Evaluating the Safety and Pharmacokinetics of Raltegravir in Infants

Sponsor
National Institute of Allergy and Infectious Diseases (NIAID) (NIH)
Overall Status
Completed
CT.gov ID
NCT01828073
Collaborator
(none)
40
19
83.2
2.1
0

Study Details

Study Description

Brief Summary

The purpose of this study was to determine the washout pharmacokinetics (PK) and safety of in utero/intrapartum exposure to maternal raltegravir (RAL) in infants born to pregnant women with HIV infection who received RAL 400 mg twice daily. The study also provided data for the development of an infant RAL starting dosing regimen for IMPAACT P1110 (NCT01780831).

Condition or Disease Intervention/Treatment Phase

Detailed Description

Study participants were enrolled in two cohorts.

  • Cohort 1 enrolled mother-infant pairs in which the infant was expected to be ≥2000 grams at birth (i.e. full term) at time of enrollment and the mother was living with HIV and received RAL 400 mg twice daily for at least 2 weeks prior to delivery and continued to receive antiretroviral (ARV) drugs during labor.

  • Cohort 2 enrolled mother-infant pairs in which the infant was expected to be ≤2500 grams at birth [i.e. low birth weight (LBW)] at time of enrollment and the mother was living with HIV and received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery.

Cohorts 1 and 2 provided pharmacokinetics and safety data of in utero and intrapartum exposure to maternal RAL in full-term and LBW infants, respectively. Also, the study data were pooled with data from IMPAACT P1066 (NCT00485264) (Cohorts IV and V) and P1026s (NCT00042289) to determine the starting RAL dosing regimen for full-term and LBW infants in IMPAACT P1110 (NCT01780831).

The study initially opened accrual to Cohort 1 under protocol Version 1.0. Upon completion of accrual and follow-up of Cohort 1, the protocol was amended and accrual to and follow-up of Cohort 2 was under protocol Version 2.0.

No study-specific treatment was given to the participants during this study. The women (mothers) received RAL for clinical indications outside of the study. Infants received standard of care ARV therapy for prophylaxis of perinatal transmission of HIV as prescribed by their primary care physicians.

Cohort 1 mother-infant pairs were enrolled prior to delivery. The women were followed-up until discharge from the labor/delivery unit. Infants were followed from birth through 20 weeks after birth. If infant was eligible for PK sampling (see "Eligibility" section), blood samples were collected at 1-5, 8-14, 18-24, and 30-36 hours after birth. Protocol defined infant safety evaluations were at birth, and at 8-14 hours, 30-36 hours, 1 week and 20 weeks after birth.

Cohort 2 mother-infant pairs were enrolled prior to delivery or within 48 hours after delivery. The women were followed-up until discharge from the labor/delivery unit. Infants were followed from birth/entry through 6 weeks after birth. If infant was eligible for PK sampling, blood samples were collected at 1-6, 12-24, 36-48, 72-84, and 108-132 hours and 7-14 days after birth. Protocol defined infant safety evaluations were at entry/birth, and at 36-48 hours, 72-84 hours, 1 week and 6 weeks after birth.

For both cohorts, all infants regardless of whether they were eligible for PK sampling were included in the safety analyses. Infant safety data included adverse birth outcomes, signs/symptoms, diagnoses and chemistry/hematology test results. Protocol required chemistry tests were AST, ALT, serum creatinine, total bilirubin and direct bilirubin. Protocol required hematology tests were CBC with differential and platelet count. Also included in the safety data were additional laboratory events done outside of the study but considered by the site as relevant information.

For both cohorts, maternal blood and cord blood for RAL concentration testing were collected at delivery when specimen collection was possible. The optional genotypic testing (i.e. testing was done only if the mother consented) was limited to infants who were eligible for PK sampling. Information obtained about the effect of UGT1A1 polymorphisms on the PK of RAL was thought to provide a better understanding of the effect of genetics on the metabolism of RAL in neonates.

Study Design

Study Type:
Observational
Actual Enrollment :
40 participants
Observational Model:
Cohort
Time Perspective:
Prospective
Official Title:
Raltegravir Pharmacokinetics and Safety in Neonates
Study Start Date :
May 19, 2011
Actual Primary Completion Date :
Apr 23, 2018
Actual Study Completion Date :
Apr 23, 2018

Arms and Interventions

Arm Intervention/Treatment
Cohort 1: Full term infants exposed in utero to maternal RAL

Infants, who were expected to be ≥2000 grams at birth (i.e. full-term) at time of enrollment, born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. The group also includes the mothers of these infants.

Drug: Raltegravir
No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
Other Names:
  • RAL
  • Cohort 2: LBW infants exposed in utero to maternal RAL

    Infants, who were expected to be ≤2500 grams at birth (i.e. LBW) at time of enrollment, born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. The group also includes the mothers of these infants.

