Association of Prenatal and Postnatal Exposure to

1 downloads 0 Views 295KB Size Report
Jul 6, 2011 - check the French CAH screening data- base and the ... The recommended timing of the test is at age 3 ..... with 9-fludrocortisone and hydrocor-.
PRELIMINARY COMMUNICATION

Association of Prenatal and Postnatal Exposure to Lopinavir-Ritonavir and Adrenal Dysfunction Among Uninfected Infants of HIV-Infected Mothers Albane Simon, MD Josiane Warszawski, MD, PhD Dulanjalee Kariyawasam, MD Jerome Le Chenadec, MSc Valerie Benhammou, PhD Paul Czernichow, MD Frantz Foissac, MD Kathleen Laborde, PhD Jean-Marc Tre´luyer, MD, PhD Ghislaine Firtion, MD Inès Layouni, MD Martine Munzer, MD Franc¸oise Bavoux, MD Michel Polak, MD, PhD Ste´phane Blanche, MD for the ANRS French Perinatal Cohort Study Group

T

HE HUMAN IMMUNODEFICIENCY

virus 1 (HIV-1) transmission rate to newborns is now less than 1% for women treated during pregnancy.1 For pregnant women not optimally treated, as in cases of HIV diagnosis late during pregnancy or persistent viral replication at delivery, several guidelines, observational reports, and the results of a recent controlled study suggest reinforcing the postnatal phase of treatment with a combination of antiretrovirals, as a “postexposure prophylaxis.”2-7 The protease inhibitor lopinavir, with its pharmacological booster ritonavir (lopinavir-ritonavir, Kaletra; Abbott Laboratories, Abbott Park, Illi70

Context Lopinavir-ritonavir is a human immunodeficiency virus 1 (HIV-1) protease inhibitor boosted by ritonavir, a cytochrome p450 inhibitor. A warning about its tolerance in prematurenewbornswasrecentlyreleased,andtransientelevationof17-hydroxyprogesterone (17OHP) was noted in 2 newborns treated with lopinavir-ritonavir in France. Objective To evaluate adrenal function in newborns postnatally treated with lopinavirritonavir. Design, Setting, and Participants Retrospective cross-sectional analysis of the database from the national screening for congenital adrenal hyperplasia (CAH) and the French Perinatal Cohort. Comparison of HIV-1–uninfected newborns postnatally treated with lopinavir-ritonavir and controls treated with standard zidovudine. Main Outcome Measures Plasma 17OHP and dehydroepiandrosterone-sulfate (DHEA-S) concentrations during the first week of treatment. Clinical and biological symptoms compatible with adrenal deficiency. Results Of 50 HIV-1–uninfected newborns who received lopinavir-ritonavir at birth for a median of 30 days (interquartile range [IQR], 25-33), 7 (14%) had elevated 17OHP levels greater than 16.5 ng/mL for term infants (⬎23.1 ng/mL for preterm) on days 1 to 6 vs 0 of 108 controls having elevated levels. The median 17OHP concentration for 42 term newborns treated with lopinavir-ritonavir was 9.9 ng/mL (IQR, 3.9-14.1 ng/mL) vs 3.7 ng/mL (IQR, 2.6-5.3 ng/mL) for 93 term controls (P⬍.001). The difference observed in median 17OHP values between treated newborns and controls was higher in children also exposed in utero (11.5 ng/mL vs 3.7 ng/mL; P⬍.001) than not exposed in utero (6.9 ng/mL vs 3.3 ng/mL; P=.03). The median DHEA-S concentration among 18 term newborns treated with lopinavir-ritonavir was 9242 ng/mL (IQR, 1347-25 986 ng/mL) compared with 484 ng/mL (IQR, 218-1308 ng/mL) among 17 term controls (P⬍.001). The 17OHP and DHEA-S concentrations were positively correlated (r=0.53; P=.001). All term newborns treated with lopinavir-ritonavir were asymptomatic, although 3 premature newborns experienced life-threatening symptoms compatible with adrenal insufficiency, including hyponatremia and hyperkalemia with, in 1 case, cardiogenic shock. All symptoms resolved following completion of the lopinavir-ritonavir treatment. Conclusion Among newborn children of HIV-1–infected mothers exposed in utero to lopinavir-ritonavir, postnatal treatment with a lopinavir-ritonavir–based regimen, compared with a zidovudine-based regimen, was associated with transient adrenal dysfunction. www.jama.com

JAMA. 2011;306(1):70-78

nois), is now the ritonavir-boosted protease inhibitor most widely prescribed in children. 8 Lopinavir-ritonavir is licensed in the United States for HIVinfected newborns older than 14 days and in Europe for children older than 2

JAMA, July 6, 2011—Vol 306, No. 1

Downloaded From: http://jama.jamanetwork.com/ on 02/25/2013

Author Affiliations are listed at the end of this article. Members of the ANRS French Perinatal Cohort Study Group are listed in the eAppendix, available at http: //www.jama.com. Corresponding Author: Ste´phane Blanche, MD, Unite´ d’Immunologie–He´matologie Pe´diatriques, Hoˆpital Necker-Enfants Malades, AP-HP, 149 rue de Sèvres, 75015 Paris, France ([email protected]).

©2011 American Medical Association. All rights reserved.

LOPINAVIR-RITONAVIR AND NEONATAL ADRENAL FUNCTION

years. However, published data concerning its use in newborns are scarce and limited to a few pharmacokinetic studies.8-12 The emergence of toxicity reports in newborns is evidence of its increasing use for postnatal prophylaxis. A possible cardiac toxicity has been described in 2 sets of premature twins in the United States and Europe.13,14 A recent Food and Drug Administration (FDA) warning15 confirms these findings, reporting other cases of lifethreatening conditions in premature newborns. The mechanism of this toxicity is unknown, but possible yet unproven factors may include lopinavir accumulation due to liver immaturity in very young children, possible effects of the propylene glycol and ethanol used as excipients in the lopinavir-ritonavir oral solution, or both. In April 2010, one of the centers of the French national screening program for congenital adrenal hyperplasia (CAH) identified a transient increase of 17hydroxyprogesterone (17OHP) in dried blood spots from 2 children treated at birth with lopinavir-ritonavir. This unusual finding prompted us to crosscheck the French CAH screening database and the French Perinatal Cohort of infants born to HIV-infected mothers. The aim of the study was to assess whether immediate postnatal exposure to lopinavir-ritonavir was associated with changes in adrenal function compared with standard prophylactic zidovudine treatment. METHODS The French Perinatal Cohort is a nationwide prospective multicenter study following up children born to HIVinfected mothers. The goals and design have been described elsewhere.16 Between September 24, 1984, and January 1, 2011, 15 376 mother-child couples have been included at 97 sites. The present analysis was restricted to 32 centers in the Paris area (re´gion Ile de France) at which 2816 couples were included during the study period (December 2004 to September 2008). All HIV-infected pregnant mothers giving informed consent (overall rate of re-

