Original Research

ajog.org

OBSTETRICS

Safety and pharmacokinetics of pravastatin used for the prevention of preeclampsia in high-risk pregnant women: a pilot randomized controlled trial Maged M. Costantine, MD; Kirsten Cleary, MD; Mary F. Hebert, PharmD, FCCP; Mahmoud S. Ahmed, PhD; Linda M. Brown, DrPH; Zhaoxia Ren, MD, PhD; Thomas R. Easterling, MD; David M. Haas, MD, MS; Laura S. Haneline, MD; Steve N. Caritis, MD; Raman Venkataramanan, PhD; Holly West, DHEd; Mary D’Alton, MD; Gary Hankins, MD; for the Eunice Kennedy Shriver National Institute of Child Health and Human Development Obstetric-Fetal Pharmacology Research Units Network

BACKGROUND: Preeclampsia complicates approximately 3e5% of pregnancies and remains a major cause of maternal and neonatal morbidity and mortality. It shares pathogenic similarities with adult cardiovascular disease as well as many risk factors. Pravastatin, a hydrophilic, 3-hydroxy-3methyl-glutaryl-coenzyme A reductase inhibitor, has been shown in preclinical studies to reverse various pathophysiological pathways associated with preeclampsia, providing biological plausibility for its use for preeclampsia prevention. However, human trials are lacking. OBJECTIVE: As an initial step in evaluating the utility of pravastatin in preventing preeclampsia and after consultation with the US Food and Drug Administration, we undertook a pilot randomized controlled trial with the objective to determine pravastatin safety and pharmacokinetic parameters when used in pregnant women at high risk of preeclampsia. STUDY DESIGN: We conducted a pilot, multicenter, double-blind, placebo-controlled, randomized trial of women with singleton, nonanomalous pregnancies at high risk for preeclampsia. Women between 120/7 and 166/7 weeks’ gestation were assigned to daily pravastatin 10 mg or placebo orally until delivery. Primary outcomes were maternal-fetal safety and pharmacokinetic parameters of pravastatin during pregnancy. Secondary outcomes included rates of preeclampsia and preterm delivery, gestational age at delivery, birthweight, and maternal and cord blood lipid profile (clinicaltrials.gov identifier NCT01717586).

P

reeclampsia is a multisystem disorder that complicates 3-5% of pregnancies and remains a major cause of maternal, fetal, and neonatal morbidity and mortality.1 It is characterized by angiogenic imbalance, exaggerated inflammation, and endothelial dysfunction, which ultimately lead to the clinical manifestations of hypertension, proteinuria, and end organ damage.1,2 Cite this article as: Costantine MM, Cleary K, Hebert MF, et al. Safety and pharmacokinetics of pravastatin used for the prevention of preeclampsia in high-risk pregnant women: a pilot randomized controlled trial. Am J Obstet Gynecol 2016;214:720.e1-17. 0002-9378/$36.00 ª 2015 Elsevier Inc. All rights reserved. http://dx.doi.org/10.1016/j.ajog.2015.12.038

RESULTS: Ten women assigned to pravastatin and 10 to placebo

completed the trial. There were no differences between the 2 groups in rates of study drug side effects, congenital anomalies, or other adverse or serious adverse events. There was no maternal, fetal, or neonatal death. Pravastatin renal clearance was significantly higher in pregnancy compared with postpartum. Four subjects in the placebo group developed preeclampsia compared with none in the pravastatin group. Although pravastatin reduced maternal cholesterol concentrations, umbilical cord cholesterol concentrations and infant birthweight were not different between the groups. The majority of umbilical cord and maternal pravastatin plasma concentrations at the time of delivery were below the lower limit of quantification of the assay. Pravastatin use was associated with a more favorable pregnancy angiogenic profile. CONCLUSION: This study provides preliminary safety and pharmacokinetic data regarding the use of pravastatin for preventing preeclampsia in high-risk pregnant women. Although the data are preliminary, no identifiable safety risks were associated with pravastatin use in this cohort. This favorable risk-benefit analysis justifies using pravastatin in a larger clinical trial with dose escalation. Key words: angiogenic, pharmacokinetics, pravastatin, preeclampsia,

safety

Preeclampsia is associated with serious short- and long-term maternal and neonatal morbidities,1,3 and its recurrence in subsequent pregnancies depends on the presence of risk factors (eg, diabetes, hypertension, and multifetal gestation) and the severity and time of onset of preeclampsia in a prior pregnancy.4,5 Despite being unique to pregnancy, preeclampsia shares pathogenic similarities and many risk factors with adult cardiovascular disease.4 Endothelial dysfunction and inflammation are fundamental for the initiation and progression of both atherosclerosis and preeclampsia.2,6-8 Numerous attempts at primary and secondary prevention of preeclampsia, using various

720.e1 American Journal of Obstetrics & Gynecology JUNE 2016

supplements and medications, have had limited success.4 Only low-dose aspirin was found to have a modest benefit in reducing the rate of preeclampsia in an individual patient metaanalysis,9 and that benefit was achieved only if the drug was started before 16 weeks’ gestational age. On the contrary, inhibitors of 3-hydroxy-3-methylglutaryl-coenzymeA (HMG-CoA) reductase (statins) are effective in primary and secondary prevention of cardiovascular mortality and morbidity.10,11 Moreover, statins have been used in animal models of preeclampsia to revert the angiogenic imbalance, a hallmark of preeclampsia, and restore endothelial dysfunction. This biological plausibility and data

ajog.org from preclinical animal studies support a role for statins in preeclampsia prevention.12-19 Our long-term goal is to evaluate the utility of pravastatin (a hydrophilic statin) to reduce the recurrence of preeclampsia in high-risk pregnant women. As an initial step in this process, and after consultation with the US Food and Drug Administration (FDA), we undertook a pilot randomized controlled trial with an objective to evaluate the maternal-fetal safety and pharmacokinetic (PK) parameters of pravastatin when used in pregnant women at high risk for preeclampsia.19 In this publication, we are reporting the first phase of a series of planned studies using a low dose (10 mg) of pravastatin.

Materials and Methods Study population We conducted a multicenter, doubleblind, placebo-controlled randomized trial involving pregnant women at high risk for preeclampsia. Eligible women were 18 years old or older, with singleton, nonanomalous pregnancy between 120/7 weeks and 166/7 weeks’ gestation (confirmed with an ultrasound examination), and with a history of severe preeclampsia in a prior pregnancy that required delivery prior to 34 weeks’ gestation (documented by chart review). We excluded women with known fetal genetic or major malformations; fetal demise; multifetal gestation; contraindications for statin therapy (eg, hypersensitivity to pravastatin, recent or active liver disease); concomitant therapy with fibrates, niacin, cyclosporine, clarithromycin, or erythromycin; pregestational diabetes mellitus; human immunodeficiency virus infection; history of solid organ transplant; chronic renal disease; epilepsy; uterine malformations; cancer; familial hypercholesterolemia; or inability to tolerate oral medications secondary to severe nausea and vomiting of pregnancy. The trial was conducted from August 2012 through February 2014 by the Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD) Obstetric-Fetal Pharmacology Research Units Network

OBSTETRICS

at 5 clinical center sites as an FDA-approved investigational new drug study (IND; number 114205).19 The institutional review boards at all the participating sites approved the study protocol. All women provided written informed consent. The study was registered on clinicaltrials.gov (identifier number NCT01717586).