    Drug: Raltegravir
    No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
    Other Names:
  • RAL
  • Outcome Measures

    Primary Outcome Measures

    1. PK Parameter: Neonatal RAL Elimination Half-life (T1/2) [Infant blood specimens were collected at 1-5, 8-14, 18-24, and 30-36 hours after birth for Cohort 1; and at 1-6, 12-24, 36-48, 72-84, and 108-132 hours after birth, and on day 7-14 for Cohort 2.]

      Time required for neonatal plasma concentration to decrease by one-half. T1/2 was estimated using the terminal 3 concentration-time points for each infant when available.

    2. Ratio of Cord Blood to Maternal Blood RAL Concentrations [Maternal blood samples were scheduled to be collected within 1 hour after delivery and cord blood sample were collected immediately after cord was clamped]

      Ratio of the neonatal cord blood RAL concentration to the mother's plasma RAL concentration at birth

    3. Number of Infants Who Met Composite Safety Endpoint (Grade 3/4 Adverse Event, Adverse Birth Outcome, Death) [Assessed at entry through Week 20 for Cohort 1 infants and through Week 6 for Cohort 2 infants.]

      An infant was said to have met the composite safety endpoint if any of the following was observed: adverse events (AEs) of Grade 3 or 4 as defined in DAIDS AE Grading Table adverse birth outcomes including stillbirth and low birth weight (LBW), or death. Stillbirth could only be observed on infants enrolled prior to delivery. Cohort 2 enrolled LBW infants and prematurity and growth restriction which were highly linked to LBW were considered as baseline events and not AEs or adverse birth outcome for Cohort 2 infants.

    4. Infant Total Bilirubin [Measured at 8-14 hours (Visit 1), 30-36 hours (Visit 2) and 1-2 weeks (Visit 3) after birth for Cohort 1; and at 36-48 hours (Visit 1), 72-84 hours (Visit 2) and 1 week (Visit 3)after birth for Cohort 2.]

      Total bilirubin measured from infant blood specimens.

    5. Infant Direct Bilirubin [Measured at 8-14 hours (Visit 1), 30-36 hours (Visit 2) and 1-2 weeks (Visit 3) after birth for Cohort 1; and at 36-48 hours (Visit 1), 72-84 hours (Visit 2) and 1 week (Visit 3)after birth for Cohort 2.]

      Direct bilirubin measured from infant blood specimens.

    6. Number of Infants Who Received Treatment to Reduce Bilirubin or for Jaundice [Assessed from entry through around week 1 after birth]

      Assessment if infant received exchange transfusion, Phototherapy, or other treatment to reduce bilirubin or for jaundice

    Secondary Outcome Measures

    1. Neonatal RAL Elimination (T1/2) by UGT1A1 Genotype Group (Normal VS Mutation) [Genotype was assessed close to birth and if this is not possible at 1-2 wks after birth. PK samples were collected at 1-5, 8-14, 18-24 and 30-36 hrs after birth for Cohort 1; 1-6, 12-24, 36-48, 72-84 and 108-132 hrs after birth, and day 7-14 for Cohort 2.]

      Neonatal RAL elimination was the time required for neonatal plasma concentration to decrease by one-half. Genotyping for polymorphisms of UGT1A1 were performed on infants who were eligible for PK sampling and were consented by their mothers/guardians(i.e. genotyping was optional) . The goal of the genotypic analysis is to determine if certain polymorphisms, particularly those with the UGT1A1*28/*28 genotype have slower RAL elimination than those with the UGT1A1*1/*1 genotype.

    Eligibility Criteria

    Criteria

    Ages Eligible for Study:
    N/A and Older
    Sexes Eligible for Study:
    All
    Accepts Healthy Volunteers:
    No

    Participant study inclusions and exclusion criteria are listed below.

    Cohort 1 M-I pairs were enrolled prior to delivery so that only maternal study inclusion and exclusion criteria were assessed at enrollment.

    Cohort 1: Maternal Study Inclusion Criteria

    • Documentation of HIV-1 infection.

    • Viable singleton pregnancy with gestational age of at least 35 weeks based on clinical or other obstetrical measurements with normal fetal anatomy

    • Currently receiving RAL 400 mg twice daily for at least 2 weeks prior to enrollment in combination with other ARV agents for clinical care

    • Plan to continue taking RAL in combination with other ARV agents through labor prior to delivery

    • Willing and intends to deliver at the study-affiliated clinic or hospital

    • Willing and able to sign informed consent for participation of herself and her infant. Participant must be of an age to provide legal informed consent as defined by the country in which she resides. If not, informed consent must be signed by a legal guardian.

