Skip to main content
Erschienen in: BMC Pregnancy and Childbirth 1/2014

Open Access 01.12.2014 | Research article

Prediction of preeclampsia and induced delivery at <34 weeks gestation by sFLT-1 and PlGF in patients with abnormal midtrimester uterine Doppler velocimetry: a prospective cohort analysis

verfasst von: Johannes Stubert, Stefanie Ullmann, Michael Bolz, Thomas Külz, Max Dieterich, Dagmar-Ulrike Richter, Toralf Reimer

Erschienen in: BMC Pregnancy and Childbirth | Ausgabe 1/2014

Abstract

Background

Women with bilateral abnormal uterine artery Doppler velocimetry (UtADV) are at increased risk for an adverse pregnancy outcome. This study aimed to determine if additional assessment of midtrimester angiogenic factors improves the predictive accuracy of Doppler results for various outcome parameters.

Methods

Women with a bilateral abnormal UtADV, which was defined as a postsystolic incision and/or an increased pulsatility index greater than the 95th centile, and a singleton pregnancy were prospectively recruited between 19 + 0 and 26 + 6 weeks of gestation. Maternal serum levels of placental growth factor (PlGF) and soluble fms-like tyrosine kinase-1 (sFLT-1) were measured with a fully automated immunoassay and their ratio was calculated.

Results

Angiogenic factors could predict the development of preeclampsia (PE), as well as induced delivery at <34 weeks of gestation, but failed to predict the development of normotensive intrauterine growth restriction. Twelve (24.0%) of the 50 recruited women developed PE. Nine of these patients had early-onset disease (<34 + 0 weeks). Six (12.0%) patients were delivered at <34 + 0 weeks. The most useful test results in the prediction of PE and induced delivery at <34 + 0 weeks were observed using the sFLT-1/PlGF >95th centile ratio with a sensitivity, specificity, positive predictive value, and negative predictive value (NPV) of 66.7%, 89.5%, 66.7%, and 89.5% for PE, and 85.7%, 86.1%, 50.1%, and 97.4% for induced delivery, respectively. Positive and negative likelihood ratios were 6.33 (95% CI 2.31–17.38) and 0.37 (95% CI 0.17–0.84) for PE, and 6.14 (95% CI 2.76–13.69) and 0.17 (0.03–1.02) for induced delivery, respectively. Corresponding odds ratios were 17.0 (95% CI 3.5–83.0) and 37.0 (95% CI 3.8–363.9), respectively.

Conclusions

Measurement of angiogenic factors improves the specificity of an abnormal UtADV for prediction of PE. Compared with prediction of PE an abnormal sFLT-1/PlGF ratio revealed higher sensitivity for prediction of induced delivery at <34 + 0 weeks. The NPV of 97% will help to reassure most patients with an abnormal UtADV and a normal sFLT-1/PlGF ratio.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2393-14-292) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

JS conceived and designed the study, analysed and interpreted the data, and drafted the manuscript. SU acquired and analysed the patients’ data, and drafted the manuscript. MB acquired patients, performed the ultrasound examination and took blood samples. TK acquired patients, performed the ultrasound examination and took blood samples. MD analysed and interpreted the data, and wrote the manuscript. DUR designed the study and acquired the lab data. TR conceived and designed the study, and wrote the manuscript. All authors have seen and approved the final version of the manuscript.