fusal, 4%) and their newborns are anonymously included no later than delivery. Care and prophylaxis strategies are the responsibility of the attending clinicians, who are encouraged to follow national guidelines. In brief, these guidelines—like those in all other Western countries—recommend a combination of antiretroviral treatment during pregnancy, an intravenous infusion of zidovudine during delivery, and zidovudine monotherapy for the newborn lasting 4 to 6 weeks. The guidelines include the possibility of reinforcing postnatal prophylaxis for every case in which the mother’s treatment is considered to be suboptimal.2-4 In French guidelines,2 one of the options recommended for reinforcement of postnatal prophylaxis includes combination therapy based on lopinavir-ritonavir alone with monitoring of plasma concentrations. The French national screening for CAH is based on assaying 17OHP using dried blood collected on filter paper.17 The recommended timing of the test is at age 3 days, and samples are generally collected between days 2 and 5. A 17OHP value of 16.5 ng/mL or greater for full-term infants and of 23.1 ng/mL or greater for preterm infants (⬍37 weeks of gestation) indicates possible 21 hydroxylase deficiency.17 Children with a positive screening result are immediately recalled for clinical evaluation and serum 17OHP determination. During the study period, 692 200 screening tests (mean, 180 253 per year) were performed in the Paris area. The coverage rate of this screening is near 100%, with fewer than 50 refusals annually. Written informed consent and written agreement of 1 of the parents are required for neonatal CAH screening. Participation in the French Perinatal Cohort is based on informed consent. All collected data are sent anonymously to the coordinating center, and only investigators in clinical centers could validate the possible cross identification between the 2 databases. The cross-checking process and the use of frozen samples for additional hormonal investigations were both expressly authorized for this study by an

©2011 American Medical Association. All rights reserved.

Downloaded From: http://jama.jamanetwork.com/ on 02/25/2013

institutional review board (Comite´ de Protection des Personnes, Paris). Study Population

Children treated with postnatal lopinavir-ritonavir prophylaxis were eligible if they were in the Paris area between December 2004 and September 2008, were not HIV-infected, and received lopinavir-ritonavir for at least 3 days or until the screening test for CAH was performed, if not done at age 3 days. Sixty-four children at 15 of 32 sites fulfilled these criteria (lopinavirritonavir–treated group). All non–HIVinfected children who received a postnatal prophylactic regimen free of protease inhibitors were eligible for the control group if they were born during the same period and at the same 15 sites as eligible lopinavir-ritonavir– treated children, to limit site and period effects (n=1402). To facilitate the complex process of confidential crosschecking with CAH, we restricted this process to a subgroup of children whose identity had already been well established on site—in strict respect of confidentiality—during a previous probabilistic cross-matching with French registers of pediatric cancers18 (eFigure, available at http://www.jama.com). Overall, 17OHP screening results were identified with certainty (and the children thus included) for 50 of 64 eligible newborns treated with lopinavirritonavir, and 108 of 142 eligible control newborns. Characteristics of included and nonincluded eligible children in each group were similar, except for a lower birth weight for included children of the group treated with lopinavir-ritonavir (eTable 1), and a higher proportion of recently born children, children of primiparous mothers, and those with late gestational age when pregnancy follow-up began in the control group (eTable 2). When compared with all remaining children of the eligible control group (n=1294), included control children had a higher proportion of recently born children, a higher proportion of undetectable maternal viral load, and a higher birth weight (eTable 3). Matching failure for children entered into JAMA, July 6, 2011—Vol 306, No. 1

71

LOPINAVIR-RITONAVIR AND NEONATAL ADRENAL FUNCTION

Table 1. Characteristics of Mothers, Term Newborns Treated With Lopinavir-Ritonavir During the Postnatal Period, and Term Control Participants a Treatment Group (n = 42) Infant sex Male Female Missing values, No. Period of birth 2004-2006 2007-2008 Missing values, No. Maternal origin Sub-Saharan Africa Other geographical origin Missing values, No. Maternal age ⬍25 y 25-34 y ⱖ35 y Missing values, No. Previous delivery Yes No Missing values, No. Timing of HIV diagnosis Before pregnancy During pregnancy Missing values, No. Gestational age when follow-up began ⬍14 wk 14-27 wk ⱖ28 wk Missing values, No. Gestational term at delivery, median (IQR), wk Type of HIV HIV-1 HIV-2 HIV-1 and -2 Missing values, No. ART during pregnancy Yes No Missing values, No. ART at conception Yes No Missing values, No. HAART based on lopinavir-ritonavir or other ritonavir-boosted protease inhibitor as last ART Yes No Missing values, No. Maternal viral load near delivery ⬍50/mL 50-10 000/mL ⱖ10 000/mL Missing values, No. CD4 cell count near delivery ⬍200 cells/mm3 200-349 cells/mm3 ⱖ350 cells/mm3 Missing values, No.

Control Group (n = 93)

P Value

17 (40.5) 25 (59.5) 0

42 (45.2) 51 (54.8) 0

.61 b

23 (54.8) 19 (45.2) 0

43 (46.2) 50 (53.8) 0

.36 b

28 (68.3) 13 (31.7) 1

79 (85.0) 14 (15.0) 0

.03 b

2 (4.8) 21 (50.0) 19 (45.2) 0

9 (9.7) 46 (49.5) 38 (40.9) 0

32 (80.0) 8 (20.0) 2

68 (73.1) 25 (26.9) 0

.40 b

39 (95.1) 2 (4.9) 1

76 (81.7) 17 (18.3) 0

.04 b

20 (51.2) 12 (30.8) 7 (18.0) 3 38 (38-39)

37 (44.0) 44 (52.4) 3 (3.6) 9 39 (38-40)