Study design and intervention Before randomization, all participants were documented to have normal liver transaminases (aminotransferase [AST] and aspartate aminotransferase [ALT]). Women were randomized to pravastatin 10 mg or placebo and were assigned a prepackaged supply of study medication corresponding to the appropriate study drug code. Randomization was performed through a central process that was prepared and maintained by the data coordinating center (RTI International, Research Triangle Park, NC). Initial stratification was by clinical site. Pravastatin and placebo capsules were manufactured by University of Iowa Pharmaceuticals and packaged in identical capsules. Subjects were asked to take 1 capsule orally daily, and treatment continued until delivery or until a condition developed that required discontinuation of the study drug. After randomization, research personnel followed up subjects at scheduled intervals. Subjects’ care and that of their infants was according to standard practice. At each study visit, medication’s side effects were assessed using a checklist, adverse events (AEs) were determined and assessed, and pill count performed. Subjects’ pregnancy management (including antenatal testing, ultrasounds, management of preeclampsia, use of low dose aspirin, and others) was left to the discretion of the treating physician and performed as recommended by standard prenatal care as defined by the respective participating institution. All data were collected or abstracted by research coordinators at the clinical centers and uploaded to a central database that was managed by the data coordinating center, which was responsible for data analysis.

Original Research

Pharmacokinetic studies Steady-state pravastatin PK studies were conducted in the second trimester (18e24 weeks’ gestation) and third trimester (30e34 weeks’ gestation) of pregnancy as well as postpartum (4e6 months after the delivery). Each subject served as her own control. Subjects recorded the time of pravastatin dosing for the 4 days prior to each study day, and pill counts were conducted to determine adherence. Women were asked to fast (except for water) for 5 hours prior to each study visit until 1 hour after dosing. Serial blood samples (6 mL each) were collected for measurement of pravastatin and 30 a-isopravastatin (a major metabolite of pravastatin, that is only 1-10th to 1-40th as potent as parent drug in inhibiting HMG-CoA reductase) concentrations in plasma at times: before the dose, and at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 10, 12, and 24 hours after the dose on each pharmacokinetic study day. Urine was collected before the dose, and then all urine over 1 dosing interval was collected as follows: 0e4, 4e8, 8e12, and 12e24 hours following the dosing on each study day. Urine from each interval was combined, mixed, and total volume measured. An aliquot from each interval was assayed for pravastatin and 30 a-isopravastatin concentrations. Maternal, umbilical cord venous and umbilical cord arterial blood samples were collected at the time of delivery for measurement of pravastatin and 30 a-isopravastatin concentrations in plasma. All samples were stored at e70 C until analysis (more details on PK studies and analysis will be found in the supplemental materials).

Outcome variables The primary outcomes were the maternal-fetal safety and the pravastatin PK parameters during pregnancy. Safety outcomes included evaluation of medication side effects (checklist), maternal AEs, and serious AEs as well as fetal or perinatal death, and congenital malformations. Pravastatin PK parameters included maximum concentration (Cmax) and time to

JUNE 2016 American Journal of Obstetrics & Gynecology

720.e2

Original Research

ajog.org

OBSTETRICS

maximum concentration (Tmax), area under the concentration time curve (AUC), apparent oral clearance (CL/F), half-life, renal clearance, and others (supplemental material). In addition, the study collected secondary maternal and fetal/neonatal outcomes including rate and severity of preeclampsia, gestational age at delivery, rate of preterm delivery, maternal lipid profile, and the concentrations of angiogenic (placental growth factor [PlGF]) and antiangiogenic factors (soluble fms-like tyrosine kinase-1 [sFlt1]; and soluble endoglin [sEng]) in the maternal circulation. Preeclampsia was diagnosed according to criteria set by the American College of Obstetricians and Gynecologists20 (see supplemental material), and the diagnosis (or absence) was confirmed by a panel of 3 maternal-fetal medicine physicians, blinded to treatment assignment, who reviewed the deidentified medical records of all enrolled subjects. Fetal and neonatal secondary outcomes included birthweight, rates of small for gestational age, failure of auditory brainstem response evoked potential, admission to the neonatal intensive care unit (NICU) and other neonatal complications, and cord blood analytes (concentrations of pravastatin and 30 a-isopravastatin, liver enzymes, lipid profile, creatine kinase, angiogenic and antiangiogenic markers, steroidogenic hormones (thyrotropin, folliclestimulating hormone, luteinizing hormone, estradiol, and total testosterone), and S100B and neuron specific enolase, 2 nonspecific markers of neurological injury).

Statistical analysis Statistical analyses were performed using SAS statistical software (SAS Institute, Cary, NC). Maternal and neonatal continuous variables were compared using Wilcoxon rank-sum and categorical variables with the c2 or Fisher exact test as appropriate. Biomarker concentrations were analyzed as continuous variables. Steady-state pravastatin PK parameters were estimated using standard noncompartmental techniques and

TABLE 1

Baseline characteristics of subjects who participated in the study Placebo groupa (n ¼ 10)

Characteristic

Pravastatin groupa (n ¼ 11)

Raceb White

9

10

African American

1

0

Asian

0

0

0

1

Hispanic

7

5

Non-Hispanic

3

6

American Indian b

Ethnicity

Age, y Body mass index, kg/m

2

Obesityc Systolic blood pressure at entry to care, mm Hg

27 [21, 34]

29.6 [27, 32.3]

36 [26, 38.2]

4 (40) d

Diastolic blood pressure at entry to care, mm Hg Parity

30 [27, 34]

d

e

8 (72.7)

115 [110, 122]

109 [107, 131]

68 [64, 72]

64 [55, 77]

2 [2, 3]

1 [1, 2]

Gestational age at randomization, wks

14.9 [13.4, 16.4] 13.9 [13.3, 16.1]

Gestational age at delivery in prior pregnancy, wks

30.7 [29.4, 32.0] 32.0 [30.7, 33.0]

Chronic hypertension

3 (30)

5 (50)

Use of low-dose aspirin

3 (30)

2 (18)

Data are reported as median [interquartile range] or n (percentage). None of the comparisons between the 2 groups is statistically significant (P > .05); b Race and ethnicity were self-reported by patients; c Obesity is defined as body mass index of  30 kg/m2 using prepregnancy weight; d Blood pressure at entry to care, measured in clinic after a 10 minute rest period, in seating position with the right arm in a roughly horizontal position at heart level, supported on a desk; e Parity is any pregnancy that lasted > 20 weeks. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016. a

normalized using actual body weights (see supplemental material). PK parameters during pregnancy were compared with those postpartum using a paired Wilcoxon signed-rank test. Our sample size was 20 subjects (10 assigned to pravastatin and 10 to placebo) for which the primary outcomes were available. This was determined a priori by the FDA as part of the IND approval process19 and was not intended to achieve power to detect hypothetical differences in primary or secondary clinical outcomes or other laboratory values. A value of P < .05 was considered statistically significant.

Results Of 22 subjects who consented for the study, 21 were randomized, with 11

720.e3 American Journal of Obstetrics & Gynecology JUNE 2016

assigned to the pravastatin group and 10 to the placebo group. One subject from the pravastatin group withdrew from the study after randomization for social reasons (Supplemental Figure 1). Ten subjects in each group completed the trial, as requested by the FDA. No subjects were lost to follow-up. There was no significant difference in estimated adherence to study medication between the pravastatin group and placebo group (94.6% vs 95.9%). At study entry, there were no differences in baseline characteristics such as gestational age at delivery in prior qualifying pregnancy and the percentage of subjects receiving low-dose aspirin. Although statistically nonsignificant, more subjects in the pravastatin group were obese (Table 1 and Supplemental Figure 2).

ajog.org

OBSTETRICS

TABLE 2

Adverse and serious adverse events experienced by subjects, irrespective of association with study medications Placebo groupa (n ¼ 10)

Pravastatin groupa (n ¼ 11)

Heartburn

3 (30)

4 (36)

Musculoskeletal pain

1 (10)

4 (36)

Dizziness

2 (20)

3 (27)

Chest pain

0

2 (18)

Diarrhea

1 (10)

2 (18)

Headache

3 (30)

2 (18)

Cough

1 (10)

2 (18)

Swelling

0

2 (18)

Nausea

1 (10)

1 (9)

Fever

2 (20)

1 (9)

Flatulence

0

1 (9)

Fatigue

0

1 (9)

Wheezing

0

1 (9)

Vomiting

1 (10)

0

Influenza-like symptoms

2 (20)