Background

The estimated incidence of preeclampsia (PE) is between 0.4–2.8% of all pregnancies in Europe [1, 2]. Only 15–20% of PE is present a severe clinical course [3]. The majority of severe PE develops early in pregnancy (<34 weeks of gestation) and is frequently associated with a serious maternal and foetal outcome [46].
Angiogenic factors have a large effect on development of preeclamptic syndrome [7]. The extent of the antiangiogenic shift, characterized by an increase in antiangiogenic soluble fms-like tyrosine kinase (sFlt)-1 and a decrease in angiogenic placental growth factor (PlGF), correlates with disease severity and precedes the clinical manifestation for several weeks [811]. Therefore, analysis of maternal sFLT-1 and PlGF can improve the diagnostic accuracy for detection of PE and allows an estimation of the clinical disease severity [12]. Moreover, removal of sFLT-1 from the maternal circulation by apheresis improves the severity of the disease and consequently prolongs the duration of pregnancy [13].
Although definitive treatment of PE is possible by delivery of the placenta alone, early prediction of high-risk patients may enhance the patient´s care before disease manifestation and could help to reduce mortality and morbidity of the mother and her foetus. A widely used noninvasive approach for identifying high-risk patients comprises the performance of midtrimester Doppler ultrasound measurements of the uterine arteries. An abnormal uterine artery Doppler velocimetry (UtADV) during the second trimester has shown detection rates for PE between 40% and 80% in patients at low risk [14]. Generally, sensitivity is better for the detection of severe and/or early-onset PE compared to mild and late onset cases, but positive predictive accuracy is quite low, and the majority of patients with an abnormal UtADV will not develop PE [15]. However, additional analysis of angiogenic markers may improve the test accuracy for detecting PE. Our prospective study aimed to evaluate sFLT-1 and PlGF as predictive markers for PE in a high-risk collective as identified by an abnormal UtADV in the second trimester.

Methods

Selection and inclusion criteria of patients

Patients with a bilateral abnormal UtADV between 19 + 0 and 26 + 6 weeks of gestation were included in this prospective cohort study. The clinical trial was conducted at the Department for Obstetrics and Gynaecology of the University of Rostock, Germany in cooperation with the outpatient centre of prenatal diagnosis “Praxiszentrum Frauenheilkunde” in Rostock between February 2011 and July 2013. The study was approved by the institutional review board of the University of Rostock (IRB No. A2010100) and written informed consent was obtained from all participating patients. An abnormal UtADV was assumed if a bilaterally increased pulsatility index (PI) greater than the 95th centile and/or a distinct postsystolic incision (“notch”) were detected [16]. All measurements were performed by two experienced observers following the recommendations for Doppler ultrasonography measurements in obstetrics [17]. For ultrasound examinations, the Voluson 730, Voluson G8 (both GE Medical Systems, Milwaukee, WI, USA), and HDI5000 SonoCT (Philips Medical Systems, Bothell, WA, USA) were used. Gestational age was calculated from the first day of the last menstrual period and was corrected by ultrasound if measurements of the crown-rump-length during the first trimester revealed a difference of more than 7 days. PE was defined as being present when blood pressure was ≥140/90 mmHg (taken twice, 6 h apart) combined with proteinuria ≥300 mg in a 24-h collection [18]. Inclusion of patients with chronic hypertension (blood pressure level ≥140/90 mmHg prior to 20 weeks of gestation) was possible. In these cases, new onset of proteinuria ≥300 mg per day was regarded as superimposed PE. According to the clinical manifestation, PE was assigned to early onset (<34 weeks of gestation) or late onset (≥34 weeks) [19]. Haemolysis, elevated liver enzymes, and low platelets (HELLP) syndrome was defined as being present, when the platelet count was less than 100 × 109/L (normal range: 150–450 × 109/L), haptoglobin serum levels were below 0.3 g/L (normal range: 0.3–2.0 g/L), and aspartate transaminase (AST) levels were >70 U/L (normal range: 3–34 U/L). Only singleton pregnancies were included. Exclusion criteria were maternal age <18 years, on-going therapy with glucocorticoids or non-steroidal analgesics (with exception of aspirin 100 mg daily), and the presence of anti-phospholipid syndrome.
Small for gestational age (SGA) was defined as a birth weight less than the 10th centile for gestational age according to Voigt et al. [20]. Intrauterine growth restriction (IUGR) was assumed when a SGA birth weight was combined with at least one of the following criteria: (1) oligohydramnios with an amniotic fluid index <5 cm, (2) persistent bilateral abnormal UtADV, (3) a pathological cardiogram, (4) a decrease in foetal growth by ultrasound biometry (“crossing centiles”), and (5) increased PI of the umbilical artery greater than the 95th centile. Because clinicians were not aware of biomarker results, an effect of decision making for the obstetric approach was excluded.