40 (95.2) 2 (4.8) 0 0

90 (96.7) 2 (2.2) 1 (1.1) 0

38 (90.5) 4 (9.5) 0

93 (100.0) 0 0

.009 d

25 (61.0) 16 (39.0) 1

44 (47.3) 49 (52.7) 0

.14 b

35 (83.3) 7 (16.7) 0

58 (62.4) 35 (37.6) 0

.01 b

22 (57.9) 14 (36.8) 2 (5.3) 4

70 (79.6) 16 (18.2) 2 (2.3) 5

4 (10.3) 10 (25.6) 25 (64.1) 3

4 (4.7) 15 (17.6) 66 (77.7) 8

.61 b

.007 b

.10 c

.72 c

Statistical Analysis

.03 d

.25 b

(continued)

72

JAMA, July 6, 2011—Vol 306, No. 1

Downloaded From: http://jama.jamanetwork.com/ on 02/25/2013

the process of cross-checking with CAH was mainly due to discrepancy between the name and surname given at birth and the names used later. Rate of matching failure was similar in treated and control groups. Characteristics of mothers and children were available from the French Perinatal Cohort database. Clinical symptoms of children were retrospectively reassessed with the investigating clinician in light of possible adrenal dysfunction. The results of 17OHP assays on dried blood collected on filter paper were available from the CAH database. Plasma samples from frozen plasma banks at the virology laboratories responsible for neonatal HIV diagnosis were available for dehydroepiandrosterone-sulfate (DHEA-S) assays in a subgroup of children and additional 17OHP assays for cases with initially elevated results. Testing for 17OHP levels using dried blood spots on filter paper collected at birth was centralized and performed using a solid-phase time-resolved fluoroimmunoassay method (AutoDELFIA Neonatal 17OHP Kit; Wallac Oy, Turku, Finland). Plasma 17OHP levels and DHEA-S levels were determined by radioimmunoassay as previously described (CISbio International, Gif-surYvette, France, and Immunotech, Marseille, France).19,20 Plasma lopinavir assays involved high-pressure liquid chromatography.10 Statistical analysis was restricted to children born at term. We used a Spearman rank correlation coefficient (r) to test for correlationbetweenlevelsofDHEA-Sand 17OHP. ␹2 or Fisher exact tests were used asappropriateforcategoricalvariablesand t test or Wilcoxon or Kruskal-Wallis tests for continuous variables. P⬍.05 was the threshold for 2-sided statistical significance. SAS version 9.2 (SAS Institute, Cary,NorthCarolina)wasusedforanalyses. The pharmacological data were analyzed using the nonlinear mixed-effect modeling program NONMEM (version VII, release 1) to calculate the analysis of variance and area under the curve for 0 through 24 hours (AUC0-24).21

©2011 American Medical Association. All rights reserved.

LOPINAVIR-RITONAVIR AND NEONATAL ADRENAL FUNCTION

17OHP and DHEA-S Levels

Among the 50 neonates treated with lopinavir-ritonavir, 7 had abnormally high 17OHP results from dried blood spots (⬎16.5 ng/mL at term or ⬎23.1 ng/mL preterm) vs 0 of 108 controls (P ⬍.001). For children born at term, 5 of 42 newborns treated with lopinavirritonavir vs 0 of 93 controls had values greater than 16.5 ng/mL (P=.003).

Table 1. Characteristics of Mothers, Term Newborns Treated With Lopinavir-Ritonavir During the Postnatal Period, and Term Control Participants a (continued) Treatment Group (n = 42) Mode of delivery Vaginal Emergency cesarean delivery Elective cesarean delivery Missing values, No.

Control Group (n = 93)

18 (42.9)

37 (42.1)

7 (16.7)

16 (18.2)

17 (40.5)

35 (39.8)

0

P Value

.97 b

5

Birth weight, median (IQR), g

3090 (2770-3360)

3200 (2820-3500)

.13 c

Birth height, median (IQR), cm

49 (47-50)

49 (47-50)

.87 c

Missing value for height, No.

1

Breastfeeding Yes

0

1 (2.5)

No

0

39 (97.5)

Missing values, No.

2

85 (100)

.32 d

8

Abbreviations: ART, antiretroviral treatment; HAART, highly active antiretroviral treatment; HIV, human immunodeficiency virus; IQR, interquartile range. a Unless otherwise indicated, values are number (percentage). Missing values were excluded from the analysis. b ␹2 Test. c t Test. d Fisher exact test.

Only children born at term who were also exposed to ritonavir-boosted protease inhibitor in utero had values greater than 16.5 ng/mL (14.3%, 5/35). The median 17OHP value for term newborns treated with lopinavir-ritonavir was 9.9 ng/mL (IQR, 3.9-14.1 ng/mL) vs 3.7 ng/mL (IQR, 2.6-5.3 ng/mL) in controls (P⬍.001) (FIGURE 1, TABLE 2, and T ABLE 3). The difference observed in median 17OHP values between treated newborns and controls was higher in children also exposed in utero (11.5 ng/mL vs 3.7 ng/mL; P⬍.001) than not exposed in utero (6.9 ng/mL vs 3.3 ng/mL; P=.03) (Table 3). The 17OHP level was also associated with maternal origin and gestational age when follow-up began and at delivery (Table 2). Frozen samples for DHEA-S determination were available from 20 newborns (18 at term) treated with lopinavirritonavir and 23 (17 at term) of the control group. The characteristics of children tested were no different from those of untested children. Age at sampling was similar in both groups (4 days of life; range, 2-6 days). The median DHEA-S values for children born at term were 9242 ng/mL (IQR, 1347-25 986 ng/ mL) for the treated group vs 484 ng/mL (IQR, 218-1308 ng/mL) for the controls (mean [SD] values for 2-6 days of

©2011 American Medical Association. All rights reserved.

Downloaded From: http://jama.jamanetwork.com/ on 02/25/2013

Figure 1. 17-Hydroxyprogesterone Levels in Term Newborns Collected at Birth Maternal ritonavir-boosted protease inhibitor treatment Mother treated during pregnancy Mother not treated during pregnancy 35 30 25

17OHP, ng/mL

RESULTS Between December 2004 and September 2008, 50 HIV-1 uninfected children received lopinavir-ritonavir just after birth, and 108 received standard prophylaxis: zidovudine alone (n=100), zidovudine and lamivudine (n = 6), or zidovudine and nevirapine (n=2). The median duration of postnatal lopinavirritonavir treatment was 30 days (interquartile range [IQR], 25-33 days), and the median daily dose was 43.8 mg/ kg/d (IQR, 14.9-91.7 mg/kg/d). Lopinavir-ritonavir was administered with lamivudine, zidovudine, or both in 26 cases and was administered alone in 24 cases. The reasons for the particular choice of prophylactic regimen are not routinely collected. Retrospective analysis revealed inadequate treatment of mother (n = 29) or zidovudine resistance of the maternal virus (n=7). The reasons were not provided or were unavailable for 14 cases. Fewer mothers of lopinavir-ritonavir– treated children than control children originated from sub-Saharan Africa (68.3% vs 85%; P=.03), and more mothers began follow-up later in their maternity (18% began follow-up at 28 gestational weeks vs 3.6%; P = .007); as expected, fewer received antiretroviral therapy during pregnancy (90.5% vs 100%; P=.009) and had an undetectable viral load near delivery (57.9% vs 79.6%; P=.03) (TABLE 1). The proportion of mothers who received ritonavirboosted protease inhibitor as the last antiretroviral therapy during pregnancy was higher in the treated group than in the control group (83.3% vs 62.4%; P=.01). In most cases (77.4%), the ritonavirboosted protease inhibitor received by the mothers was lopinavir-ritonavir.