0

0

0

0

0

Condition Adverse events

Serious adverse events Maternal, fetal, or infant death Rhabdomyolysis Liver injury

b

b

0

0

Polydactylyc Ventriculomegaly

Hypospadiasc Coarctation of aorta

HTN/BP exacerbation,

3 (30)

2 (18)

preeclampsia workup

1 (10)

0

Vaginal bleeding

1 (10)

0

Influenza infection

1 (10)

0

Migraine

1 (10)

0

Syncope

0

1 (9)

Congenital anomalies Hospitalization > 24 h

Data are reported as n (percentage). ALT, aspartate aminotransferase; AST, aminotransferase; BP, blood pressure; CK, creatinine kinase; HTN, hypertension. None of the comparisons between the 2 groups is statistically significant (P > .05); b Rhabdomyolysis was defined as muscle pain or muscle weakness in conjunction with increase in CK values to greater than 10 times the upper limit of normal. Liver injury was diagnosed with elevation of transaminases (AST or ALT) values greater than 3 times the upper limit of normal. Data on maternal AST, ALT, and CK concentrations are in Supplemental Table 1; c Family history of polydactyly and hypospadias in the father of each child, respectively. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016. a

The rates and types of side effects and AEs, irrespective of relation to study medication, were not different between the 2 groups (Table 2). The most common side effects reported by subjects who received pravastatin were

musculoskeletal pain and heartburn. There were no reports of myopathy/ rhabdomyolysis or liver injury (data on maternal concentrations of creatine kinase, AST, and ALT are in Supplemental Table 1). None of the

Original Research

participants discontinued their study medication. In addition, there were no maternal, fetal, or infant deaths in either group. One fetus in the pravastatin group had hypospadias and another had coarctation of the aorta (diagnosed postnatally), whereas in the placebo group, one fetus had polydactyly and another had ventriculomegaly. One subject in the placebo group underwent postpartum hysterectomy secondary to hemorrhage from placenta previa and uterine atony. Pravastatin Cmax, Tmax, AUC, T1/2, percentage of the dose excreted unchanged as parent drug, and CL/F were not significantly different between the 3 time periods (Table 3). The average steady-state concentration-time profiles are depicted graphically in Supplemental Figure 3. For subjects in whom we were able to quantify the 24 hour postdose concentration, pravastatin appears to exhibit a 2-compartment PK model. The apparent half-life of pravastatin based on concentration data until 12 hours was estimated to be 2.1  0.9 hours in the second trimester (n ¼ 11), 3.0  1.6 hours in the third trimester (n ¼ 10), and 2.4  1.3 hours postpartum (n ¼ 9). However, in the small subset of subjects (n ¼ 1e3 per PK study day) in whom we were able to quantify the 24 hour postdose concentration, the estimated terminal half-life was much longer. Renal clearance and net renal secretion clearance of pravastatin were significantly higher during pregnancy compared with postpartum (Table 3). We had an adequate sample to assay for pravastatin concentrations in 6 umbilical cord arterial and 7 umbilical cord venous samples. In the majority of umbilical cord and maternal samples at the time of delivery, pravastatin concentrations were below the limit of quantification of the assay (Supplemental Table 2). Four subjects in the placebo group developed preeclampsia (with 3 of 4 having severe disease) compared with none in the pravastatin group. There were 5 indicated preterm deliveries before 37 weeks in the placebo group compared with 1 in the pravastatin group. (Table 4) Other obstetric

JUNE 2016 American Journal of Obstetrics & Gynecology

720.e4

Original Research

ajog.org

OBSTETRICS

outcomes were similar between the 2 groups. The concentrations of PlGF were increased in subjects receiving pravastatin, and those of sFlt-1 and sEng were decreased; however, the differences for these markers did not reach statistical significance. Of note, the 4 women in the placebo arm who developed preeclampsia had the highest sFlt-1 concentrations near term (Figure and Supplemental Table 1). Birthweight was similar between the 2 groups. One infant in the placebo group was diagnosed as small for gestational age. Five infants born to women in the placebo group were admitted either to an intermediate nursery (n ¼ 2) or the NICU (n ¼ 3) compared with 2 in the pravastatin group (intermediate nursery [n ¼ 1] and the NICU [n ¼ 1]; Table 4). None of the newborns in either group failed their auditory brainstem responseeevoked potential or similar hearing screening tests. Of note, 8 of 10 women in both groups breast-fed or provided breast milk to their newborns. Maternal total cholesterol (TC) and low-density lipoproteins (LDL) were similar at baseline but lower in subjects receiving pravastatin compared with placebo in the second trimester (TC, 188.6  31 vs 230  48.3 mg/dL, and LDL, 81.1  20.9 vs 124.1  42 mg/dL) and in the third trimester (TC, 201.7  33.5 vs 250  25.3 mg/dL, P ¼ .02, and LDL, 85.6  25.7 vs 126.1  44.4 mg/dL; Supplemental Table 1). Despite the decrease in maternal TC and LDL, cord blood concentrations of TC and LDL were similar between the pravastatinand placebo-exposed fetuses (TC, 56.2  11.5 vs 63.9  18.8 mg/dL, and LDL, 28.2  10.2 vs 31.8  13.3 mg/dL). There were no differences in other cord blood parameters assayed (Supplemental Table 3).

Comment This pilot randomized controlled trial provides preliminary safety and PK data regarding the use of pravastatin, a drug traditionally avoided in pregnancy, for preventing preeclampsia, a pregnancy complication with serious morbidity. Initiation and completion of this trial was a direct result of collaboration between

TABLE 3

Estimated steady-state pravastatin pharmacokinetics in subjects during the second and third trimesters of pregnancy compared with postpartum

Parameter

30e34 wks gestation (n ¼ 10)

18e24 wks gestation (n ¼ 11)

4e6 mo postpartum (n ¼ 9)

14.9  11.3

11.1  6.2

17.2  11.5

Tmax, h

1.6  0.6

1.5  0.4

1.6  1.0

Half-lifeapparent, h

2.1  0.9

3.0  1.6

2.4  1.3

Cmax, ng/mL

CL/F, L/h CL/F, L/h/kg AUC(0-24), ng/h/mL Amount excreted(0-24 h), mg

396  190

4.2  2.0

31  16

32  16

0.98  0.60

Percent excreted unchanged

10  6

CLrenal, L/h

34  16

CLsecretion, mL/min

389  215

4.6  2.4

1.04  0.57 10  6

a

480  273a

289  142 3.2  1.5 43  20 0.93  0.60 9 6

34  11

23  4

471  151a

325  65

a

Data are reported as mean  SD. AUC(0-24), area under the concentration time curve; CL/F, apparent oral clearance; CLrenal, renal clearance; CLsecretion, net renal secretion clearance; Cmax, maximum concentration; Tmax, time to maximum concentration. a P < .05, second and third trimester compared with postpartum. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016.

the NICHDeObstetric-Fetal Pharmacology Research Units network and the FDA. Although the data are preliminary, no identifiable safety risks were associated with the use of pravastatin at a dose of 10 mg in this cohort of high-risk pregnant women, with strong signals for possible efficacy (lower rates of preeclampsia and indicated preterm delivery and a maternal proangiogenic profile). The lack of a reduction in cholesterol concentration in the fetuses exposed to pravastatin is reassuring. This favorable risk-benefit analysis justifies continued research using pravastatin in a larger clinical trial with dose escalation. Pravastatin classification as a category X medication was due to a lack of indications that warranted its use during pregnancy rather than for observed risk. An increased risk of congenital malformations has not been demonstrated in multiple cohorts of subjects exposed to pravastatin during pregnancy.21-26 In this trial, pravastatin was started in the second trimester (ie, after completion of organ formation), and the rate of anomalies was similar between subjects receiving pravastatin or placebo. Additionally, despite maternal concentrations