    Cohort 1: Maternal Study Exclusion Criteria

    • Receipt of disallowed medications (phenobarbital, phenytoin, rifampin) within 4 weeks prior to enrollment

    Cohort 1 Infants were enrolled prior to delivery so there were no infant study inclusion/exclusion criteria. However, only infants who met the following criteria were eligible for PK blood sampling. Infants ineligible for PK sampling remained in the study and were followed-up for safety.

    Cohort 1: Infant PK Sampling Inclusion Criteria

    • Infant born to women who received at least 2 weeks of RAL prior to delivery and continue to receive RAL during labor prior to delivery in addition to their other ARV drugs

    • Infant birth weight of at least 2 kg

    • Infant at least 37 weeks gestation at delivery

    • Infant not receiving disallowed medications (phenobarbital, phenytoin, rifampin). If these medications are required for the infant's care, the infant will be ineligible for further PK sampling. PK samples will be obtained up to the time of the introduction of the disallowed medication.

    Cohort 1: Infant PK Sampling Exclusion Criteria

    • Infant has a severe congenital malformation or other medical condition not compatible with life or that would interfere with study participation or interpretation, as judged by the examining clinician

    Cohort 2 enrolled M-I pairs at two time points: prior to delivery and within 48 hours after delivery.

    • For M-I pairs enrolled prior to delivery, the maternal study eligibility criteria were assessed at enrollment. There were no infant study eligibility criteria. However, only infants who met the PK sampling eligibility criteria had PK blood sampling. Infants ineligible for PK sampling remained in the study and were followed-up for safety.

    • For M-I pairs enrolled within 48 hours delivery, the maternal and infant study eligibility criteria were assessed at enrollment. A M-I pair was enrolled only if both the mother and the infant were eligible for the study. For multiple births, only infants who met the study eligibility criteria were enrolled.

    Cohort 2: Maternal Study Inclusion Criteria: M-I pairs enrolled prior to delivery

    • Documentation of HIV-1 infection.

    • Viable singleton or multiple birth pregnancy based on clinical or other obstetrical measurements with infant birth weight anticipated to be less than or equal to 2,500 grams

    • RAL is currently used as part of maternal ARV regimen and planned to continue through labor and delivery

    • Willing and intends to deliver at the study-affiliated clinic or hospital

    • Willing and able to sign informed consent for participation of herself and her infant. Participant must be of an age to provide legal informed consent as defined by the country in which she resides. If not, informed consent must be signed by a legal guardian.

    Cohort 2: Maternal Study Exclusion Criteria: M-I pairs enrolled prior to delivery

    • Receipt of disallowed medications (phenobarbital, phenytoin, rifampin) within 4 weeks prior to enrollment or intent to be on any of the disallowed medications prior to delivery.

    Cohort 2: Infant PK Blood Sampling Eligibility Criteria: M-I pairs enrolled prior to delivery

    Infants were enrolled prior to delivery so there were no infant study eligibility criteria.

    Only infants who met the following criteria were eligible for PK blood sampling:
    • Infant born to woman who received at least one dose of RAL within 2 to 24 hours prior to delivery. Dose administered to mother must have been at least 2 hours prior to delivery to allow time for adequate absorption and distribution.

    • Infant birth weight less than or equal to 2,500 grams

    • Infant not receiving disallowed medications (phenobarbital, phenytoin, rifampin) as described in the protocol. If these medications are required for the infant's care, the infant will be ineligible for further PK sampling. PK data will be obtained up to the time of the introduction of the disallowed medication.

    • Infant less than or equal to 48 hours of age

    • Infant does not have any severe congenital malformation or other medical condition not compatible with life or that would interfere with study participation or interpretation, as judged by the examining clinician

    Cohort 2: Maternal Study Inclusion Criteria: M-I pairs enrolled after delivery

    • Documentation of HIV-1 infection.

    • Received at least one dose of RAL within 2 to 24 hours prior to delivery

    • Willing and able to sign informed consent for participation of herself and her infant. Participant must be of an age to provide legal informed consent as defined by the country in which she resides. If not, informed consent must be signed by a legal guardian.

