JD. Metabolism of 3' -azido-3' -
    deoxythymidine (AZT) in human placental trophoblasts and hofbauer 
    cells. Biochem Pharmacol 1994; 48:383-9.
    Dancis J, Lee JD, Mendoza S, Liebes L. Transfer and metabolism of 
    dideoxyinosine by the perfused human placenta. JAIDS 1993; 6:2-6.
    Sandberg JA, Binienda Z, Lipe G, Slikker Jr W. Placental transfer 
    and fetal disposition of dideoxycytidine (ddC) and dideoxyinosine 
    (ddI). Toxicologist 1994; 14; 434 (abstract).
    
    [[Page 36821]]
    
    Antiretroviral Pregnancy Registry for didanosine, lamivudine, 
    saquinavir, stavudine, zalcitabine, zidovudine. Interim Report, 1 
    January 1989 through 31 December 1996. A Collaborative Project 
    Managed by Bristol Myers Squibb Co., Glaxo Wellcome, and Hoffman-La 
    Roche, Inc. Research Triangle Park, NC 1997.
    Connor E, Sperling R, Shapiro D, et al. Long term effect of 
    zidovudine exposure among uninfected infants born to HIV-infected 
    mothers in Pediatric AIDS Clinical Trials Group protocol 076. 
    Proceedings of the 35th Interscience Conference on Antimicrobial 
    Agents and Chemotherapy. San Francisco, CA, September 17-20 1995:205 
    (Abstract I1).
    Eastman PS, Shapiro DE, Coombs RW, et al. Maternal genotypic ZDV 
    resistance and failure of ZDV therapy to prevent mother-child HIV-1 
    transmission. Abstracts of the 4th Conference on Retroviruses and 
    Opportunistic Infections. Washington, D.C., January 22-26, 1997; 160 
    (Abstract 516).
    Hanson C, Cooper E, Antonelli T, et al. Lack of tumors in infants 
    with perinatal HIV exposure and fetal/neonatal exposure to 
    zidovudine. National Conference on Women and HIV. Pasadena CA, May 
    4-7, 1997.
    Bardegeuz A, Spino C. Interim analysis PACTG 288. Presented at 
    Workshop on Antiretroviral Therapy to Reduce the Risk of Perinatal 
    Transmission. Herndon, Virginia; May 9, 1997.
    Pediatric ACTG Protocol 185 Executive Summary. National Institute of 
    Child Health & Human Development, National Institutes of Health. 
    Bethesda, MD: March 25, 1997.
    Simpson BJ, Shapiro ED, Andiman WA. Reduction in the risk of 
    vertical transmission of HIV-1 associated with treatment of pregnant 
    women with orally administered zidovudine alone. JAIDS 1997; 14:145-
    52.
    Dickover RE, Garratty EM, Horman SA, et al. Identification of levels 
    of maternal HIV-1 RNA associated with risk of perinatal 
    transmission: effect of maternal zidovudine treatment on viral load. 
    JAMA 1996; 275:599-605.
    Mayaux M-J, Dussaix E, Isopet J, et al. Maternal virus load during 
    pregnancy and the mother-to-child transmission of human 
    immunodeficiency virus type 1: the French Perinatal Cohort Studies. 
    J Infect Dis 1997; 175:172-5.
    Cao Y, Krogstad P, Korber BT, et al. Maternal HIV-1 viral load and 
    vertical transmission of infection: The Ariel Project for the 
    prevention of HIV transmission from mother to infant. Nature 
    Medicine 1997; 3:549-52.
    Thea DM, Steketee RW, Bornshlegel K, et al. The effect of maternal 
    viral load on the risk of perinatal transmission of HIV-1. J Infect 
    Dis 1997; 175:707-11.
    Burchett SK, Kornegay J, Pitt J, et al. Assessment of maternal 
    plasma HIV viral load as a correlate of vertical transmission. 
    Proceedings of the Third National Conference on Retroviruses and 
    Opportunistic Infections. Washington, D.C., January 28-February 1, 
    1996:161 (Abstract LB3).
    Rasheed S, Li Z, Xu D, Kovacs A. Presence of cell-free human 
    immunodeficiency virus in cervicovaginal secretions is independent 
    of viral load in the blood of human immunodeficiency virus-infected 
    women. Am J Obstet Gynecol 1996; 175: 222-9.
    Melvin AJ, Burchett SK, Watts DH, et al. Effect of pregnancy and 
    zidovudine therapy on viral load in HIV-1-infected women. JAIDS 
    1997; 14:232-6.
    Centers for Disease Control. Recommendations for assisting in the 
    prevention of perinatal transmission of human T-lymphotropic virus 
    type III/lymphadenopathy-associated virus and acquired 
    immunodeficiency syndrome. MMWR 1985; 34:721-6.
    Rodman JH, Ribbins BL, Flynn PM, et al. A systemic and cellular 
    model for zidovudine plasma concentrations and intracellular 
    phosphorylation in patients. J Infect Dis 1996; 174:490-99.
    Barry MG, Khoo SH, Beal GJ, et al. The effect of zidovudine dose on 