Sample collection and immunoassays

From each study patient, 2 × 7.5 mL of venous blood was taken at the time of Doppler measurement using a serum collection tube (Sarstedt, Nümbrecht, Germany). The blood clot was immediately separated by centrifugation at 2000 × g for 10 min at 15°C and serum was stored in 1-mL aliquots at -80°C until further investigations were performed. Serum concentrations of PlGF and sFLT-1 were measured with the fully automated Elecsys® sandwich immunoassays (Roche Diagnostics, Penzberg, Germany) based on streptavidin-biotin technology. The assays were performed using the Roche immunoanalyser Elecsys® 2010/cobas e 411 according to the recommendations of the manufacturer. Within and between run coefficients of variation were below 4% for sFLT-1 > 60 pg/mL and PlGF >20 pg/mL. Functional sensitivity was <5 pg/mL [21, 22]. Results were classified using gestational age-dependent centile curves that were provided by the manufacturer and based on data from Verlohren and colleagues [10]. Results were regarded as pathological if sFLT-1 or the ratio of sFLT-1/PlGF was greater than the 95th centile and/or PlGF was less than the 5th centile.

Statistical analysis

All data were stored and analysed using the IBM SPSS statistical package 19.0 (SPSS Inc. Chicago, IL, USA) and Excel 2010 (Microsoft Corporation, Redmond, WA, USA). Descriptive statistics were computed for continuous and categorical variables. The statistics included mean, median, standard deviation (SD), minimum, maximum, and number of available observations of continuous variables. Values of continuous variables are presented as mean ± SD. For categorical variables, frequencies and relative frequencies are shown. Testing for differences of continuous variables between the groups was accomplished by the ANOVA or the Kruskal–Wallis test, as appropriate. Test selection was based on evaluation of the variables for normal distribution by using the Kolmogorov–Smirnov test. Comparison between the groups for categorical variables was performed using the chi-square test. All P values resulted from two-sided statistical tests and values of P < 0.05 were considered as statistically significant. Criteria of diagnostic validity for sFLT-1 and PlGF alone, as well as the sFLT-1/PlGF ratio, were computed and presented as sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), positive likelihood ratio, and negative likelihood ratio. Furthermore, receiver operating characteristic (ROC) curves were calculated and the areas under the curves (AUCs) were reported. The most useful cut-off values were calculated by identification of the highest score for sensitivity and specificity. For the sFLT-1/PlGF ratio >95th centile, odds ratios (OR) were computed for various outcome parameters. The logistic regression model was used to assess the independence of specific outcome parameters. Variables yielding P values ≤0.05 in univariate analysis were entered in the multivariate model and adjusted for maternal body mass index, parity, onset of PE, existence of chronic hypertension, week of gestation at the moment of blood sampling, and use of acetylsalicylic acid. We performed post hoc analysis for estimation of the statistical power of our study collective by logistic regression analysis using G*Power 3.1.9.2 [23]. One-tailed power analysis for prediction of PE by the sFLT-1/PlGF ratio with an α error of 0.05 revealed a statistical power of 95.5% for our sample size.

Results

Patients’ characteristics

The patients’ characteristics are shown in Table 1 with the following prevalence of subgroups: PE was found in 24.0% (12/50) of patients, IUGR in 12.0% (6/50), gestational hypertension in 32.0% (16/50), and others in 32.0% (16/50). Subgroup analysis did not show any differences between maternal characteristics, such as age, gravidity, parity, and body mass index. In contrast, significant intergroup differences were found regarding foetal/neonatal parameters. More than 40% of all preeclamptic newborns showed a SGA birth weight. Newborns in the PE subgroup also presented with the lowest mean gestational age. Consequently, the mean birth weight in cases of PE was significantly lower compared with that in the other subgroups, including newborns with normotensive IUGR. Delivery at <34 + 0 weeks occurred in 14.0% (7/50) of all patients. Five of them (71.4%) had PE and two had severe hypertension without proteinuria. All of these patients had an induced delivery because of a severe clinical course indicated by the physicians on duty. One patient with chronic hypertension and concomitant diabetes mellitus type II had an intrauterine foetal death at 26 weeks of gestation. Induction of lung maturation with betamethasone was not performed in three newborns with induced delivery at <34 + 0 weeks, including the patient with intrauterine foetal death. Doppler measurements were repeated in all of the patients after 4 weeks. An abnormal UtADV normalized in 32.0% (16/50) of all cases. There were significant differences between subgroups (P = 0.047) with normalization of an abnormal uterine Doppler in 0.0% of PE (0/12), 33.3% of IUGR (2/6), 31.3% of gestational hypertension (5/16), and 56.3% in the remaining patients (9/16).
Table 1
Maternal and foetal characteristics of the study population
Characteristic
All patients (n = 50)
PE (n = 12)
IUGR (n = 6)#
GH (n = 16)
Others (n = 16)
P-value
Age, y
      