    Cohort 2: Maternal Study Exclusion Criteria: M-I pairs enrolled after delivery

    • Receipt of disallowed medications (phenobarbital, phenytoin, rifampin) within 4 weeks prior to delivery

    Cohort 2: Infant Study Inclusion Criteria: M-I pairs enrolled after delivery

    • Infant birth weight less than or equal to 2,500 grams

    • Infant less than or equal to 48 hours of age

    Cohort 2: Infant Study Exclusion Criteria: M-I pairs enrolled after delivery

    • Received disallowed medications (phenobarbital, phenytoin, rifampin)

    • Infant has a severe congenital malformation or other medical condition not compatible with life or that would interfere with study participation or interpretation, as judged by the examining clinician

    Contacts and Locations

    Locations

    Site City State Country Postal Code
    1 University of California, UC San Diego CRS- Mother-Child-Adolescent HIV Program La Jolla California United States 92093-0672
    2 Miller Children's Hosp. Long Beach CA NICHD CRS Long Beach California United States 90806
    3 Usc La Nichd Crs Los Angeles California United States 90089
    4 David Geffen School of Medicine at UCLA NICHD CRS Los Angeles California United States 90095-1752
    5 Univ. of California San Francisco NICHD CRS San Francisco California United States 94143
    6 Univ. of Florida Jacksonville NICHD CRS Jacksonville Florida United States 32209
    7 Johns Hopkins Univ. Baltimore NICHD CRS Baltimore Maryland United States 21287
    8 Boston Medical Center Ped. HIV Program NICHD CRS Boston Massachusetts United States 02118
    9 Bronx-Lebanon Hospital Center NICHD CRS Bronx New York United States 10457
    10 Jacobi Med. Ctr. Bronx NICHD CRS Bronx New York United States 10461
    11 St. Jude Children's Research Hospital CRS Memphis Tennessee United States 38105-3678
    12 Seattle Children's Research Institute CRS Seattle Washington United States 98101
    13 Hospital Nossa Senhora da Conceicao NICHD CRS Porto Alegre Rio Grande Do Sul Brazil 91350-200
    14 Hospital Federal dos Servidores do Estado NICHD CRS Rio de Janeiro Brazil 20221-903
    15 Hosp. Geral De Nova Igaucu Brazil NICHD CRS Rio de Janeiro Brazil 26030
    16 Univ. of Sao Paulo Brazil NICHD CRS Sao Paulo Brazil 14049-900
    17 Soweto IMPAACT CRS Johannesburg Gauteng South Africa 1862
    18 Kilimanjaro Christian Medical Centre (KCMC) Moshi Tanzania
    19 Chiangrai Prachanukroh Hospital NICHD CRS Chiang Mai Thailand 50100

    Sponsors and Collaborators

    • National Institute of Allergy and Infectious Diseases (NIAID)

    Investigators

    • Study Chair: Diana F. Clarke, PharmD, Boston Medical Center

    Study Documents (Full-Text)

    More Information

    Additional Information:

    Publications

    Responsible Party:
    National Institute of Allergy and Infectious Diseases (NIAID)
    ClinicalTrials.gov Identifier:
    NCT01828073
    Other Study ID Numbers:
    • P1097
    • 11790
    • IMPAACT P1097
    First Posted:
    Apr 10, 2013
    Last Update Posted:
    Nov 8, 2021
    Last Verified:
    Feb 1, 2020
    Additional relevant MeSH terms:

    Study Results

    Participant Flow

    Recruitment Details Cohort 1 participants were enrolled from 11 sites in the USA. Enrollment period was from May 2011 through September 2012. Cohort 2 participants were enrolled from 4 sites in Brazil, 1 site in South Africa, 1 site in Tanzania, 1 site in Thailand, and 3 sites in the USA. Enrollment period was from January 2015 through March 2018.
    Pre-assignment Detail
    Arm/Group Title Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Arm/Group Description Infants, who were expected to be ≥2000 grams at birth (i.e. full-term), born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. Raltegravir: No study-specific drugs was given to women or infants during this study. Women received RAL for clinical indications outside of the study. Infants, who were expected to be ≤2500 grams at birth (i.e. LBW), born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
    Period Title: Overall Study
    STARTED 22 18
    COMPLETED 21 16
    NOT COMPLETED 1 2

    Baseline Characteristics

    Arm/Group Title Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL Total
    Arm/Group Description Infants, who were expected to be ≥2000 grams at birth (i.e. full-term), born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study. Infants, who were expected to be ≤2500 grams at birth (i.e. LBW), born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study. Total of all reporting groups
    Overall Participants 22 18 40
    Age (Count of Participants)
    <=18 years
    22
    100%
    18
    100%
    40
    100%
    Between 18 and 65 years
    0
    0%
    0
    0%
    0
    0%
    >=65 years
    0
    0%
    0
    0%
    0
    0%
    Age, Customized (Count of Participants)
    Enrolled prior to birth
    22
    100%
    0
    0%
    22
    55%
    Enrolled after birth
    0
    0%
    18
    100%
    18
    45%
    Sex: Female, Male (Count of Participants)
    Female
    6
    27.3%
    12
    66.7%
    18
    45%
    Male
    16
    72.7%
    6
    33.3%
    22
    55%
    Ethnicity (NIH/OMB) (Count of Participants)
    Hispanic or Latino
    8
    36.4%
    9
    50%
    17
    42.5%
    Not Hispanic or Latino
    13
    59.1%
    9
    50%
    22
    55%
    Unknown or Not Reported
    1
    4.5%
    0
    0%
    1
    2.5%
    Birth Weight (g) (Count of Participants)
    1500 - <2000 g
    0
    0%
    7
    38.9%
    7
    17.5%
    2000 - 2500 g
    1
    4.5%
    11
    61.1%
    12
    30%
    >2500 g
    21
    95.5%
    0
    0%
    21
    52.5%