720.e5 American Journal of Obstetrics & Gynecology JUNE 2016

of TC and LDL being reduced in the second and third trimesters with pravastatin use, neither cord blood TC and LDL concentrations nor infant birthweight differed between the 2 groups. These findings support prior studies that showed independence of fetal cholesterol concentrations from maternal cholesterol levels or diet.27,28 The pattern and rates of AEs and serious AEs in our study are consistent with data from prior large pravastatin trials in nonpregnant women and men,29 suggesting that pregnancy does not adversely affect the occurrence of these AEs. Although not statistically significant, a 10 mg dose of pravastatin was associated with favorable pregnancy outcomes including lower rates of preeclampsia, indicated preterm delivery, and neonatal admissions to intermediate nurseries or the NICU as well as improved proangiogenic profile (lower sFlt-1, sEng, and higher PlGF). The high rate of preeclampsia recurrence in the placebo group is consistent with prior studies.4,5 The exact mechanism of how pravastatin may prevent preeclampsia is unknown but is thought to be associated with pravastatin’s ability

ajog.org

OBSTETRICS

Original Research

TABLE 4

FIGURE

Maternal and neonatal outcomes of participants in the study

Longitudinal plots of serum concentrations of sFlt-1, sEng, and PlGF within subjects

Placebo groupa (n ¼ 10)

Outcomes

Pravastatin groupa (n ¼ 10)

Maternal outcomes Preeclampsia

4 (40)

0 (0)

Severe features

3

0 b

Postpartum preeclampsia

1 (10)

0 (0)

Gestational hypertension

1 (10)

1 (10)

Gestational age at delivery, wks

36.7  2.1

37.7  0.9

Indicated preterm delivery less than 37 wks

5 (50)c

1 (10)d

Indicated preterm delivery less than 34 wks

1 (10)

0 (0)

Blood transfusion

1 (10)

1 (10)

4 [3e7]; range, 2e43

3 [3e4]; range, 1e6

Length of hospital stay, d

e

Neonatal outcomes 2877  630

3018  260

Well-baby/routine

5 (50)

8 (80)

Intermediate (level 2)

2 (20)

1 (10)

NICU

3 (30)

1 (10)

NICU length of stay  48 h

3 (30)

0

Respiratory distress syndrome

2 (20)

1 (10)

Birthweight, g Highest level of care

Data are reported as n (percentage), mean  SD, or median [interquartile range]. NICU, neonatal intensive care unit. None of the comparisons between the 2 groups is statistically significant (P > .05); b This subject developed preeclampsia and was delivered at 353/7 weeks because of spontaneous preterm labor and a history of prior classical cesarean delivery. She received magnesium sulfate and on discharge had normal blood pressure. She then presented 7 days after delivery with elevated blood pressure and was diagnosed with postpartum preeclampsia; c Three patients were delivered at 336/7, 343/7, and 35 2/7 for preeclampsia with severe features, 1 patient was delivered at 361/7 for worsening gestational hypertension and history of classical cesarean delivery, and 1 patient was delivered at 354/7 for placenta previa; d One patient was delivered at 355/7 weeks for worsening chronic hypertension; e Length of hospital stay was for the hospitalization resulting in delivery. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016. a

to reverse the pregnancy-specific angiogenic imbalance and oxidative and inflammatory stress and to restore global endothelial health.19 The ability of pravastatin to restore the angiogenic balance is also being tested in a proof of concept Statins to Ameliorate early onset Preeclampsia trial in the United Kingdom (www.controlled-trials.com; number ISRCTN23410175). Pravastatin’s reassuring data with its use in human pregnancy and in animal models were not unexpected. Pravastatin is one of the most hydrophilic statins and is a substrate for the efflux transporters P-glycoprotein and multidrug resistance-associated protein 2, which

would potentially limit its transplacental transfer.30-32 Consistent with these characteristics, recent transplacental studies have shown that the clearance of pravastatin in third-trimester placentas was higher in the fetal-to-maternal than the maternal-to-fetal direction.32 In this study, the majority of the umbilical cord and maternal pravastatin concentrations at delivery were below the lower limit of quantification of the assay (< 0.1 ng/mL). Only 2 subjects had measurable concentrations in the umbilical cord plasma (Supplemental Table 2), which may also be related to the low dose of pravastatin used in this study and its short apparent half-life.

Longitudinal plots of serum concentrations of sFlt-1 (panel A), sEng (panel B), and PlGF (panel C) within individual subjects who received pravastatin (n ¼ 10, red ) or placebo (n ¼ 10, blue ) according to the gestational age window at time of collection: 120/7 to 166/7 weeks (baseline and before treatment), 24 0/7 to 276/7, and 340/7 to 366/7 weeks. D designates the subjects who developed preeclampsia. PlGF, placental growth factor; sEng soluble endoglin; sFlt-1, soluble fms-like tyrosine kinase. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016.

Postpartum values for pravastatin CL/F, renal clearance, AUC, half-lifeinitial, Cmax, Tmax, and fraction of dose excreted unchanged in the urine are consistent with those previously reported in nonpregnant subjects.33,34 Although significance was not reached because of the small sample size, there is a strong trend toward an increase in CL/F and a decrease in pravastatin AUC in

JUNE 2016 American Journal of Obstetrics & Gynecology

720.e6

Original Research

OBSTETRICS

pregnancy compared with postpartum. As expected, the renal clearance of pravastatin was significantly increased during pregnancy, consistent with the increased glomerular filtration as measured by the creatinine clearance, accounting for some of the variability. In addition, the net renal secretion clearance was also increased in pregnancy, likely because of changes in active transport by renal excretory transporters such as P-glycoprotein, multidrug resistance-associated protein 2, breast cancer resistance protein, or renal uptake transporters such as organic anion transporter (OAT)-3 and OAT1.31,32,35 The PK parameters of 30 a-isopravastatin, a major metabolite of pravastatin with only 1-10th to 1-40th of its HMG-CoA reductase inhibition,30 are reported in Supplemental Table 4. Because to our knowledge this is the first preeclampsia prevention trial using statins, it was designed to have multiple layers of safety including being conducted under an IND, use of an independent medical monitor who reviewed all serious AEs in real time, oversight by a data safety monitoring board that reviewed all AEs and serious AEs quarterly, and use of well-defined criteria to withdraw subjects or stop the study. Maternal, fetal, and neonatal safety was determined using clinical and laboratory outcomes at multiple time points. An independent panel, blinded to study drug allocation, reviewed the preeclampsia outcomes. At the recommendation of the FDA, we were limited by our sample size, which prevented us from conducting subgroup and other more detailed analyses. In addition, our study does not have the power to detect differences in individual outcomes such as congenital anomalies or other clinical or safety outcomes of low prevalence. Other limitations include the use of a low dose of pravastatin (10 mg/d) and the short-term follow-up for subjects and infants included in this report. Infant follow-up is planned at 5 years of age, and subjects’ contact information is updated regularly to facilitate long-term follow-up.

Conclusion This study provides preliminary safety and pharmacokinetic data regarding the use of pravastatin for preventing preeclampsia in high-risk pregnant women. Although the data are preliminary, no identifiable safety risks were associated with pravastatin use in this cohort. This favorable risk-benefit analysis justifies continued research with dose escalation in a future larger clinical trial to evaluate pravastatin’s effectiveness in preventing preeclampsia. Finally, collaboration between the NICHD and the FDA was essential for the success of this trial. n Acknowledgments We thank Ms Charlene Williamson and members of the FDA Division of Reproductive and Urologic Products who reviewed our Investigational New Drug application for their thoughtful study design comments. In addition, we thank the following members who participated in protocol development, oversight, data management, and coordination between the clinical research centers: Katrina Burson, RN, and Julie Croxford, RN, MPH. They also thank the following collaborators and study research personnel: University of Texas Medical Branch, Galveston, TX, George Saade, MD; Shannon Clark MD; Wayne Snodgrass, MD; Xing Zhang, PhD; Svetlana Patrikeeva, MS; Tatiana Nanovskaya, PhD; Angela Jones, RN; Sonia Jordan, RN; Carly Oliver, RN; Margaret Zimmerle, RN; and Maria Garza; Columbia University, New York, NY, Mary Talucci, MSN, CCRP; Indiana University, Indianapolis, IN, David Flockhart, MD; and Janie Klank, RN, MSN; University of Washington, Seattle, WA, Michael Z. Liao, BS; Yvonne Lin, PhD; Kenneth Thummel, PhD; Karan Hays, DNP, CNM, ARNP; Erin Michelson; Claudine Hernandez; and Anna Lemchen, RN; University of Pittsburgh, Pittsburgh, PA, Dawn Fischer, RN; and Donna DeAngeles, BS; and RTI International, Research Triangle Park, NC, Lei Li, PhD; Matthew Westlake, MS; and Kelly Rooney, PhD.