    the information of intracellular phosphorylated metabolites. AIDS 
    1996; 10:1361-7.
    Gambertoglio JG, Peter K. Zidovudine phosphorylation after short 
    term and long term therapy with zidovudine in patients infected with 
    HIV. Clin Pharmacol Therapy 1996; 60:168-76.
    Mulder JW, Cooper DA, Mathiesen L, et al. Zidovudine twice daily in 
    asymptomatic subjects with HIV infection and a high risk of 
    progression to AIDS: a randomized, double-blind placebo controlled 
    study. AIDS 1994; 8:313-21.
    Mannucci PM, Gringeri A, Savidge G, et al. Randomized, double-blind, 
    placebo-controlled trial of twice-daily zidovudine in asymptomatic 
    haemophiliacs infected with the human immunodeficiency virus type 1. 
    Brit J Haematol 1994;86:174-9.
    Cooper DA, Gatell JM, Kroon S, et al. Zidovudine in persons with 
    asymptomatic HIV infection and CD4+ cell counts greater than 400 per 
    cubic millimeter. N Engl J Med 1993;329:297-303.
    Boucher FD, Modlin JF, Weller S, et al. Phase I evaluation of 
    zidovudine administered to infants exposed at birth to the human 
    immunodeficiency virus. J Pediatr 1993;122:1137-44.
    Capparelli EV, Mirochnick MH, Dankner WM. Zidovudine 
    pharmacokinetics in premature infants exposed to HIV. Pediatr Res 
    1996;39:169A.
    Shafer RW, Iversen AKN, Winters MA, et al. Drug resistance and 
    heterogeneous long-term virologic responses of human 
    immunodeficiency virus type 1-infected subjects to zidovudine and 
    didanosine combination therapy. J Infect Dis 1995;175:70-8.
    The Delta Coordinating Committee. Delta: a randomized double-blind 
    controlled trial comparing combinations of zidovudine plus 
    didanosine or zalcitabine with zidovudine alone in HIV-infected 
    individuals. Lancet 1996;348:283-91.
    Hammer SM, Katzenstein DA, Hughes MD, et al. A trial comparing 
    nucleoside monotherapy with combination therapy in HIV-infected 
    adults with CD4 cell counts from 200 to 500 per cubic millimeter. N 
    Engl J Med 1996;335:1081-90.
    Saravolatz LD, Winslow DL, Collins G et al. Zidovudine alone or in 
    combination with didanosine or zalcitabine in HIV-infected patients 
    with acquired immunodeficiency syndrome or fewer than 200 CD4 cells 
    per cubic millimeter. N Engl J Med 1996;335:1099-106.
    Kuritzkes DR. Clinical significance of drug resistance in HIV-1 
    infection. AIDS 1996;10 (suppl5):S27-31.
    Japour AJ, Welles S, D'Aquila RT, et al. Prevalence and clinical 
    significance of zidovudine resistance mutations in human 
    immunodeficiency virus isolated from patients after long-term 
    zidovudine treatment. J Infect Dis 1995;171;1172-9.
    Dunn DT, Brandt CD, Krivine A, et al. The sensitivity of HIV-1 DNA 
    polymerase chain reaction in the neonatal period and the relative 
    contributions of intra-uterine and intra-partum transmission. AIDS 
    1995;9:F7-F11.
    Centers for Disease Control. Update: provisional public health 
    service recommendations for chemoprophylaxis after occupational 
    exposure to HIV. MMWR 1996;45:468-72.
    Centers for Disease Control. Case-control study of HIV 
    seroconversion in health-care workers after percutaneous exposure to 
    HIV-infected blood--France, United Kingdom, and United States, 
    January 1988-August 1994. MMWR 1995;44:929-33.
    Van Rompay KKA, Otsyula MG, Marthas ML, et al. Immediate zidovudine 
    treatment protects simian immunodeficiency virus-infected newborn 
    macaques against rapid onset of AIDS. Antimicrob Ag Chemother 
    1995;39:125-31.
    Bottiger D, Johansson N-G, Samuelsson B, et al. Prevention of simian 
    immunodeficiency virus, SIVsm, or HIV-2 infection in cynomolgus 
    monkeys by pre- and postexposure administration of BEA-005. AIDS 
    1997;11:157-62.
    Tsai C-C, Follis KE, Sabo A, et al. Prevention of SIV infection in 
    macaques by (R)-9-(2-phosphonylmethoxypropyl)adenine. Science 
    1995;270:1197-9.
    Mathes LE, Polas PJ, Hayes KA, et al. Pre- and post-exposure 
    chemoprophylaxis: evidence that 3'-azido-3'dideoxythymidine (AZT) 
    inhibits feline leukemia virus diseases by a drug-induced vaccine 
    effect. Antimicrob Ag Chemother 1992;36:2715-21.
    Miotti PB, Liomba G, Dallabetta GA, et al. T-lymphocyte subsets 
    during and after pregnancy: analysis in human immunodeficiency virus 
    type 1-infected and uninfected Malawian mothers. J Infect Dis 
    1992;165:146-9.
    