    Outcome Measures

    1. Primary Outcome
    Title PK Parameter: Neonatal RAL Elimination Half-life (T1/2)
    Description Time required for neonatal plasma concentration to decrease by one-half. T1/2 was estimated using the terminal 3 concentration-time points for each infant when available.
    Time Frame Infant blood specimens were collected at 1-5, 8-14, 18-24, and 30-36 hours after birth for Cohort 1; and at 1-6, 12-24, 36-48, 72-84, and 108-132 hours after birth, and on day 7-14 for Cohort 2.

    Outcome Measure Data

    Analysis Population Description
    Infants with RAL concentration (conc) for whom T1/2 could be calculated. Excluded (i) 5 Cohort 1 infants: 2 had no data, 3 had data but could not calculate T1/2 (terminal RAL conc below level of quantification (BLQ), higher RAL conc at later collection time); and (ii) 1 Cohort 2 infants:1 had terminal RAL conc BLQ.
    Arm/Group Title Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Arm/Group Description Infants, who were expected to be ≥2000 grams at birth (i.e. full-term), born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study. Infants, who were expected to be ≤2500 grams at birth (i.e. LBW), born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
    Measure Participants 17 17
    Median (Full Range) [Hours]
    26.6
    24.4
    2. Primary Outcome
    Title Ratio of Cord Blood to Maternal Blood RAL Concentrations
    Description Ratio of the neonatal cord blood RAL concentration to the mother's plasma RAL concentration at birth
    Time Frame Maternal blood samples were scheduled to be collected within 1 hour after delivery and cord blood sample were collected immediately after cord was clamped

    Outcome Measure Data

    Analysis Population Description
    Mother-Infant (M-I) pairs with maternal blood and cord blood samples. Excluded were (i) 3 Cohort 1 M-I pairs with neither cord blood nor maternal blood samples; and (ii) 16 Cohort 2 M-I pairs: 5 had neither cord blood nor maternal blood specimens, 11 had no cord blood specimen.
    Arm/Group Title Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Arm/Group Description Infants, who were expected to be ≥2000 grams at birth (i.e. full-term), born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. The group also includes the mothers of these infants. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study. Infants, who were expected to be ≤2500 grams at birth (i.e. LBW), born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. The group also includes the mothers of these infants. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
    Measure Participants 19 2
    Median (Full Range) [ratio]
    1.48
    2.62
    3. Primary Outcome
    Title Number of Infants Who Met Composite Safety Endpoint (Grade 3/4 Adverse Event, Adverse Birth Outcome, Death)
    Description An infant was said to have met the composite safety endpoint if any of the following was observed: adverse events (AEs) of Grade 3 or 4 as defined in DAIDS AE Grading Table adverse birth outcomes including stillbirth and low birth weight (LBW), or death. Stillbirth could only be observed on infants enrolled prior to delivery. Cohort 2 enrolled LBW infants and prematurity and growth restriction which were highly linked to LBW were considered as baseline events and not AEs or adverse birth outcome for Cohort 2 infants.
    Time Frame Assessed at entry through Week 20 for Cohort 1 infants and through Week 6 for Cohort 2 infants.

    Outcome Measure Data

    Analysis Population Description
    All infants.
    Arm/Group Title Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Arm/Group Description Infants, who were expected to be ≥2000 grams at birth (i.e. full-term), born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study. Infants, who were expected to be ≤2500 grams at birth (i.e. LBW), born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
    Measure Participants 22 18
    Count of Participants [Participants]
    7
    31.8%
    9
    50%
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL
    Comments Point and 90% CI estimates of percentage of full term infants meeting the composite safety endpoint (grade 3/4 adverse event, adverse birth outcome, death)
    Type of Statistical Test Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Clopper-Pearson Confidence Interval (CI)
    Estimated Value 31.82
    Confidence Interval (2-Sided) 90%
    16.00 to 51.50
    Parameter Dispersion Type:
    Value:
    Estimation Comments With the small sample size, the CI estimate tend to be wide.
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Comments Point and 90% CI estimates of percentage of full term infants meeting the composite safety endpoint (grade 3/4 adverse event, death)
    Type of Statistical Test Other
    Comments
    Statistical Test of Hypothesis p-Value
    Comments
    Method
    Comments
    Method of Estimation Estimation Parameter Clopper-Pearson Confidence Interval (CI)
    Estimated Value 50.00
    Confidence Interval (2-Sided) 90%
    29.10 to 70.90
    Parameter Dispersion Type:
    Value:
    Estimation Comments With the small sample size, the CI estimate tend to be wide.
    4. Primary Outcome
    Title Infant Total Bilirubin
    Description Total bilirubin measured from infant blood specimens.
    Time Frame Measured at 8-14 hours (Visit 1), 30-36 hours (Visit 2) and 1-2 weeks (Visit 3) after birth for Cohort 1; and at 36-48 hours (Visit 1), 72-84 hours (Visit 2) and 1 week (Visit 3)after birth for Cohort 2.