References 1. American College of Obstetricians and Gynecologists. Hypertension in pregnancy. Report of the American College of Obstetricians and Gynecologists’ Task Force on Hypertension in Pregnancy. Obstet Gynecol 2013;122: 1122-31. 2. Baumwell S, Karumanchi SA. Preeclampsia: clinical manifestations and molecular mechanisms. Nephron Clin Pract 2007;106: c72-81. 3. Mongraw-Chaffin ML, Cirillo PM, Cohn BA. Preeclampsia and cardiovascular disease death: prospective evidence from the child

720.e7 American Journal of Obstetrics & Gynecology JUNE 2016

ajog.org health and development studies cohort. Hypertension 2010;56:166-71. 4. Barton JR, Sibai B. Prediction and prevention of recurrent preeclampsia. Obstet Gynecol 2008;112:359. 5. Hernández-Dıaz S, Toh S, Cnattingius S. Risk of pre-eclampsia in first and subsequent pregnancies: prospective cohort study. BMJ 2009;338:b2255. 6. Young BC, Levine RJ, Karumanchi SA. Pathogenesis of preeclampsia. Annu Rev Pathol 2010;5:173-92. 7. Ramma W, Ahmed A. Is inflammation the cause of preeclampsia? Biochem Soc Trans 2011;39:1619-27. 8. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med 2005;352:1685-95. 9. Askie LM, Duley L, Henderson-Smart DJ, Stewart LA; PARIS Collaborative Group. Antiplatelet agents for prevention of preeclampsia: a meta-analysis of individual patient data. Lancet 2007;369:1791-8. 10. Mills EJ, Rachlis B, Wu P, Devereaux PJ, Arora P, Perri D. Primary prevention of cardiovascular mortality and events with statin treatments: a network meta-analysis involving more than 65,000 patients. J Am Coll Cardiol 2008; 52:1769-81. 11. Brugts JJ, Yetgin T, Hoeks SE, et al. The benefits of statins in people without established cardiovascular disease but with cardiovascular risk factors: meta-analysis of randomised controlled trials. BMJ 2009;338:b2376. 12. Cudmore M, Ahmad S, Al-Ani B, et al. Negative regulation of soluble Flt-1 and soluble endoglin release by heme oxygenase-1. Circulation 2007;115:1789-97. 13. Costantine M, Tamayo E, Bytautiene E, et al. Using pravastatin to improve the vascular reactivity in a mouse model of soluble Fms-like tyrosine kinase-1-induced preeclampsia. Obstet Gynecol 2010;116:114-20. 14. Fox KA, Longo M, Tamayo E, et al. Effects of pravastatin on mediators of vascular function in a mouse model of soluble Fms-like tyrosine kinase-1einduced preeclampsia. Am J Obstet Gynecol 2011;205:366.e1-5. 15. Ahmed A, Singh J, Khan Y, Seshan SV, Girardi G. A new mouse model to explore therapies for preeclampsia. PLoS One 5. 2010;10: e13663. 16. Singh J, Ahmed A, Girardi G. Role of complement component C1q in the onset of preeclampsia in mice. Hypertension 2011;58: 716-24. 17. Kumasawa K, Ikawa M, Kidoya H, et al. Pravastatin induces placental growth factor and ameliorates preeclampsia in a mouse model. Proc Natl Acad Sci USA 2011;108: 1451-5. 18. Saad AF, Kechichian T, Yin H, et al. Effects of pravastatin on angiogenic and placental hypoxic imbalance in a mouse model of preeclampsia. Rep Sci 2014;21:138-45. 19. Costantine MM, Cleary K; for the Eunice Kennedy Shriver National Institute of Child

ajog.org Health and Human Development Obstetric-Fetal Pharmacology Research Units Network. Pravastatin for the prevention of preeclampsia in high risk pregnant women. Obstet Gynecol 2013;121:349-53. 20. American College of Obstetricians and Gynecologists. Diagnosis and management of preeclampsia and eclampsia. ACOG Practice bulletin. Obstet Gynecol 2002;99: 159-67. 21. Edison R, Muenke M. Mechanistic and epidemiologic considerations in the evaluation of adverse birth outcomes following gestational exposure to statins. Am J Med Genet 2004; 131A:287. 22. Ofori B, Rey E, Berard A. Risk of congenital anomalies in pregnant users of statin drugs. Br J Clin Pharmacol 2007;64:496. 23. Taguchi N, Rubin ET, Hosokawa A, et al. Prenatal exposure to HMG-CoA reductase inhibitor: effects on fetal and neonatal outcomes. Reprod Toxicol 2008;26:175. 24. Petersen EE, Mitchell AA, Carey JC, et al. Maternal exposure to statins and risk for birth defects. Am J Med Genet 2008;146A: 2701-5. 25. Winterfeld U, Allignol A, Panchaud A, et al. Pregnancy outcome following maternal exposure to statins: a multicentre prospective study. Br J Obstet Gynaecol 2013;120: 463-71. 26. Bateman BT, Hernandez-Diaz S, Fischer MA, et al. Statins and congenital malformations: Cohort study. BMJ 2015;350: h1035. 27. Woollett LA. Maternal cholesterol in fetal development: transport of cholesterol from the

OBSTETRICS

maternal to the fetal circulation. Am J Clin Nutr 2005;82:1155-61. 28. Ethier-Chiasson M, Duchesne A, Forest JC, et al. Influence of maternal lipid profile on placental protein expression of LDLr and SR-BI. Biochem Biophys Res Commun 2007;359: 8-14. 29. Pfeffer MA1, Keech A, Sacks FM, et al. Safety and tolerability of pravastatin in longterm clinical trials: prospective Pravastatin Pooling (PPP) Project. Circulation 2002;105: 2341-6. 30. Hatanaka T. Clinical pharmacokinetics of pravastatin: mechanisms of pharmacokinetic events. Clin Pharmacokinet 2000;39: 397-412. 31. Zarek J, DeGorter MK, Lubetsky A, et al. The transfer of pravastatin in the dually perfused human placenta. Placenta 2013;34: 719-21. 32. Nanovskaya TN, Patrikeeva SL, Paul J, Costantine M, Hankins GDV, Ahmed MS. Transplacental transfer and distribution of pravastatin. Am J Obstet Gynecol 2013;209:373. e1-5. 33. Singhvi SM, Pan HY, Morrison RA, Willard DA. Disposition of pravastatin sodium, a tissue-selective HMG-CoA reductase inhibitor, in healthy subjects. Br J Clin Pharmacol 1990;29:239-43. 34. Ogawa K, Hasegawa S, Udaka Y, Nara K, Iwai S, Oguchi K. Individual difference in the pharmacokinetics of a drug, pravastatin, in healthy subjects. J Clin Pharmacol 2003;43: 1268-73. 35. Hasegawa M, Kusuhara H, Sugiyama D, et al. Functional involvement of rat organic anion

Original Research

transporter 3 (rOAT3; Slc22a8) in the renal uptake of organic anions. J Pharmacol Exp Ther 2002;300:746-53.