Mean ± SD
29.7 ± 4.9
29.8 ± 3.9
33.3 ± 4.8
28.3 ± 3.6
29.6 ± 6.3
0.206*
Minimum - maximum
19 - 40
24 - 38
28 - 40
22 - 34
19 - 39
 
Gravidity
     
0.613**
Median
1
1
2
1
2
 
Minimum - maximum
1 - 9
1 - 9
1 - 8
1 - 5
1 - 5
 
Parity
     
0.612**
Median
0
0
0
0
0
 
Minimum - maximum
0 - 5
0 - 4
0 - 5
0 - 2
0 - 1
 
Body mass index (pregravid), kg/m 2
     
0.132*
Mean ± SD
27.1 ± 8.2
30.6 ± 8.0
21.7 ± 3.2
28.1 ± 9.1
25.6 ± 6.3
 
Minimum - maximum
14.5 - 47.0
19.8 - 45.0
18.7 - 27.3
14.5 - 45.7
18.8 - 47.0
 
Chronic hypertension, n (%)
8 (16.0%)
4 (33.3%)
0 (0.0%)
4 (25.0%)
0 (0.0%)
0.049***
Thrombophilia, n (%)
5 (10.0%)
0 (0.0%)
0 (0.0%)
3 (18.8%)
2 (12.5%)
0.755***
Nicotine abuse, n (%)
14 (28.0%)
5 (41.7%)
1 (16.7%)
6 (37.5%)
2 (12.5%)
0.238***
Highest systolic blood pressure, mmHg
     
< 0.001*
Mean ± SD
143.2 ± 23.1
165.8 ± 21.2
128.7 ± 5.4
149.4 ± 20.0
124.7 ± 9.9
 
Minimum - maximum
110 - 200
140 - 200
120 - 135
130 - 200
110 - 140
 
Highest diastolic blood pressure, mmHg
     
< 0.001*
Mean ± SD
90.4 ± 17.6
110.5 ± 13.4
79.7 ± 6.2
91.8 ± 15.9
77.3 ± 7.1
 
Minimum - maximum
60 - 130
90 - 130
70 - 86
60 - 124
60 - 90
 
Proteinuria, mg/d
      
Mean ± SD
1180 ± 2524
2613 ± 3394
n.a.
157 ± 196
n.a.
 
Minimum - maximum
0 - 9986
109 - 9986
n.a.
0 - 283
n.a.
 
Placenta weight, g
     
0.129*
Mean ± SD
324 ± 98
300 ± 86
322 ± 29
286 ± 145
443 ± 71
 
Minimum - maximum
124 - 510
182 - 461
283 - 364
124 - 476
369 - 510
 
Amnion fluid index (AFI), n (%)
     
0.839***
Oligohydramnion (AFI ≤ 8)
13 (26.0%)
3 (25.0%)
2 (33.3%)
4 (25.0%)
4 (25.0%)
 
Polyhydramnion (AFI > 18)
7 (14.0%)
3 (25.0%)
1 (16.7%)
1 (6.3%)
2 (12.5%)
 
Birth weight, g
     
< 0.001*
Mean ± SD
2588 ± 809
1859 ± 658
2270 ± 303
2588 ± 750
3175 ± 626
 
Minimum - maximum
620 - 3940
890 - 2980
1750 - 2620
620 - 3610
1860 - 3940
 
Percentile of birth weight, n (%)
      
SGA (<10th centile)
16 (32.0%)
5 (41.7%)
6 (100.0%)
6 (37.5%)
0 (0.0%)
 
LGA (>90th centile)
0 (0.0%)
0 (0.0%)
0 (0.0%)
0 (0.0%)
0 (0.0%)
 
5´-APGAR-Score
     
0.398**
Median
10
9
10
10
10
 
Minimum - maximum
8 - 10
8 - 10
9 - 10
8 - 10
9 - 10
 
Induction of lung maturation, n (%)
13 (26.0%)
4 (33.3%)
6 (100.0%)
2 (12.5%)
1 (6.3%)
0.019***
Newborns < 34 + 0 weeks, n (%)
7 (14.0%)
5 (41.7%)
0 (0.0%)
2 (12.5%)
0 (0.0%)
0.008***
Gestational age at delivery, weeks + days
     