    Outcome Measure Data

    Analysis Population Description
    Infants with total bilirubin results
    Arm/Group Title Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Arm/Group Description Infants, who were expected to be ≥2000 grams at birth (i.e. full-term), born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study. Infants, who were expected to be ≤2500 grams at birth (i.e. LBW), born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
    Measure Participants 22 18
    Visit 1
    3.7
    6.6
    Visit 2
    5.7
    10.7
    Visit 3
    2.7
    4.6
    5. Primary Outcome
    Title Infant Direct Bilirubin
    Description Direct bilirubin measured from infant blood specimens.
    Time Frame Measured at 8-14 hours (Visit 1), 30-36 hours (Visit 2) and 1-2 weeks (Visit 3) after birth for Cohort 1; and at 36-48 hours (Visit 1), 72-84 hours (Visit 2) and 1 week (Visit 3)after birth for Cohort 2.

    Outcome Measure Data

    Analysis Population Description
    Infants with Direct Bilirubin results
    Arm/Group Title Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Arm/Group Description Infants, who were expected to be ≥2000 grams at birth (i.e. full-term), born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study. Infants, who were expected to be ≤2500 grams at birth (i.e. LBW), born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
    Measure Participants 22 18
    Visit 1
    0.3
    0.5
    Visit 2
    0.4
    0.4
    Visit 3
    0.3
    0.5
    6. Primary Outcome
    Title Number of Infants Who Received Treatment to Reduce Bilirubin or for Jaundice
    Description Assessment if infant received exchange transfusion, Phototherapy, or other treatment to reduce bilirubin or for jaundice
    Time Frame Assessed from entry through around week 1 after birth

    Outcome Measure Data

    Analysis Population Description
    All infants.
    Arm/Group Title Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Arm/Group Description Infants, who were expected to be ≥2000 grams at birth (i.e. full-term), born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study. Infants, who were expected to be ≤2500 grams at birth (i.e. LBW), born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
    Measure Participants 22 18
    Exchange transfusion therapy
    0
    0%
    0
    0%
    Phototherapy
    1
    4.5%
    4
    22.2%
    Other treatment
    0
    0%
    0
    0%
    7. Secondary Outcome
    Title Neonatal RAL Elimination (T1/2) by UGT1A1 Genotype Group (Normal VS Mutation)
    Description Neonatal RAL elimination was the time required for neonatal plasma concentration to decrease by one-half. Genotyping for polymorphisms of UGT1A1 were performed on infants who were eligible for PK sampling and were consented by their mothers/guardians(i.e. genotyping was optional) . The goal of the genotypic analysis is to determine if certain polymorphisms, particularly those with the UGT1A1*28/*28 genotype have slower RAL elimination than those with the UGT1A1*1/*1 genotype.
    Time Frame Genotype was assessed close to birth and if this is not possible at 1-2 wks after birth. PK samples were collected at 1-5, 8-14, 18-24 and 30-36 hrs after birth for Cohort 1; 1-6, 12-24, 36-48, 72-84 and 108-132 hrs after birth, and day 7-14 for Cohort 2.