    [[Page 36822]]
    
    Tuomala RE, Kalish LA, Zorilla C, et al. A longitudinal study of 
    changes in total, CD4+ and CD8+ lymphocytes during pregnancy and one 
    postpartum year in HIV-infected women. Obstet Gynecol 1997;in press.
    Centers for Disease Control. 1995 revised guidelines for prophylaxis 
    against Pneumocystis carinii pneumonia for children infected with or 
    perinatally exposed to human immunodeficiency virus. MMWR 1995;44:1-
    11.
    Kovacs A, Xu J, Rasheed S, et al. Comparison of a rapid nonisotopic 
    polymerase chain reaction assay with four commonly used methods for 
    the early diagnosis of human immunodeficiency virus type 1 infection 
    in neonates and children. Pediatr Infect Dis J 1995;14:948-54.
    
        Dated: June 26, 1997.
    John M. Eisenberg,
    Principal Deputy Assistant Secretary for Health, U.S. Department of 
    Health and Human Services.
    
                                             Table 1.--PACTG 076 ZDV Regimen                                        
    ----------------------------------------------------------------------------------------------------------------
                                                                                                                    
    ----------------------------------------------------------------------------------------------------------------
    Antepartum..........................................  Oral administration of 100 mg ZDV five times daily,       
                                                           initiated at 14-34 weeks gestation and continued         
                                                           throughout the pregnancy.                                
    Intrapartum.........................................  During labor, intravenous administration of ZDV in a 1-   
                                                           hour loading dose of 2 mg per kg of body weight, followed
                                                           by a continuous infusion of 1 mg per kg of body weight   
                                                           per hour until delivery.                                 
    Postpartum..........................................  Oral administration of ZDV to the newborn (ZDV syrup at 2 
                                                           mg per kg body weight per dose every 6 hours) for the    
                                                           first 6 weeks of life, beginning at 8-12 hours after     
                                                           birth (Note: intravenous dosage for infants who cannot   
                                                           tolerate oral intake is 1.5 mg per kg body weight        
                                                           intravenously every 6 hours).                            
    ----------------------------------------------------------------------------------------------------------------
    