0.009**
Mean (w + d) ± SD (d)
37 + 2 ± 24
34 + 4 ± 26
37 + 5 ± 5
37 + 4 ± 25
38 + 6 ± 17
 
Minimum - maximum
26 + 4 - 42 + 0
27 + 4 - 40 + 3
36 + 5 - 39 + 0
26 + 2 – 41 + 0
34 + 1 - 42 + 0
 
# normotensive IUGR only.
* = ANOVA.
** = Kruskal–Wallis test.
*** = Chi-Square test.
n.a. = not available, PE = Preeclampsia, GH = Gestational hypertension, IUGR = Intrauterine growth restriction, SGA = Small for gestational age, LGA = Large for gestational age.

Prediction of PE by UtADV, PlGF, sFLT-1, and the sFLT-1/PlGF ratio

The prevalence of PE in our high-risk study collective was 24.0% (12/50), with 75.0% (9/12) early-onset and 25.0% (3/12) late-onset diseases. PE was also present with chronic hypertension in four cases (all early onset). One patient developed concomitant HELLP syndrome. For prediction of PE, bilateral UtADV showed a PPV of only 24.0% with a false-positive rate (FPR) of 76.0%. Performance of ROC analysis using the mean PI of the uterine arteries showed an AUC of 0.630. At the optimal cut-off with a mean uterine PI ≥1.86, the specificity increased to 72.2%, with a PPV of 38.2%. However, the false negative rate also increased to 45.5%, resulting in a predictive accuracy of only 70.0%. In contrast, serum levels of PlGF and sFLT-1, as well as the sFLT-1/PlGF ratio, clearly improved discrimination between patients with and without PE (Figure 1, Table 2). If values were greater than the 95th centile of gestation, the sFLT-1/PlGF ratio showed most useful test characteristics for PE, with a sensitivity of 66.7% and an FPR of only 10.5% (Table 3). The corresponding PPV was 66.7%, with an NPV of 89.5% and a predictive accuracy of the ratio of 84.0%. An elevated sFLT-1/PlGF ratio greater than the 95th centile was associated with an increased risk for development of PE (OR 17.0; 95% confidence interval [CI] 3.5–83.0, P < 0.001) (Table 3). Results remained significant after weighting of parameters in multiple regression analysis with an adjusted OR for PE (adjusted OR 14.1; 95% CI 1.1–182.6, P = 0.043).
Table 2
Maternal serum PlGF and sFLT-1 levels and their ratio
 
PE (n = 12)
No PE (n = 38)
P-valuea
Others (n = 16) (Uncomplicated)
GH (n = 16)
P-valueb
Normotensive IUGR (n = 6)
P-valuec
Delivery < 34 weeks (n = 7)
Delivery ≥ 34 weeks (n = 44)
P-valued
PlGF (pg/mL)
105.6 (70.7-248.7)
218.5 (131.2-322.5)
0.041
274.6 (157.9-413.6)
198.7 (91.5-305.3)
0.142
165.9 (72.1-220.7)
0.033
75.1 (64.6-131.7)
221.7 (117.4-336.4)
0.003
sFLT-1 (pg/mL)
1879.0 (1379.5-2168.3)
1144.0 (842.2-1882.8)
0.046
1458.0 (1018.8-1896.3)
1032.6 (826.8-1649.8)
0.258
1324.4 (673.1-2012.0)
0.768
2158.0 (1858.0-3463.0)
1298.0 (846.1-1876.0)
0.008
sFLT-1/PlGF ratio
18.8 (4.8-29.1)
6.2 (3.4-8.8)
0.022
5.9 (2.9-8.6)
5.9 (3.6-9.1)
0.366
8.6 (4.0-19.6)
0.161
29.4 (14.1-53.9)
6.0 (3.4-9.0)
0.001
Data are given as median (interquartile range).
a Mann–Whitney statistical test comparing PE vs No PE.
b Mann–Whitney statistical test comparing GH vs Others.
c Mann–Whitney statistical test comparing IUGR vs Others.
d Mann–Whitney statistical test comparing delivery < 34 weeks vs delivery ≥ 34 weeks.
PE = Preeclampsia, GH = Gestational hypertension, IUGR = Intrauterine growth restriction.
Table 3
Diagnostic indices of maternal PlGF, sFLT-1, and their ratio for identifying patients who will develop preeclampsia, hypertension without proteinuria, normotensive IUGR, and induced delivery at <34 weeks of gestation
  