    Outcome Measure Data

    Analysis Population Description
    Infants with data on UGT1A1 genotype and RAL half-life (T1/2)
    Arm/Group Title Cohort 1 Infants With UGT1A1 Mutation Cohort 1 Infants With Normal UGT1A1 Phenotype Cohort 2 Infants With UGT1A1 Mutation Cohort 2 Infants With Normal UGT1A1 Phenotype
    Arm/Group Description Cohort 1 (full term) infants with the presence of UGT1A1 *28/*28 genetic variant Cohort 1 (full term) infants with the absence of UGT1A1 *28/*28 genetic variant Cohort 2 (LBW) infants with the presence of UGT1A1 *28/*28 genetic variant Cohort 2 (LBW) infants with the absence of the UGT1A1 *28/*28 genetic variant
    Measure Participants 8 6 9 7
    Median (Inter-Quartile Range) [Hours]
    40.85
    32.75
    21.1
    39.7
    Statistical Analysis 1
    Statistical Analysis Overview Comparison Group Selection Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL, Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Comments To investigate the relationship between neonatal RAL elimination and UGT1A1 genotype in full term infants.
    Type of Statistical Test Other
    Comments
    Statistical Test of Hypothesis p-Value 0.747
    Comments The study was not powered to do the comparison. This was an exploratory analysis.
    Method Wilcoxon (Mann-Whitney)
    Comments
    Statistical Analysis 2
    Statistical Analysis Overview Comparison Group Selection Cohort 2 Infants With UGT1A1 Mutation, Cohort 2 Infants With Normal UGT1A1 Phenotype
    Comments To investigate the relationship between neonatal RAL elimination and UGT1A1 genotype in LBW infants.
    Type of Statistical Test Other
    Comments
    Statistical Test of Hypothesis p-Value 0.341
    Comments The study was not powered to do the comparison. This was an exploratory analysis.
    Method Wilcoxon (Mann-Whitney)
    Comments

    Adverse Events

    Time Frame Birth through 20 weeks and 6 weeks for Cohort 1 and Cohort 2, respectively.
    Adverse Event Reporting Description All Adverse Events (AEs) including diagnoses, signs/symptoms and abnormal laboratory test results mentioned in the "Study Description". LBW is an eligibility criterion for Cohort 2. Prematurity and growth restrictions, which were AEs linked with LBW, were defined in the protocol as baseline events for Cohort 2 infants and were excluded from the "Serious Adverse Events" and "Other Adverse Events tables" for Cohort 2.The DAIDS AE Grading Table (V1.0) and Expedited AE Manual (V2.0) were used.
    Arm/Group Title Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Arm/Group Description Infants, who were expected to be ≥2000 grams at birth (i.e. full-term), born to women with HIV-1 infection who received RAL 400 mg twice daily for at least two weeks prior to delivery and continued to receive ARVs during labor. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study. Infants, who were expected to be ≤2500 grams at birth (i.e. LBW), born to women with HIV-1 infection who received at least one dose of RAL 400 mg within 2 to 24 hours prior to delivery. Raltegravir: No study-specific drugs were given to women or infants during this study. Women received RAL for clinical indications outside of the study.
    All Cause Mortality
    Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 0/22 (0%) 0/18 (0%)
    Serious Adverse Events
    Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 0/22 (0%) 5/18 (27.8%)
    Congenital, familial and genetic disorders
    Congenital syphilis 0/22 (0%) 1/18 (5.6%)
    Hypospadias 0/22 (0%) 1/18 (5.6%)
    Gastrointestinal disorders
    Abdominal distension 0/22 (0%) 1/18 (5.6%)
    Infections and infestations
    Pneumonia 0/22 (0%) 1/18 (5.6%)
    Surgical and medical procedures
    Infection prophylaxis 0/22 (0%) 1/18 (5.6%)
    Other (Not Including Serious) Adverse Events
    Cohort 1: Full Term Infants Exposed in Utero to Maternal RAL Cohort 2: LBW Infants Exposed in Utero to Maternal RAL
    Affected / at Risk (%) # Events Affected / at Risk (%) # Events
    Total 21/22 (95.5%) 16/18 (88.9%)
    Cardiac disorders
    Bradycardia neonatal 0/22 (0%) 1/18 (5.6%)
    Congenital, familial and genetic disorders
    Atrial septal defect 0/22 (0%) 1/18 (5.6%)