    
                                     Table 2.--Preclinical and Clinical Data Relevant to Use of Antiretrovirals in Pregnancy                                
    --------------------------------------------------------------------------------------------------------------------------------------------------------
                                                                                                                                      Long-term animal      
              Antiretroviral drug               FDA pregnancy category*      Placental passage [newborn: maternal drug ratio]     carcinogenicity  studies  
    --------------------------------------------------------------------------------------------------------------------------------------------------------
    Nucleoside Analogue Reverse                                                                                                                             
     Transcriptase Inhibitors:                                                                                                                              
        Zidovudine (ZDV)..................  C                               Yes (human) [0.85]...............................  Positive (rodent, noninvasive
                                                                                                                                vaginal epithelial tumors). 
        Zalcitabine (ddC).................  C                               Yes (rhesus) [0.30-0.50].........................  Positive (rodent, thymic     
                                                                                                                                lymphomas).                 
        Didanosine (ddI)..................  B                               Yes (human) [0.5]................................  Negative (no tumors, lifetime
                                                                                                                                rodent study).              
        Stavudine (d4T)...................  C                               Yes (rhesus) [0.76]..............................  Not completed.               
        Lamivudine (3TC)..................  C                               Yes (human) [1.0]................................  Negative (no tumors, lifetime
                                                                                                                                rodent study).              
    Non-Nucleoside Reverse Transcriptase                                                                                                                    
     Inhibitors:                                                                                                                                            
        Nevirapine........................  C                               Yes (human) [1.0]................................  Not completed.               
        Delavirdine.......................  C                               Unknown..........................................  Not completed.               
    Protease Inhibitors:                                                                                                                                    
        Indinavir.........................  C                               Yes (rats) ``Significant'' in rats, but low in     Not completed.               
                                                                             rabbits.                                                                       
        Ritonavir.........................  B                               Yes (rats) [mid-term fetus, 1.15; late-term        Not completed.               
                                                                             fetus, 0.15-0.64].                                                             
        Saquinavir........................  B                               Unknown..........................................  Not completed.               
        Nelfinavir........................  B                               Unknown..........................................  Not completed.               
    --------------------------------------------------------------------------------------------------------------------------------------------------------
    * FDA Pregnancy Categories are:                                                                                                                         
      A--Adequate and well-controlled studies of pregnant women fail to demonstrate a risk to the fetus during the first trimester of pregnancy (and there  
      is no evidence of risk during later trimesters);                                                                                                      
      B--Animal reproduction studies fail to demonstrate a risk to the fetus and adequate but well-controlled studies of pregnant women have not been       
      conducted;                                                                                                                                            
      C--Safety in human pregnancy has not been determined, animal studies are either positive for fetal risk or have not been conducted, and the drug      
      should not be used unless the potential benefit outweighs the potential risk to the fetus;                                                            
      D--Positive evidence of human fetal risk based on adverse reaction data from investigational or marketing experiences, but the potential benefits from
      the use of the drug in pregnant women may be acceptable despite its potential risks;                                                                  
      X--Studies in animals or reports of adverse reactions have indicated that the risk associated with the use of the drug for pregnant women clearly     
      outweighs any possible benefit.                                                                                                                       
    
    
      Table 3.--Summary: Clinical Situations and Recommendations for Use of 
            Antiretroviral Drugs To Reduce Perinatal HIV Transmission       
    ------------------------------------------------------------------------
          Clinical scenario                     Recommendation*             
    ------------------------------------------------------------------------
    Scenario #1: HIV-infected      HIV-1 infected pregnant women must       
     pregnant women without prior   receive standard clinical, immunologic  
     antiretroviral therapy.        and virologic evaluation, and           
                                    recommendations for initiation and      
                                    choice of antiretroviral therapy should 
                                    be based on the same parameters used in 
                                    non-pregnant individuals, with          
                                    consideration and discussion of the     
                                    known and unknown risks and benefits of 
                                    such therapy during pregnancy.          
                                   The 3-part ZDV chemoprophylaxis regimen  
                                    should be recommended for all HIV-      
                                    infected pregnant women to reduce the   
                                    risk of perinatal transmission.         
                                   If the woman's clinical, immunologic and 
                                    virologic status indicates that more    
                                    aggressive therapy is recommended to    
                                    treat her infection (Panelrec, 1997),   
                                    other antiretroviral drugs should be    
                                    recommended in addition to ZDV.         
    