20 15 10 5 0 Control Group (n = 93)

Newborn Postnatal Lopinavir-Ritonavir Treatment Group (n = 42)

Results represent 42 children treated postnatally with lopinavir-ritonavir and 93 controls. All children were born at term, and tests were performed within 2 to 6 days of life. Solid circles indicate mothers treated with ritonavirboosted protease inhibitor during pregnancy (n=35 in treatment group and 58 in control group). Open circles indicate no ritonavir-boosted protease inhibitor during pregnancy (n=7 in treatment group and 35 in control group). Horizontal bar indicates the median value; 17OHP, 17-hydroxyprogesterone.

life: girls at term, 680 [650] ng/mL; boys at term, 820 [640] ng/mL20) (FIGURE 2 and Table 3). DHEA-S and 17OHP values for individual children were JAMA, July 6, 2011—Vol 306, No. 1

73

LOPINAVIR-RITONAVIR AND NEONATAL ADRENAL FUNCTION

Table 2. Factors Associated With Elevated 17-Hydroxyprogesterone Levels in Term Newborns From Univariable Analysis a

Postnatal lopinavir-ritonavir treatment Yes No Missing values Exposure in utero to ritonavir-boosted protease inhibitor, including lopinavir-ritonavir c Yes No Missing values Exposure to ritonavir-boosted protease inhibitor in utero and postnatally Both in utero exposure and postnatal treatment Only postnatal treatment

No. of Participants

17OHP Level, Median (IQR), ng/mL

42

9.9 (3.9-14.1)

93

3.7 (2.6-5.3)

93

4.5 (3.2-8.3)

42

3.5 (2.5-6.1)

35

11.5 (3.7-15.5)

7

6.9 (4.5-9.5) 3.7 (2.8-5.8)

No exposure

35

3.3 (2.4-5.3)

Other geographical origin Missing values Maternal age, y ⬍25

107

3.7 (2.8-6.7)

27

7.2 (3.4-13.8)

11

3.3 (2.2-4.8) 4.9 (3.2-8.2)

ⱖ35

57

3.7 (3.1-7.2)

No Missing values Timing of HIV diagnosis Before pregnancy During pregnancy Missing values Gestational age when follow-up began, wk ⬍14

100

4.2 (3.0-7.3)

33

4.7 (3.3-7.5)

115

4.2 (3.1-8.2)

19

3.8 (2.5-7.2)

4.2 (3.1-8.5)

ⱖ28

10

7.9 (6.9-9.5)

Missing values

12

ART during pregnancy c Yes

131

4.0 (3.1-7.4)

No

4

8.0 (6.0-9.4)

Missing values

0

ART at conception Yes

69

4.5 (3.1-8.6)

65

3.7 (2.6-7.0)

Missing values

92

3.9 (3.0-6.7) 5.3 (3.2-11.6)

ⱖ10 000/mL

4

3.9 (3.0-5.5)

Missing values

9

CD4 cell count near delivery, cells/mm3 ⬍200

.55 e

.005 d

.10 e

.16 b

1

30

50-10 000/mL

.49 b

1

3.7 (3.1-6.0)

Viral load at delivery ⬍50/mL

.10 d

2

57

No

.002 e

0

56

14-27

⬍.001 d

1

67

Missing values

.004 b

0

25-34

Previous delivery Yes

Subsequent Postnatal Hormonal Assays

0

58

Maternal origin Sub-Saharan Africa

⬍.001 b

0

Only in utero exposure Missing values

P Value

8

3.9 (3.5-10.3)

200-350

25

4.5 (3.3-8.3)

ⱖ350

91

4.0 (2.8-7.0)

Missing values

11

.32 d

.62 d

(continued)

74

JAMA, July 6, 2011—Vol 306, No. 1

Downloaded From: http://jama.jamanetwork.com/ on 02/25/2013

strongly positively correlated (n = 35, r=0.53; 95% confidence interval [CI], 0.23-0.73; P=.001). Consistent with the findings for 17OHP, the DHEA-S values were significantly higher only in cases also exposed in utero to ritonavirboosted protease inhibitor.

For 6 of 7 newborns with abnormal screening results, the second test either with dried blood (n=2) or plasma (n=4) on day 9 (n=4) or day 32 (n=1) while still receiving lopinavir-ritonavir treatment indicated normal 17OHP concentrations. One child was lost to follow-up and could not be retested. In addition to the screening control procedure, hormonal assays (17OHP and DHEA-S) were performed retrospectively using frozen samples from the virology centers. Serum 17OHP was assayed in samples collected at 1 month (1 patient), 6 months (2 patients), and 7 months (1 patient) of life from fullterm newborns treated with lopinavirritonavir (and thus after completion of lopinavir-ritonavir treatment). In all 4 cases, the 17OHP levels were normal despite a high 17OHP value at birth for 1 of them. DHEA-S levels after lopinavir-ritonavir completion were normal in 6 cases; in a seventh case, the DHEA-S level was extremely high on day 4 (41 times the normal value), and substantially lower at 1 month, although it remained above normal (2.3 times the normal value). Pharmacological Study

The median prescribed dose of lopinavir (under the responsibility of individual clinicians) was 43.8 mg/kg per day. Two children (premature twins) received 91.7 mg/kg/d for 3 days after birth (this dose is almost 3 times the recommended dose of 30 mg/kg/d for newborns). Plasma concentrations of lopinavir (but not ritonavir) were available for 36 newborns, at a mean (SD) age of 12.8 (12.2) days. The mean (SD) delay between drug intake and lopinavir-ritonavir sampling was 6.2 (4.4) hours. No relationship was found be-

©2011 American Medical Association. All rights reserved.