    Congenital cardiovascular anomaly 0/22 (0%) 1/18 (5.6%)
    Congenital laryngeal stridor 0/22 (0%) 1/18 (5.6%)
    Congenital umbilical hernia 1/22 (4.5%) 2/18 (11.1%)
    Cryptorchism 0/22 (0%) 1/18 (5.6%)
    Eye disorders
    Conjunctival haemorrhage 0/22 (0%) 1/18 (5.6%)
    Conjunctival pallor 0/22 (0%) 2/18 (11.1%)
    Eyelid oedema 0/22 (0%) 1/18 (5.6%)
    Gastrointestinal disorders
    Constipation 2/22 (9.1%) 0/18 (0%)
    Perianal erythema 0/22 (0%) 1/18 (5.6%)
    Vomiting 2/22 (9.1%) 2/18 (11.1%)
    General disorders
    Decreased activity 0/22 (0%) 1/18 (5.6%)
    Pyrexia 4/22 (18.2%) 2/18 (11.1%)
    Hepatobiliary disorders
    Hepatomegaly 0/22 (0%) 1/18 (5.6%)
    Hyperbilirubinaemia neonatal 0/22 (0%) 1/18 (5.6%)
    Jaundice 0/22 (0%) 4/18 (22.2%)
    Infections and infestations
    Bacterial sepsis 0/22 (0%) 2/18 (11.1%)
    Oral candidiasis 2/22 (9.1%) 0/18 (0%)
    Pneumonia bacterial 1/22 (4.5%) 1/18 (5.6%)
    Upper respiratory tract infection 0/22 (0%) 1/18 (5.6%)
    Investigations
    Aspartate aminotransferase increased 15/22 (68.2%) 3/18 (16.7%)
    Blood bilirubin increased 2/22 (9.1%) 1/18 (5.6%)
    Blood creatinine abnormal 0/22 (0%) 1/18 (5.6%)
    Blood creatinine increased 2/22 (9.1%) 5/18 (27.8%)
    Breath sounds abnormal 2/22 (9.1%) 0/18 (0%)
    Haemoglobin abnormal 1/22 (4.5%) 1/18 (5.6%)
    Haemoglobin decreased 8/22 (36.4%) 5/18 (27.8%)
    Neutrophil count abnormal 0/22 (0%) 2/18 (11.1%)
    Neutrophil count decreased 5/22 (22.7%) 6/18 (33.3%)
    Metabolism and nutrition disorders
    Feeding disorder 0/22 (0%) 1/18 (5.6%)
    Hyperkalaemia 0/22 (0%) 1/18 (5.6%)
    Hypoglycaemia neonatal 1/22 (4.5%) 1/18 (5.6%)
    Malnutrition 0/22 (0%) 1/18 (5.6%)
    Poor feeding infant 0/22 (0%) 1/18 (5.6%)
    Pregnancy, puerperium and perinatal conditions
    Cephalhaematoma 2/22 (9.1%) 0/18 (0%)
    Jaundice neonatal 6/22 (27.3%) 4/18 (22.2%)
    Psychiatric disorders
    Irritability 3/22 (13.6%) 0/18 (0%)
    Respiratory, thoracic and mediastinal disorders
    Cough 2/22 (9.1%) 1/18 (5.6%)
    Dyspnoea 3/22 (13.6%) 1/18 (5.6%)
    Grunting 0/22 (0%) 1/18 (5.6%)
    Nasal congestion 3/22 (13.6%) 0/18 (0%)
    Neonatal hypoxia 0/22 (0%) 1/18 (5.6%)
    Neonatal respiratory distress 0/22 (0%) 1/18 (5.6%)
    Neonatal respiratory distress syndrome 0/22 (0%) 5/18 (27.8%)
    Neonatal tachypnoea 1/22 (4.5%) 2/18 (11.1%)
    Respiratory distress 0/22 (0%) 1/18 (5.6%)
    Rhinorrhoea 2/22 (9.1%) 0/18 (0%)
    Snoring 0/22 (0%) 1/18 (5.6%)
    Use of accessory respiratory muscles 1/22 (4.5%) 1/18 (5.6%)
    Wheezing 2/22 (9.1%) 0/18 (0%)
    Skin and subcutaneous tissue disorders
    Erythema 1/22 (4.5%) 1/18 (5.6%)
    Erythema toxicum neonatorum 0/22 (0%) 1/18 (5.6%)
    Papule 1/22 (4.5%) 1/18 (5.6%)
    Rash 2/22 (9.1%) 0/18 (0%)
    Seborrhoeic dermatitis 2/22 (9.1%) 0/18 (0%)
    Skin exfoliation 1/22 (4.5%) 1/18 (5.6%)
    Skin swelling 0/22 (0%) 1/18 (5.6%)
    Vascular disorders
    Pallor 1/22 (4.5%) 2/18 (11.1%)

    Limitations/Caveats

    [Not Specified]

    More Information

    Certain Agreements

    Principal Investigators are NOT employed by the organization sponsoring the study.

    In accordance with the Clinical Trials Agreement between NIAID (DAIDS) and company collaborators, NIAID (DAIDS) provides companies with a copy of any abstract, press release, or manuscript prior to submission for publication with sufficient time for company review and comment. The publication/other disclosure can be delayed for up to 30 additional business days for manuscripts and five (5) business days for abstracts, to preserve U.S. or foreign patent or other intellectual property rights

    Results Point of Contact

    Name/Title Melissa Allen, Director, IMPAACT Operations Center
    Organization Family Health International (FHI 360)
    Phone (919) 405-1429
    Email mallen@fhi360.org
    Responsible Party:
    National Institute of Allergy and Infectious Diseases (NIAID)
    ClinicalTrials.gov Identifier:
    NCT01828073
    Other Study ID Numbers:
    • P1097
    • 11790
    • IMPAACT P1097
    First Posted:
    Apr 10, 2013
    Last Update Posted:
    Nov 8, 2021
    Last Verified:
    Feb 1, 2020