    [[Page 36823]]
    
                                                                            
                                   If the woman's status is such that       
                                    therapy would be considered optional,   
                                    the use of additional antiretrovirals   
                                    may be offered, although whether this   
                                    will provide additional benefit to the  
                                    woman or her child is not known.        
                                   Women who are in the first trimester of  
                                    pregnancy may wish to consider delaying 
                                    initiation of therapy at least until    
                                    after 10 to 12 weeks gestation.         
    Scenario #2: HIV-infected      HIV-1 infected women receiving           
     women receiving                antiretroviral therapy in whom pregnancy
     antiretroviral therapy         is identified after the first trimester 
     during the current pregnancy.  should continue therapy.                
                                   For women receiving antiretroviral       
                                    therapy in whom pregnancy is recognized 
                                    during the first trimester, the woman   
                                    should be counseled regarding the       
                                    benefits and potential risks of         
                                    antiretroviral administration during    
                                    this period, and continuation of therapy
                                    should be considered.                   
                                   If therapy is discontinued during the    
                                    first trimester, all drugs should be    
                                    stopped and reintroduced simultaneously 
                                    to avoid the development of resistance. 
                                   If the current therapeutic regimen does  
                                    not contain ZDV, the addition of ZDV or 
                                    substitution of ZDV for another         
                                    nucleoside analogue antiretroviral is   
                                    recommended after 14 weeks gestation.   
                                    Intrapartum and newborn ZDV             
                                    administration is recommended regardless
                                    of the antepartum antiretroviral        
                                    regimen.                                
    Scenario #3: HIV-infected      Administration of intrapartum intravenous
     women in labor who have had    ZDV should be recommended along with the
     no prior therapy.              6-week newborn ZDV regimen.             
                                   In the immediate postpartum period, the  
                                    woman should have appropriate           
                                    assessments (e.g., CD4 count, HIV-1 RNA 
                                    copy number) to determine if            
                                    antiretroviral therapy is recommended   
                                    for her own health.                     
    Scenario #4: Infants born to   The 6 week neonatal ZDV component of the 
     mothers who have received no   ZDV chemoprophylactic regimen should be 
     antiretroviral therapy         discussed with the mother and offered   
     during pregnancy or            for the newborn.                        
     intrapartum.                                                           
                                   ZDV should be initiated as soon as       
                                    possible after birth, preferably within 
                                    12-24 hours after birth.                
                                   Some clinicians may choose to use ZDV in 
                                    combination with other antiretroviral   
                                    drugs, particularly if the mother has   
                                    known or suspected ZDV-resistant virus. 
                                    However, the efficacy of this approach  
                                    is unknown and appropriate dosing       
                                    regimen for neonates are incompletely   
                                    defined.                                
                                   In the immediate postpartum period, the  
                                    woman should undergo appropriate        
                                    assessments (e.g., CD4 count, HIV-1 RNA 
                                    copy number) to determine if            
                                    antiretroviral therapy is required for  
                                    her own health.                         
    ------------------------------------------------------------------------
    * General note: Discussion of treatment options and recommendations     
      should be noncoercive, and the final decision regarding the use of    
      antiretroviral drugs is the responsibility of the woman. A decision to
      not accept treatment with ZDV or other drugs should not result in     
      punitive action or denial of care, nor should use of ZDV be denied to 
      a woman who wishes to minimize exposure of the fetus to other         
      antiretroviral drugs and therefore chooses to receive only ZDV during 
      pregnancy to reduce the risk of perinatal transmission.               
    
    [FR Doc. 97-17854 Filed 7-8-97; 8:45 am]
    BILLING CODE 4140-01-P
    
    
    

Document Information

Published:
07/09/1997
Department:
Health and Human Services Department
Entry Type:
Notice
Action:
Notice.
Document Number:
97-17854
Dates:
Comments on the proposed guidelines must be received on or before August 8, 1997 in order to ensure that NIH will be able to consider the comments in preparing the final guidelines.
Pages:
36809-36823 (15 pages)
PDF File:
97-17854.pdf