Chi-Square P-value
Sensitivity
Specificity
PPV
NPV
LR + (95% CI)
LR- (95% CI)
OR (95% CI)
PE (n = 12)
PlGF
0.027
58.3%
78.9%
46.7%
85.7%
2.77 (1.27-6.04)
0.53 (0.27-1.05)
5.25 (1.31-21.03)
 
sFLT-1
0.240
16.7%
94.7%
50.0%
78.3%
3.17 (0.50-20.13)
0.88 (0.68-1.15)
3.60 (0.45-28.86)
 
sFLT-1/PlGF ratio
<0.001
66.7%
89.5%
66.7%
89.5%
6.33 (2.31-17.38)
0.37 (0.17-0.84)
17.0 (3.50-83.02)
Gestational hypertension (n = 16)
PlGF
0.514
37.5%
73.5%
40.0%
71.4%
1.42 (0.61-3.30)
0.85 (0.55-1.31)
1.67 (0.47-5.92)
 
sFLT-1
1.000
6.3%
91.2%
25.0%
67.4%
0.71 (0.08-6.29)
1.03 (0.87-1.21)
0.69 (0.067-7.19)
 
sFLT-1/PlGF ratio
0.292
12.5%
70.6%
16.7%
63.2%
0.43 (0.11-1.72)
1.24 (0.93-1.65)
0.34 (0.07-1.79)
IUGR (n = 6)
PlGF
0.654
16.7%
68.2%
6.7%
85.7%
0.52 (0.08-3.30)
1.22 (0.81-1.84)
0.43 (0.05-4.02)
 
sFLT-1
1.000
0.0%
90.9%
0.0%
87.0%
0.00
1.10 (1.00-1.21)
0.00
 
sFLT-1/PlGF ratio
1.000
16.7%
75.0%
8.3%
86.8%
0.67 (0.10-4.29)
1.11 (0.75-1.65)
0.60 (0.06-5.71)
Induced delivery < 34 weeks (n = 7)
PlGF
0.176
57.1%
74.4%
26.7%
91.4%
2.23 (0.98-5.07)
0.58 (0.34-1.38)
3.88 (0.75-20.12)
 
sFLT-1
0.089
28.6%
95.4%
50.3%
89.1%
6.14 (1.03-36.79)
0.75 (0.47-1.20)
8.20 (0.94-71.73)
 
sFLT-1/PlGF ratio
<0.001
85.7%
86.1%
50.1%
97.4%
6.14 (2.76-13.69)
0.17 (0.03-1.02)
37.00 (3.76-363.91)
Early-onset PE (n = 9)
PlGF
0.220
50.0%
73.8%
26.7%
88.6%
1.91 (0.81-4.51)
0.68 (0.33-1.39)
2.82 (0.60-13.24)
 
sFLT-1
0.115
25.0%
95.2%
50.0%
87.0%
5.25 (0.86-32.02)
0.79 (0.53-1.18)
6.67 (0.79-56.64)
 
sFLT-1/PlGF ratio
0.001
75.0%
85.7%
50.0%
94.7%
5.25 (2.26-12.18)
0.29 (0.09-0.98)
18.00 (2.92-110.96)
Late-onset PE (n = 3)
PlGF
0.075
75.0%
73.9%
20.0%
97.1%
2.88 (1.36-6.06)
0.34 (0.06-1.86)
8.50 (0.81-89.75)
 