LOPINAVIR-RITONAVIR AND NEONATAL ADRENAL FUNCTION

Clinical Symptoms of Possible Adrenal Insufficiency

Retrospective review of all clinical symptoms reported in children treated with lopinavir-ritonavir revealed 17 hospitalizations for 17 children during the neonatal period for suspected maternal-fetal bacterial infection (n=4), prematurity (n=4), transient respiratory distress (n=3), acute fetal distress syndrome (n=1),poorsocialcontextprecludingsafe neonatal care and optimal antiretroviral treatment at home (n=2), or various reasons linked to birth (n=3). Three of these children presented with clinical and biological symptoms consistent with adrenal insufficiency (eTable 4). All 3 were premature (30, 34, and 34 weeks of gestation; 2 of them twins) and were exposed in utero to a lopinavir-ritonavir– based combination: lopinavir-ritonavir was administered from birth in all 3 cases. Lopinavir-ritonavir was associated with zidovudine and lamivudine for 1 child and was given alone for 2. For the twins, the dose of lopinavir given was above recommendations (3 times higher), with a residual plasma concentration that was double that expected. Severe metabolic abnormalities were detected 24 hours, 72 hours, and 96 hours, respectively, after the start of treatment. All 3 children had profound hyponatremia with salt loss and hyperkalemia. Electrolytic disorders were severe enough to induce bradyarrhythmia and cardiogenic shock in 1 child. No hypoglycemia was detected. In all cases, treatment was required to normalize the electrolytic disturbances: sodium chloride supplementation was administered in all cases, with 9␣-fludrocortisone and hydrocortisone succinate in 1 case and sodium polystyrene sulfonate in 1 case. In all 3 cases, hormonal disturbances were discovered retrospectively: 17OHP lev-

Table 2. Factors Associated With Elevated 17-Hydroxyprogesterone Levels in Term Newborns From Univariable Analysis a (continued) 17OHP Level, Median (IQR), ng/mL

P Value

55 23 52 5

4.2 (3.2-6.5) 3.3 (2.2-6.9) 4.8 (3.1-8.5)

.15 d

59 76 0 135

4.8 (3.4-7.4) 3.7 (2.7-7.8)

.53 b

−0.25 (−0.40 to −0.08)

.004

No. of Participants Mode of delivery Vaginal Emergency cesarean delivery Elective cesarean delivery Missing values Infant sex Male Female Missing values Gestational term at delivery, Spearman correlation (95% CI)

Abbreviations: 17OHP, 17-hydroxyprogesterone; ART, antiretroviral treatment; CI, confidence interval; HIV, human immunodeficiency virus; IQR, interquartile range. a Missing values were excluded from the analysis. b t Test. c In most cases, the ritonavir-boosted protease inhibitor during pregnancy was lopinavir-ritonavir; for children, only lopinavirritonavir was used. d Kruskal-Wallis test. e Wilcoxon rank sum test.

els in dried blood spots were in the normal range for 2 newborns in contrast with very high DHEA-S levels. 17OHP level was clearly in the pathological range in the third newborn. In all cases, the metabolic perturbations resolved within a few days of the completion of lopinavir-ritonavir treatment. COMMENT In this cohort of HIV-1–uninfected newborns of HIV-1 infected mothers, we found the postnatal treatment with lopinavir-ritonavir was associated with transiently increased 17OHP levels. 17OHP was assayed as part of the national screening program for CAH using dried blood spots collected between 2 and 5 days after birth. Complementary retrospective assays of DHEA-S, using frozen plasma samples, revealed extremely high values, up to 64 times normal levels, during the treatmentperiod(⬍28days).The17OHP and DHEA-S values were strongly positively correlated, although a few children with normal 17OHP results had abnormally high DHEA-S concentrations. Most newborns were asymptomatic during the short period of treatment (4 weeks). However, we retrospectively identified clinical and biological features compatible with a transient but potentially life-threatening adrenal insufficiency in 3 preterm newborns.

©2011 American Medical Association. All rights reserved.

Downloaded From: http://jama.jamanetwork.com/ on 02/25/2013

Figure 2. Plasma DehydroepiandrosteroneSulfate Concentrations During the First Week of Life in Term Newborns Maternal ritonavir-boosted protease inhibitor treatment Mother treated during pregnancy Mother not treated during pregnancy 55 000 50 000 45 000 40 000

DHEA-S, ng/mL

tween 17OHP level and the plasma concentration of lopinavir: tests for correlation between 17OHP levels and AUC 0-24 (r = 0.047, P = .78) and the analysis of variance between normal or abnormal levels of 17OHP and AUC0-24 (P=.50) were not significant.

35 000 30 000 25 000 20 000 15 000 10 000 5000 0 Control Group (n = 17)

Newborn Postnatal Lopinavir-Ritonavir Treatment Group (n = 18)

Results represent 18 term children treated postnatally with lopinavir-ritonavir and 17 controls. Solid circles indicate mothers treated with ritonavir-boosted protease inhibitor during pregnancy (n=16 in treatment group and n=13 in control group). Open circles indicate no ritonavir-boosted protease inhibitor during pregnancy (n=2 in treatment group and n=4 in control group). Horizontal bar indicates the median value; DHEA-S, dehydroepiandrosterone-sulfate.

The incidence of transient abnormal 17OHP levels, as assessed from the screening program in the general French JAMA, July 6, 2011—Vol 306, No. 1

75

LOPINAVIR-RITONAVIR AND NEONATAL ADRENAL FUNCTION

population, is around 0.03%.22 In contrast, 7 of 50 newborns (14%) receiving lopinavir-ritonavir in our cohort had abnormal 17OHP values. Moreover, the difference relative to a control group of infants born to HIV-1–infected mothers receiving a standard postnatal zidovudine treatment was statistically significant. Despite reportedly weak transplacental transfer of lopinavir and ritonavir,23,24 we found that 17OHP levels were highest among infants exposed to lopinavir-ritonavir both during pregnancy and postnatally. Nevertheless, the small number of women not receiving ritonavir-boosted protease inhibitor limits the power of this study to detect a possible small association with postnatal exposure alone. Although these findings cannot establish a causal relationship, further studies are needed to test the hypothesis of whether lopinavirritonavir may act as an inhibitor of adrenal steroid synthesis in fetuses and newborns.