Author and article information From the Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX (Drs Costantine, West, Ahmed, and Hankins); Columbia University, New York, NY (Drs Cleary and D’Alton); Departments of Pharmacy and Obstetrics and Gynecology, University of Washington, Seattle, WA (Drs Hebert and Easterling); University of Pittsburgh, Pittsburgh, PA (Drs Caritis and Venkataramanan); Departments of Obstetrics and Gynecology and Pediatrics, Indiana University, Indianapolis, IN (Drs Haas and Haneline); RTI International, Rockville, MD (Dr Brown); and the Eunice Kennedy Shriver National Institute of Child Health and Human Development (Dr Ren). Received Sept. 30, 2015; revised Dec. 9, 2015; accepted Dec. 17, 2015. The views expressed herein are those of the authors and do not necessarily represent the official views of Eunice Kennedy Shriver National Institute of Child Health and Human Development or the National Institutes of Health. This study was supported by grants U10HD047891, U10HD063094, U10HD047892, U10HD047905, and U10HD057753 from the Eunice Kennedy Shriver National Institute of Child Health and Human Development and grants UL1TR000423 and UL1TR001439 from National Institutes of Health and National Center for Advancing Translational Sciences through the Clinical and Translational Science Awards Program. The authors report no conflict of interest. Corresponding author: Maged Costantine MD. [email protected]

JUNE 2016 American Journal of Obstetrics & Gynecology

720.e8

Original Research

OBSTETRICS

Supplemental material Patients and methods Pharmacokinetic (PK) sample analysis

Concentrations of pravastatin and 3’a-isopravastatin in plasma and urine were measured by liquid chromatographyemass spectrometry using an in-house validated analytical method, following Food and Drug Administration guidance. (Food and Drug Administration, Guidance for industry bioanalytical method validation, 2013.1 Briefly, the method includes a 1-step liquid-liquid extraction for plasma and direct dilution for urine samples. The lower limit of quantitation (LLOQ) of the analytes in plasma samples was 0.1 ng/mL for pravastatin and 0.1 ng/mL for 3’a-isopravastatin. The LLOQ in urine samples was 19.7 ng/mL for pravastatin and 2.0 ng/mL for 3a-hydroxypravastatin. The relative deviation of this method was < 10% for intraday and interday assays in plasma and urine samples, and the recovery of the method ranged between 97% and 106% in plasma and between 98% and 105% in urine. PK analysis

Steady-state pravastatin PK parameters were estimated using standard noncompartmental techniques. Maximum concentration (Cmax) and time to maximum concentration (Tmax) were determined from the measured concentrations. Area under the concentration time curve (AUC0-24h) was estimated using the linear trapezoidal rule. When concentrations fell below the lower limit of quantification for the assay prior to the end of the dosing interval, the 24 hour concentration was estimated by extrapolating the predicted concentration based on the regression line from the time of the last measurable concentration (Clast) using the equation, 24 hour concentration ¼ Clast $ e-kt, where k was the elimination rate constant and t was the time between Clast and 24 hours.

The elimination rate constant was determined by log-linear regression of the terminal slope of the plasma concentration vs time data. Apparent oral clearance (CL/F) was estimated by the formula, CL/F ¼ dose/AUC0-24h. Halflife was determined by half-life ¼ ln(2)/k. The apparent half-life was determined using plasma concentrations up to 12 hours. In a subset of subjects who had measurable concentrations up to 24 hours, the terminal elimination half-life was determined utilizing plasma concentrations through the 24 hour time point. The metabolic ratio was estimated by metabolic ratio ¼ 3’a-isopravastatin AUC0-24h/pravastatin AUC0-24h corrected for molecular weight. The amount of pravastatin and 3’a-isopravastatin excreted into the urine was calculated by summing the amount excreted over 1 dosing interval. The percent of pravastatin dose excreted unchanged in the urine was determine by the (amount excreted over 1 dosing interval divided by the dose)  100. Renal clearance was estimated by the amount of pravastatin excreted unchanged in the urine over 1 dosing interval/AUC0-24h. Net renal secretion clearance was estimated by CLsecretion ¼ CLrenal e fu$CrCl in which fu is the fraction unbound and CrCl is the creatinine clearance. The fraction unbound for pravastatin was assumed to be 0.55.2,3 PK parameters are reported as mean  SD and were adjusted using actual body weights.

Preeclampsia diagnosis Preeclampsia was defined as the presence of either a systolic blood pressure of  140 mm Hg and/or diastolic blood pressure of  90 mm Hg on 2 occasions at least 4 hours (but less than 7 days) apart, with proteinuria (either  1þ on urine dipstick 4 hours apart or  300 mg in an adequately collected, timed urine sample) after the 20th week of gestation. Severe preeclampsia was defined as preeclampsia with any one of the following: either systolic blood pressure of  160 mm Hg or diastolic blood

720.e9 American Journal of Obstetrics & Gynecology JUNE 2016

ajog.org pressure of  110 mm Hg on at least 2 occasions at least 4 hours apart, persistent cerebral disturbances, visual disturbances (headache, blurring of vision, scotomas), or right upper quadrant or epigastric pain, thrombocytopenia (platelet count < 100,000/mm3), elevated liver enzymes (aminotransferase [AST] or aspartate aminotransferase [ALT]) > 2 times the upper limit of normal, eclamptic seizure, HELLP (hemolysis, elevated liver enzymes, and low platelet count) syndrome, pulmonary edema, acute kidney injury, or intrauterine growth restriction. Chronic hypertension with superimposed preeclampsia was diagnosed in patients with hypertension if 1 or more of the following were present: persistent cerebral disturbances, visual disturbances (headache, blurring of vision, scotomas), or right upper quadrant or epigastric pain, thrombocytopenia (platelet count < 100,000/mm3), elevated liver enzymes (ASTor ALT) > 2 times upper normal, eclamptic seizure, or HELLP syndrome. At the time of the protocol development, the previously mentioned definition of preeclampsia was based on the American College of Obstetricians and Gynecologists (ACOG) practice bulletin.4 In 2013 ACOG revised the definition of preeclampsia.5 In this study, the rates and severity of preeclampsia were not different in each group, whether the previously or newly published ACOG criteria were used.

Biomarkers measured with an enzyme-linked immunosorbent assay All samples available from the different study sites were shipped on dry ice for batch analysis in a single laboratory at the University of Texas Medical Branch in Galveston, TX. Personnel performing the laboratory analysis were blinded to treatment assignment. Maternal and umbilical cord serum concentration of sFlt-1, sEng, and PlGF as well as umbilical cord serum neuron specific enolase (NSE) and S100B were measured with commercially available

ajog.org enzyme-linked immunosorbent assay kits and according to the manufacturer’s instructions. Immunoassay kits were purchased from R&D Systems (Minneapolis, MN) for sFlt-1, sEng, and PlGF; Alpha Diagnostic International (San Antonio, TX) for NSE; and BioVendor LLC (Chandler, NC) for S100B. The detection limits for the 5 assays were 13.3 pg/mL, 7 pg/mL, 0.03 ng/mL, 1 mg/L, and 5 pg/mL, respectively. The inter- and intraassay coefficients of variation were < 10% for all analytes. All samples were run in duplicate and the mean values used in analyses.

Serious adverse event definition A serious adverse event was defined as any adverse event occurring that results in any of the following outcomes: maternal,

OBSTETRICS

fetal, or neonatal death; maternal, fetal, or neonatal life-threatening adverse experience; maternal, fetal, or neonatal inpatient hospitalization or prolongation of existing hospitalization; a persistent or significant disability/ incapacity; or a congenital anomaly/ birth defect. Other adverse events were considered serious if they jeopardized the subject or required intervention to prevent one of the previous outcomes listed; examples of those include rhabdomyolysis and liver injury. A life-threatening adverse event is defined as any adverse event that places the patient at immediate risk of death from the reaction as it occurred. It does not include a reaction that, had it occurred in a more severe form, might have caused death.