Drug-induced adrenal insufficiency is a well-known phenomenon in adults25 but to our knowledge has not previously been described in neonates. Several drugs act on adrenal enzymes, primarily but not only those belonging to the p450 cytochrome (CYP) family. Concerning adrenal cytochrome enzymes, drugs can either inhibit cortisol synthesis (CYP19A1, CYP11A1, CYP11B1) or activate cortisol metabolism (CYP2B1, CYP2B2, CYP3A4). The fixed-dose combination lopinavir-ritonavir used in this cohort includes ritonavir, a potent inducer or inhibitor of several p450 cytochromes.8 This secondary effect led to its use only as a pharmacological enhancer to “boost” the antiviral effects of other protease inhibitors, notably lopinavir, a substantially less potent cytochrome disruptor. Interaction between ritonavir and adrenal enzymes has been described,26 but with the opposite effect to what we observed here:

the inhibition of CYP3A4 involved in glucocorticoid metabolism leads to deleterious interactions between ritonavir and synthetic steroids, with iatrogenic Cushing syndrome.26 The absence of any reported adrenal insufficiency associated with long-term treatment with lopinavir-ritonavir— either in adults or children—suggests the possibility of a specific susceptibility during the neonatal period. Severe clinical symptoms were only observed in preterm newborns, reinforcing this notion. A relative adrenal insufficiency during the neonatal period transiently worsened by cytochrome inhibition is thus the main hypothesis.27-30 Abnormally high 17OHP and DHEA-S titers are compatible with a 21 hydroxylase deficiency.31 This enzyme (CYP21) is a cytochrome and therefore a potential target for ritonavir inhibition. However, the enzymatic activity—induction, inhibition, or both—of lopinavir-ritonavir on the various newborn adrenal cyto-

Table 3. 17-Hydroxyprogesterone in Dried Blood Spots Collected at Birth (2-6 Days of Life) and Dehydroepiandrosterone Sulfate in Frozen Plasma Collected During the First Week of Life From Term Newborns Treated With Lopinavir-Ritonavir and Control Participants From Bivariable Analysis Treatment Group (n = 42)

Control Group (n = 93)

No. of Participants (Missing Values)

No. (%) or Mean/Median (IQR)

No. of Participants (Missing Values)

42 (0) 35 (0)

5 (11.9) 5 (14.3)

93 (0) 58 (0)

0 0

0

35 (0)

0

17OHP values ⬎16.5 ng/mL a Overall Maternal ART containing ritonavir-boosted protease inhibitor c Maternal ART not containing ritonavir-boosted protease inhibitor 17OHP values, ng/mL a Overall Maternal ART containing ritonavir-boosted protease inhibitor c Maternal ART not containing ritonavir-boosted protease inhibitor DHEA-S values, ng/mL f Overall Maternal ART containing ritonavir-boosted protease inhibitor c Maternal ART not containing ritonavir-boosted protease inhibitor

7 (0)

No. (%) or Mean/Median (IQR)

P Value .003 b .006 b

42 (0) 35 (0)

10.4/9.9 (3.9-14.1) 11.1/11.5 (3.7-15.5)

93 (0) 58 (0)

4.3/3.7 (2.6-5.3) 4.3/3.7 (2.8-5.8)

⬍.001 d ⬍.001 d

7 (0)

6.9/6.9 (4.5-9.5)

35 (0)

4.2/3.3 (2.4-5.3)

.03 e

18 (24) 16 (19)

14 747/9242 (1347-25 986) 16 462/12 252 (4264-28 527)

17 (76) 13 (45)

870/484 (218-1308) 791/484 (218-1308)

⬍.001 e ⬍.001 e

2 (5)

1027/1027 (958-1096)

4 (31)

1124/504 (224-2025)

.53 e

Abbreviations: 17OHP, 17-hydroxyprogesterone; ART, antiretroviral treatment; DHEA-S, dehydroepiandrosterone sulfate; IQR, interquartile range. a Normal values for 17OHP (term newborn): ⬍16.5 ng/mL. b Fisher exact test. c In cases of several different treatments during the pregnancy, the composition reported (with or without ritonavir-boosted protease inhibitor) is based on the last regimen received during pregnancy. d t Test. e Wilcoxon rank-sum test. f Normal values for DHEA-S (term newborn): ⬍1300 ng/mL.

76

JAMA, July 6, 2011—Vol 306, No. 1

Downloaded From: http://jama.jamanetwork.com/ on 02/25/2013

©2011 American Medical Association. All rights reserved.

LOPINAVIR-RITONAVIR AND NEONATAL ADRENAL FUNCTION

chromes remains to be evaluated precisely in vitro, as does the potential role of geographical gene polymorphisms.32 The use of lopinavir-ritonavir in newborns is recent but increasing.15 Although data concerning newborn tolerance are still limited, asymptomatic grade 3 serum hyponatremia and hyperkalemia disturbances in 2 newborns were noted in the first pharmacology study.9 These unusual biological disturbances in children born to HIVinfected mothers may have been linked to adrenal dysfunction. Electrolyte analyses were not recorded in the routine follow-up of the newborns in our study, and therefore, we cannot exclude occult electrolyte disequilibrium. The cardiac rhythm abnormalities and cardiogenic shock observed in 1 premature infant were considered probably related to electrolyte disturbances rather than direct cardiac toxicity. The triad of “shock—hyponatremia with salt loss and renal insufficiency— hyperkalemia” is the classical presentation of the early and severe form of adrenal congenital deficiency.33 Specific cardiac toxicity of lopinavir has been discussed in case reports,13,14 but potassium determinations were not reported. Cardiac dysfunction was also noted in 6 of the 10 cases included in the recent FDA warning.15 The authors considered possible specific lopinavir toxicity but also the possibility of a toxic effect of the propylene glycol and ethanol excipients. Four of those 10 children had hyperkalemia, and it is possible that some of them also had cardiac dysfunction induced by hyperkalemia and salt loss–induced dehydration. There is not necessarily a single mechanism of the apparent toxicity of lopinavir-ritonavir in newborns. Although the drug dose administered to some newborns in our study was higher than recommended, there was no clear link between circulating concentrations of 17OHP and lopinavir. An evaluation of ritonavir titers would be interesting, although cytochrome inhibitory or induction effects can be observed at very low concentrations. Further studies are needed to investigate in