Original Research

References 1. Zhang X, Vernikovskaya DI, Wang X, et al. Quantitative determination of pravastatin and its metabolite 3a-hydroxy pravastatin in plasma and urine of pregnant patients by LC-MS/MS. Biomed Chromatogr 2016;30:548-54. 2. Pan HY. Clinical pharmacology of pravastatin, a selective inhibitor of HMG-CoA reductase. Eur J Clin Pharmacol 1991;40(Suppl 1):S15-8. 3. Singhvi SM, Pan HY, Morrison RA, et al. Disposition of pravastatin sodium, a tissueselective HMG-CoA reductase inhibitor, in healthy subjects. Br J Clin Pharmacol 1990;29: 239-43. 4. American College of Obstetricians and Gynecologists. Diagnosis and management of preeclampsia and eclampsia. ACOG Practice bulletin no. 33. Obstet Gynecol 2002;99: 159-67. 5. American College of Obstetricians and Gynecologists. Hypertension in pregnancy. Report of the American College of Obstetricians and Gynecologists’ Task Force on Hypertension in Pregnancy. Obstet Gynecol 2013;122:1122-31.

JUNE 2016 American Journal of Obstetrics & Gynecology

720.e10

Original Research

OBSTETRICS

SUPPLEMENTAL FIGURE 1

Study flow chart

Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016.

720.e11 American Journal of Obstetrics & Gynecology JUNE 2016

ajog.org

ajog.org

OBSTETRICS

Original Research

SUPPLEMENTAL FIGURE 2

GA at delivery in current pregnancy compared with prior qualifying pregnancy

Red lines represent patients assigned to pravastatin, and blue lines represent patients assigned to placebo. GA, gestational age. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016.

JUNE 2016 American Journal of Obstetrics & Gynecology

720.e12

Original Research

OBSTETRICS

ajog.org

SUPPLEMENTAL FIGURE 3

Pravastatin concentration vs time profiles in women second and third trimesters and postpartum

Mean (SD) steady-state plasma pravastatin concentration vs time profiles in women studied during the second (A) and third trimesters (B) of pregnancy, and postpartum (C). Pravastatin plasma concentrations at 24 hours were extrapolated from the predicted concentration at the last measurable time point. Pravastatin plasma concentrations at predose (0 hour) were observed concentrations; when they were below the limit of quantification, the predicted 24 hour concentrations were used. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016.

720.e13 American Journal of Obstetrics & Gynecology JUNE 2016

ajog.org

SUPPLEMENTAL TABLE 1

Maternal laboratory assays Placebo group

Pravastatin group

Variable

Rand Mean (n, SD)

BS1 Mean (n, SD)

AST, U/L

16.1 (7, 4.2)

18 (9, 5.9)

17.5 (6, 3.8)

18.9 (8, 6.4)

15.5 (10, 4.2)

20.5 (10, 7.3)

ALT, U/L

16.7 (7, 7.2)

19.6 (9, 10.8)

16 (6, 5.3)

19.9 (8, 7.1)

16.9 (10, 5.7)

21.6 (10, 8.7)

CK, U/L

72.5 (10, 38.4)

75.4 (9, 44)

74.3 (7, 58.7)

79.8 (11, 79.3)

43.3 (10, 23.5)

68.7 (10, 53.8)

LDL, mg/dL

87.4 (10, 25.8)

124.1 (7, 42)

126.1 (7, 44.4)

87.6 (11, 25.6)

81.1 (10, 20.9)

85.6 (10, 25.7)

HDL, mg/dL

67.2 (10, 19)

74.6 (7, 23.7)

59.1 (11, 10.8)

72.6 (10, 15.1)

73.5 (10, 10.2)

TG, mg/dL

137 (10, 41.6)

195 (9, 77.8)

246.4 (7, 85.2)

150.6 (11, 60.8)

174.9 (10, 48.7)

213.2 (10, 45.4)

TC, mg/dL

185.6 (9, 32.4)

230 (9, 48.3)

250 (7, 25.3)a

176.6 (11, 34.3)

188.6 (10, 31)

201.7 (10, 33.5)a

80.6 (9, 26.2)

BS2 Mean (n, SD)

Rand Mean (n, SD)

BS1 Mean (n, SD)

BS2 Mean (n, SD)

76.4 (10, 34.3)

270.8 (10, 194.7)

249.4 (10, 187.5)

71.3 (10, 34.8)

410.7 (10, 292.3)

453.6 (10, 459.2)

sFlt1, pg/mL

803.8 (10, 238.7)

1249.9 (10, 1154.9)

7504.8 (10, 6817.2)

785.9 (10, 240.4)

658.1 (10, 195.6)

3430.4 (10, 2102.1)

6.1 (10, 2.1)

21.9 (10, 23.7)

6.1 (10, 2.4)

5.6 (10, 2.3)

12.3 (10, 9.6)

sEng, ng/mL

5.6 (10, 1.3)

Rand indicates randomization visit (120/7 to 166/7 weeks) and before start of study medication; BS1 indicates biospecimen collection visit 1, between 240/7 and 276/7 weeks; BS2 indicates biospecimen collection visit 2, between 340/7 and 366/7 weeks. ALT, alanine aminotransferase; AST, aspartate aminotransferase; CK, creatine kinase; HDL, high density lipoproteins; LDL, low density lipoproteins; PlGF, placental growth factor; sEng, soluble endoglin; sFl-1, soluble FMS-like tyrosine kinase 1; TC, total cholesterol; TG, triglycerides. P ¼ .02 for comparison between the 2 groups. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016.

a

OBSTETRICS

720.e14

Original Research

JUNE 2016 American Journal of Obstetrics & Gynecology

PLGF, pg/mL

Original Research

ajog.org

OBSTETRICS

SUPPLEMENTAL TABLE 2

Maternal and cord blood plasma drug and metabolite concentrations at time of delivery in patients randomized to the pravastatin group 3’a-Isopravastatin, ng/mL

Pravastatin, ng/mL Subject UA

UV

Maternal UA

UV

Maternal

1

< LLOQ

< LLOQ

< LLOQ

< LLOQ

< LLOQ

< LLOQ

2

< LLOQ

< LLOQ

< LLOQ

< LLOQ

< LLOQ

< LLOQ

3

0.29

< LLOQ

0.13

< LLOQ

< LLOQ

< LLOQ

4

< LLOQ

< LLOQ

< LLOQ

< LLOQ

< LLOQ

< LLOQ

5

NA

VNE

< LLOQ

NA

VNE

< LLOQ

6

0.34

0.11

0.43

0.72

< LLOQ

0.15

7

VNE



VNE

0.21

< LLOQ

8

Not collected Not collected 0.54

9

Not collected VNE

10

NA

< LLOQ

Not collected Not collected < LLOQ

< LLOQ

Not collected VNE

< LLOQ

NA

< LLOQ

< LLOQ < LLOQ

< LLOQ indicates that the plasma concentration is below the lower limit of quantification. LLOQ, lower limit of quantitation; NA, concentration not reported; UA, umbilical artery concentration; UV, umbilical vein concentration; maternal, maternal plasma concentration within 30 minutes of delivery; VNE, volume not enough. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016.

720.e15 American Journal of Obstetrics & Gynecology JUNE 2016

ajog.org

OBSTETRICS

Original Research

SUPPLEMENTAL TABLE 3

Cord blood laboratory data Placebo groupa Mean, n (SD)

Variable S100b, pg/mLb b

NSE, ng/mL

sFlt-1, pg/mL

179.8 (10, 54.9)

Pravastatin groupa Mean, n (SD) 155.7 (7, 43.7)

12.4 (10, 8.8)

12.3 (7, 8.2)

281.5 (10, 188.4)

169.5 (8, 65.9)

sEng, ng/mL

4 (10, 1)

4.3 (8, 1.6)

PLGF, pg/mL

8.5 (10, 1.8)

9.1 (8, 2.5)

LDL, mg/dL

31.8 (9, 13.3)

28.2 (5, 10.2)

HDL, mg/dL

27.9 (10, 9.5)

24.5 (6, 5.6)

TG, mg/dL

23.8 (10, 9.2)

21.3 (6, 5.8)

TC, mg/dL

63.9 (10, 18.8)

56.2 (6, 11.6)

AST, U/L

37.2 (10, 23.1)

32.5 (6, 14.9)

ALT, U/L

17.8 (10, 8.9)

21 (6, 8.2)