more detail both the consequences of lopinavir and its booster ritonavir on adrenal function and the mechanism of cardiac toxicity in premature newborns. In summary, our findings of the association between lopinavir-ritonavir and transient adrenal dysfunction in HIV-1 uninfected newborns suggest that lopinavir-ritonavir and more generally ritonavir boosting should be used with caution, if at all, in premature infants, and if this drug regimen is administered to full-term infants, it should be used under electrolyte monitoring. Whether more prolonged exposure of HIV-1-infected or uninfected infants via breast milk34 is associated with endocrine disruption should be carefully investigated, and the apparent risk associated with prenatal ritonavir exposure also merits further evaluation. Author Affiliations: Unite´ d’Endocrinologie Pe´diatrique (Drs Simon, Kariyawasam, and Polak), Association Franc¸aise pour le De´pistage et la Pre´vention des Handicaps de l’Enfant (Dr Czernichow), Unite´ de Recherche Clinique Paris-Centre (Drs Foissac and Tre´luyer), Laboratoire d’Explorations Fonctionnelles (Dr Laborde), INSERM U845 (Dr Polak), and Unite´ d’Immunologie–He´ matologie Pe´ diatriques (Dr Blanche), Hoˆpital Necker-Enfants Malades, Assistance Publique-Hoˆpitaux de Paris (AP-HP), Paris, France; EA 3620 (Drs Foissac, Tre´luyer, and Blanche), Universite´ Paris Descartes (Drs Simon, Kariyawasam, and Polak), Paris; INSERM CESP U1018 Equipe VIH et IST (Dr Warszawski, Mr Le Chenadec, and Dr Benhammou) and Universite´ et Faculte´ de Me´decine ParisSud (Dr Warszawski), Le Kremlin-Biceˆtre, France; Service d’Epide´miologie et Sante´ Publique, Hoˆpital Biceˆtre, AP-HP (Dr Warszawski); Service de Ne´onatologie, Hoˆpital Cochin/Port Royal, AP-HP (Dr Firtion); Service de Ne´onatologie, Hoˆpital Intercommunal, Cre´teil, France (Dr Layouni); Service de Pe´diatrie, Centre Hospitalo-Universitaire de Reims, Reims, France (Dr Munzer); and Unite´ de Pharmacovigilance Pe´ diatrique, Hoˆpital Cochin/Saint Vincent de Paul, AP-HP (Dr Bavoux). Author Contributions: Dr Blanche had full access to all of the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis. Study concept and design: Simon, Warszawski, Kariyawasam, Polak, Blanche. Acquisition of data: Simon, Warszawski, Kariyawasam, Benhammou, Foissac, Laborde, Tre´luyer, Firtion, Layouni, Munzer, Bavoux, Polak, Blanche. Analysis and interpretation of data: Simon, Warszawski, Kariyawasam, Le Chenadec, Czernichow, Foissac, Laborde, Polak, Blanche. Drafting of the manuscript: Simon, Warszawski, Kariyawasam, Le Chenadec, Benhammou, Czernichow, Foissac, Tre´luyer, Polak, Blanche. Critical revision of the manuscript for important intellectual content: Warszawski, Le Chenadec, Laborde, Tre´luyer, Firtion, Layouni, Munzer, Bavoux, Polak, Blanche. Statistical analysis: Warszawski, Le Chenadec, Foissac, Tre´luyer.

©2011 American Medical Association. All rights reserved.

Downloaded From: http://jama.jamanetwork.com/ on 02/25/2013

Obtained funding: Warszawski, Blanche. Administrative, technical, or material support: Simon, Benhammou, Czernichow, Laborde, Blanche. Study supervision: Polak, Blanche. Conflict of Interest Disclosures: All authors have completed and submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Dr Warszawski reported having obtained a research grant from Abbott Laboratories for another study concerning lopinavir-ritonavir in children. Dr Polak reported having received support for educational presentations and grant support from Pfizer, IPSEN, and Sandoz-SAS Laboratories. Dr Blanche reported having received support for travel to meetings from Abbott Laboratories and honoraria for lectures and for development of educational presentations from Astellas, Boehringer Ingelheim, GlaxoSmithKline, and Tibotec Laboratories. No other disclosures were reported. Funding/Support: This study was supported by the French Agence Nationale de Recherche sur le SIDA (ANRS) and Agence Franc¸aise de Se´curite´ Sanitaire et des Produits de Sante´ (AFSSAPS). Role of the Sponsor: The sponsors had no role in the design and conduct of the study; in the collection, analysis, and interpretation of the data; or in the preparation, review, or approval of the manuscript. Online-Only Material: The eAppendix, eFigure, and eTables 1 through 4 are available at http://www.jama .com. Additional Contributions: We thank Jean Paul Teglas, MSc, INSERM CESP U1018, for his help with database management and statistical analysis, for which he did not receive compensation. We are indebted to all families who agreed to participate in the French Perinatal Cohort.

REFERENCES 1. Coovadia H. Current issues in prevention of motherto-child transmission of HIV-1. Curr Opin HIV AIDS. 2009;4(4):319-324. 2. Prise en charge me´dicale des personnes infecte´es par le VIH: recommandations du groupe d’experts, rapport 2010. http://www.sante.gouv.fr /rapport-2010-sur-la-prise-en-charge-medicale -des-personnes-infectees-par-le-vih-sous-la -direction-du-pr-patrick-yeni.html. Accessed May 19, 2011. 3. Recommendations for use of antiretroviral drugs in pregnant HIV-1-infected women for maternal health and interventions to reduce perinatal HIV transmission in the United States. Panel on Treatment of HIV-Infected Pregnant Women and Prevention of Perinatal Transmission. http://aidsinfo.nih.gov /ContentFiles/PerinatalGL.pdf. Accessed May 19, 2011. 4. Lyall EG, Blott M, de Ruiter A, et al; British HIV Association. Guidelines for the management of HIV infection in pregnant women and the prevention of mother-to-child transmission. HIV Med. 2001; 2(4):314-334. 5. Chiappini E, Galli L, Tovo PA, et al. Use of postnatal antiretroviral prophylaxis for prevention of mother-to-child transmission of HIV is increasing in Italy. Clin Infect Dis. 2009;49(10):1618-1619. 6. Haile-Selassie H, Townsend C, Tookey P. Use of neonatal post-exposure prophylaxis for prevention of mother-to-child HIV transmission in the UK and Ireland, 2001-2008 [published online January 20, 2011]. HIV Med. doi:10.1111/j.1468-1293.2010.00902.x. 7. Nielsen KN, Watts H, Gonc¸alves Veloso V, et al. Phase III randomized trial of the safety and efficacy of 3 neonatal ARV regimes for prevention of intrapartum HIV-1 transmission: NICHD HPTN 040 /PACTG 1043 [abstract 124 LB]. Paper presented at: 18th Conference on Retrovirus and Opportunistic infection (CROI); March 2, 2011; Boston, MA. 8. Croxtall JD, Perry CM. Lopinavir/ritonavir: a reJAMA, July 6, 2011—Vol 306, No. 1