CK, U/L Estradiol (only females), pg/mL

218.8 (10, 93.8) 5143 (1, NA)

234.3 (6, 140) 6117.7 (3, 1354.7)

TSH, mIU/mL

9.5 (8, 5.7)

8.1 (6, 2.9)

FSH, mIU/mL

0.2 (5, 0.05)

0.4 (4, 0.6)

LH, mIU/mL

0.2 (5, 0.08)

0.5 (4, 0.7)

Total testosterone (only males), ng/dL

153.4 (5, 62.7)

102.4 (2, 22.1)

Data are reported as mean (n, SD). S100B is a calcium-binding protein, predominantly expressed and released by astrocytes in the central nervous system and has been used as a marker of neurological injury. Neuron-specific enolase is a glycolytic enzyme localized primarily within neurons and neuroendocrine cells, and its release has been used as another marker of neurological injury. ALT, alanine aminotransferase; AST, aspartate aminotransferase; CK, creatine kinase; FSH, follicular stimulating hormone; HDL, high-density lipoproteins; LDL, low-density lipoproteins; LH, luteinizing hormone; NSE, neuron-specific enolase; PlGF, placental growth factor; sEng, soluble endoglin; sFl-1, soluble FMS-like tyrosine kinase 1; TC, total cholesterol; TG, triglycerides; TSH, thyroid stimulating hormone. None of the comparisons between the two groups is statistically significant (P > 0.05); b One infant was excluded from both the NSE and S100b analysis because of the sample being hemolyzed. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016.

a

JUNE 2016 American Journal of Obstetrics & Gynecology

720.e16

Original Research

ajog.org

OBSTETRICS

SUPPLEMENTAL TABLE 4

Estimated steady-state 3’a-isopravastatin pharmacokinetics in women during the second and third trimesters of pregnancy compared with postpartum

Variable

18-24 wks gestation (n ¼ 11)

30-34 wks gestation (n ¼ 10)

4-6 mo postpartum (n ¼ 9)

Cmax, ng/mL

19.7  15.4

8.0  5.9

10.9  7.2

1.3  0.7

1.2  0.5

1.3  0.8

Tmax, h

1.3  0.8

Half-life, h

27  14

AUC(0-24h), ng/h/mL

2.7  1.6 19  11

2.0  1.2 21  12

1.0  0.7

0.6  0.3

0.7  0.5

Amount excreted(0-24 h), mg

0.2  0.1

0.2  0.1

0.2  0.1

Percentage excreted as 3 a-isopravastatin

2.1  1.4

1.5  1.4

1.7  1.0

Metabolic ratio 0

Data are reported as mean  SD. AUC(0-24 h), area under the concentration time curve; Cmax, maximum concentration; Tmax, time to maximum concentration. Costantine et al. Pravastatin for prevention of preeclampsia. Am J Obstet Gynecol 2016.

720.e17 American Journal of Obstetrics & Gynecology JUNE 2016

A4 Junio.pdf

database that was managed by the data. coordinating center, which was. responsible for data analysis. Pharmacokinetic studies. Steady-state pravastatin PK ...

889KB Sizes 1 Downloads 355 Views

Recommend Documents

A4.GTAV.Xero.Market.Paper.Model.by.Papermau.2014.pdf
A4.GTAV.Xero.Market.Paper.Model.by.Papermau.2014.pdf. A4.GTAV.Xero.Market.Paper.Model.by.Papermau.2014.pdf. Open. Extract. Open with. Sign In.

A4 Septiembre.pdf
A receiver operating characteristic curve. analysis revealed strong diagnostic performance of plasma decorin in the. prediction of preeclampsia status. Although ...

a4.pdf
∆ιάλειμμα. Whoops! There was a problem loading this page. Retrying... Whoops! There was a problem loading this page. Retrying... a4.pdf. a4.pdf. Open. Extract.

A4 brochure Mphil
www.iiitmk.ac.in. Ecological Informatics (EI) is a trans-disciplinary subject that integrates ecology, information technology, computational and social sciences in.

A4 Octubre.pdf
METHODS: Data were obtained from the U.S. Zika Preg- nancy Registry, an enhanced surveillance system of pregnant. women with laboratory evidence of ...

¢2£%a4 I
Sep 24, 1979 - anamid HDS-9 Trilobe). The hydrotreating was con ducted at a reactor temperature of 650° F., hydrogen pressure of 900 psig, a liquid hourly ...

A4.Desktop.Architecture.Four.Simple.Little.Houses.by.Papermau.2015 ...
Page 1 of 4. Page 1 of 4. Page 2 of 4. Page 2 of 4. Page 3 of 4. Page 3 of 4. A4.Desktop.Architecture.Four.Simple.Little.Houses.by.Papermau.2015.pdf. A4.

A4-Eschenfelder.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item.

A4 brochure Mphil
All students admitted to the program, with minimum 65% marks or 7.0 CGPA in M.Sc. will be awarded SPEED-IT. First class M.Sc. in Natural/ Physical Sciences ...

Exhibitor Directory - A4 - Austrade
Jul 28, 2014 - Fax: +61 3 6231 0604 ... 3. The rise of Chinese civilisation over the past millennia is said to be ..... Room 1215, A building, Galaxy Central.

A4-Eschenfelder.pdf
There was a problem loading more pages. Retrying... Whoops! There was a problem previewing this document. Retrying... Download. Connect more apps.

a4.pdf
Whoops! There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. a4.pdf.

A4-Eschenfelder.pdf
Page 1 of 17. 1/17. Social Science Data Archives: A Historical Social Network Analysis1. Kristin R. Eschenfelder, Morgaine Gilchrist Scott, Kalpana Shankar, ...

A4.R2D2.Paper.Model.by.Disney.Family.Custom.by.Papermau.pdf
Page 3 of 3. Page 3 of 3. A4.R2D2.Paper.Model.by.Disney.Family.Custom.by.Papermau.pdf. A4.R2D2.Paper.Model.by.Disney.Family.Custom.by.Papermau.pdf. Open. Extract. Open with. Sign In. Main menu. Displaying A4.R2D2.Paper.Model.by.Disney.Family.Custom.b

Maundy Thursday Reflection A4
“One of you will betray me.” 4 ... He will be betrayed by a friend, one who sat with him as he dined that night had already put plans in .... love will triumph over all.

A4 Septiembre.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. A4 Septiembre.

A4.The.Dirty.Dog.Saloon.Vignette.Paper.Model.by.Papermau.2015.pdf
A4.The.Dirty.Dog.Saloon.Vignette.Paper.Model.by.Papermau.2015.pdf. A4.The.Dirty.Dog.Saloon.Vignette.Paper.Model.by.Papermau.2015.pdf. Open. Extract.

A4 Julio.pdf
There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. A4 Julio.pdf.

A4 Agosto.pdf
data were provided by the laboratory for. the purpose of this study because this. information is not routinely included in. the laboratory reports. Statistical analysis.

Maundy Thursday Reflection A4
He had told them that on the night before their liberation they should eat a hasty meal. • With sandals on their feet, they were to eat bread made in haste without ...

dvds-A4.pdf
Sign in. Loading… Page 1. Whoops! There was a problem loading more pages. Retrying... dvds-A4.pdf. dvds-A4.pdf. Open. Extract. Open with. Sign In.

CORE-pdf-A4-P.cdr - GitHub
Page 1. EL - TEC. - *

2017_LIF_Brochure A4.pdf
Lennon © Thomas House/Jinx Lennon. 7. Song of Love © Angelica ... Burke: No 10. The market © Liverpool ..... 2017_LIF_Brochure A4.pdf. 2017_LIF_Brochure ...

A4.Ammu.Natin.Gun.Shop.Paper.Model.V2.by.Papermau.2015.pdf
Whoops! There was a problem loading more pages. Whoops! There was a problem previewing this document. Retrying... Download. Connect more apps... Try one of the apps below to open or edit this item. A4.Ammu.Natin.Gun.Shop.Paper.Model.V2.by.Papermau.20