77

LOPINAVIR-RITONAVIR AND NEONATAL ADRENAL FUNCTION view of its use in the management of HIV-1 infection. Drugs. 2010;70(14):1885-1915. 9. Chadwick EG, Capparelli EV, Yogev R, et al; P1030 Team. Pharmacokinetics, safety and efficacy of lopinavir /ritonavir in infants less than 6 months of age: 24 week results. AIDS. 2008;22(2):249-255. 10. Jullien V, Urien S, Hirt D, et al. Population analysis of weight-, age-, and sex-related differences in the pharmacokinetics of lopinavir in children from birth to 18 years. Antimicrob Agents Chemother. 2006; 50(11):3548-3555. 11. Chadwick EG, Pinto J, Yogev R, et al; International Maternal Pediatric Adolescent Clinical Trials Group (IMPAACT) P1030 Team. Early initiation of lopinavir/ritonavir in infants less than 6 weeks of age: pharmacokinetics and 24-week safety and efficacy. Pediatr Infect Dis J. 2009;28(3):215-219. 12. Urien S, Firtion G, Anderson ST, et al. Lopinavir /ritonavir population pharmacokinetics in neonates and infants. Br J Clin Pharmacol. 2011;71(6):956-960. 13. McArthur MA, Kalu SU, Foulks AR, Aly AM, Jain SK, Patel JA. Twin preterm neonates with cardiac toxicity related to lopinavir/ritonavir therapy. Pediatr Infect Dis J. 2009;28(12):1127-1129. 14. Lopriore E, Rozendaal L, Gelinck LB, Bo¨kenkamp R, Boelen CC, Walther FJ. Twins with cardiomyopathy and complete heart block born to an HIVinfected mother treated with HAART. AIDS. 2007; 21(18):2564-2565. 15. Kaletra (lopinavir/ritonavir): label change: serious health problems in premature babies. US Food and Drug Administration. http://www.fda.gov /Safety/MedWatch/SafetyInformation/Safety AlertsforHumanMedicalProducts/ucm246167.htm. Accessed March 10, 2011. 16. Blanche S, Rouzioux C, Moscato ML, et al; HIV Infection in Newborns French Collaborative

78

Study Group. A prospective study of infants born to women seropositive for human immunodeficiency virus type 1. N Engl J Med. 1989;320(25):16431648. 17. Dhondt JL, Dorche C, Farriaux JP, Courte C. Neonatal screening for congenital adrenal hyperplasia: a pilot study in France. J Inherit Metab Dis. 1986; 9(suppl 1):147-151. 18. Benhammou V, Warszawski J, Bellec S, et al; ANRSEnqueˆte Pe´rinatale Franc¸aise. Incidence of cancer in children perinatally exposed to nucleoside reverse transcriptase inhibitors. AIDS. 2008;22(16):2165-2177. 19. Forest MG. Adrenal function tests. In: Ranke MB, ed. Diagnostics of Endocrine Function in Children and Adolescents. Basel, Switzerland: Karger; 2003: 372-426. 20. de Peretti E, Forest MG. Pattern of plasma dehydroepiandrosterone sulfate levels in humans from birth to adulthood: evidence for testicular production. J Clin Endocrinol Metab. 1978;47(3):572-577. 21. Beal SL, Sheiner LB, ed. NONMEM Users’ Guide. San Francisco, CA: NONMEM Project Group;1992. 22. Cavarzere P, Samara-Boustani D, Flechtner I, et al. Transient hyper-17-hydroxyprogesteronemia: a clinical subgroup of patients diagnosed at neonatal screening for congenital adrenal hyperplasia. Eur J Endocrinol. 2009;161(2):285-292. 23. Gingelmaier A, Kurowski M, Ka¨stner R, et al. Placental transfer and pharmacokinetics of lopinavir and other protease inhibitors in combination with nevirapine at delivery. AIDS. 2006;20(13):1737-1743. 24. Yeh RF, Rezk NL, Kashuba AD, et al. Genital tract, cord blood, and amniotic fluid exposures of seven antiretroviral drugs during and after pregnancy in human immunodeficiency virus type 1-infected women. Antimicrob Agents Chemother. 2009;53(6):23672374.

JAMA, July 6, 2011—Vol 306, No. 1

Downloaded From: http://jama.jamanetwork.com/ on 02/25/2013

25. Bornstein SR. Predisposing factors for adrenal insufficiency. N Engl J Med. 2009;360(22):23282339. 26. Foisy MM, Yakiwchuk EMK, Chiu I, Singh AE. Adrenal suppression and Cushing’s syndrome secondary to an interaction between ritonavir and fluticasone: a review of the literature. HIV Med. 2008; 9(6):389-396. 27. Watterberg KL. Adrenocortical function and dysfunction in the fetus and neonate. Semin Neonatol. 2004;9(1):13-21. 28. Fernandez EF, Watterberg KL. Relative adrenal insufficiency in the preterm and term infant. J Perinatol. 2009;29(suppl 2):S44-S49. 29. Martinerie L, Viengchareun S, Delezoide AL, et al. Low renal mineralocorticoid receptor expression at birth contributes to partial aldosterone resistance in neonates. Endocrinology. 2009;150(9):44144424. 30. Martinerie L, Pussard E, Foix-L’He´lias L, et al. Physiological partial aldosterone resistance in human newborns. Pediatr Res. 2009;66(3):323-328. 31. Neary N, Nieman L. Adrenal insufficiency: etiology, diagnosis and treatment. Curr Opin Endocrinol Diabetes Obes. 2010;17(3):217-223. 32. Ozturk IC, Wei WL, Palaniappan L, Rubenfire M, Killeen AA. Analysis of CYP21 coding polymorphisms in three ethnic populations: further evidence of nonamplifying CYP21 alleles among whites. Mol Diagn. 2000;5(1):47-52. 33. White PC; Medscape. Neonatal screening for congenital adrenal hyperplasia. Nat Rev Endocrinol. 2009; 5(9):490-498. 34. Shapiro RL, Hughes MD, Ogwu A, et al. Antiretroviral regimens in pregnancy and breast-feeding in Botswana. N Engl J Med. 2010;362(24):22822294.

©2011 American Medical Association. All rights reserved.