sFLT-1
1.000
0.0%
91.3%
0.0%
91.3%
0.00
1.10 (1.00-1.20)
0.00
 
sFLT-1/PlGF ratio
0.240
50.0%
78.3%
16.7%
94.7%
2.30 (0.75-7.07)
0.64 (0.24-1.72)
3.60 (0.46-28.86)
Cut-off levels were defined as < 5th centile for PlGF and > 95th centile for sFLT-1 and the sFLT-1/PlGF ratio according to gestational specific reference values.
PE = Preeclampsia, GH = Gestational hypertension, IUGR = Intrauterine growth restriction, PPV = Positive predictive value, NPV = Negative predictive value, OR = Odds ratio, LR+/- = positive/negative Likelihood ratio.
The sensitivity increased to 75.0% if only early-onset PE was included, but FPR also increased to 14.3%. We found four false-positive cases, where one patient showed a normotensive IUGR and two had hypertension without proteinuria (one with concomitant IUGR and one with intrauterine foetal death at 26 weeks of gestation). Two out of the four false-negative patients developed early-onset PE (one patient with concomitant HELLP syndrome at <34 weeks of gestation) and two developed late-onset PE (with only a mild clinical course in one patient). The fourth patient with false-negative serum markers developed late-onset PE, but fulfilled the criteria of severity with hypertension >160/110 mmHg. ROC analysis showed an AUC of 0.721 for the sFLT-1/PlGF ratio, with the best predictive cut-off of 13.0, resulting in a sensitivity of 66.7% with an FPR of 13.2% (Figure 2, Table 4). If a fixed FPR of 10% was assumed, the cut-off level of the ratio changed to 22.4, with a sensitivity of only 41.7%.
Table 4
Results of receiver operating characteristic curve analysis with AUC values and cut-off levels of maternal serum PlGF (pg/mL) and sFLT-1 (pg/mL) levels and their ratio, resulting in optimal sensitivity and specificity
  
AUC
Cut-off
Sensitivity
Specificity
PPV
NPV
PE (n = 12)
PlGF
0.697
133.8
75.0%
76.3%
50.0%
90.6%
 
sFLT-1
0.693
1171.5
91.7%
52.6%
37.9%
95.3%
 
sFLT-1/PlGF ratio
0.721
13.0
66.7%
86.8%
61.5%
89.2%
EO PE (n = 9)
sFLT-1/PlGF ratio
0.735
13.0
75.0%
83.3%
51.4%
93.4%
Gestational hypertension (n = 16)
PlGF
0.494
117.6
37.5%
70.6%
37.5%
70.6%
 
sFLT-1
0.364
590.5
100.0%
11.8%
34.8%
100.0%
 
sFLT-1/PlGF ratio
0.456
2.3
100.0%
8.8%
34.0%
100.0%
IUGR (n = 6)
PlGF
0.640
262.3
100.0%
38.6%
18.2%
100.0%
 
sFLT-1
0.439
1751.0
50.0%
63.6%
15.8%
90.3%
 
sFLT-1/PlGF ratio
0.564
8.0
66.7%
56.8%
17.4%
92.6%
Induced delivery < 34 weeks (n = 7)
PlGF
0.834
133.8
85.7%
72.1%
33.3%
96.9%
 
sFLT-1
0.807
1850.5
85.7%
74.4%
35.3%
97.0%
 
sFLT-1/PlGF ratio
0.877
13.0
85.7%
83.7%
46.1%
97.3%
PE = Preeclampsia, EO = Early-onset, IUGR = Intrauterine growth restriction, ID = Induced delivery, PPV = positive predictive value, NPV = negative predictive value.

Prediction of hypertension without proteinuria, IUGR, and induced delivery at <34 weeks of gestation

Results were further analysed in respect to development of hypertension without proteinuria and IUGR. As shown in Table 2, serum levels of PlGF were significantly lower in cases of IUGR compared to uncomplicated controls (P = 0.033), but failed significance if used as predictive parameter throughout the whole study population (Table 3). However, we found significant differences of the angiogenic markers if results were analysed in relation to gestational age at delivery (Figure 3). Measurements of sFLT-1 and PlGF were useful predictive markers for delivery at <34 + 0 weeks of gestation. If ROC analysis was performed, the AUC for the sFLT-1/PlGF ratio was 0.877, with a best cut-off of 13.0 (Figure 2). This cut-off resulted in a sensitivity of 85.7% with an FPR of 16.3%. If gestational age-dependent threshold levels greater than the 95th centile were used, the FPR improved to 13.9% with the same sensitivity. The PPV and NPV were 50.1% and 97.4%, respectively. The ratio of sFLT-1/PlGF showed a predictive accuracy of 86.0%. An elevated sFLT-1/PlGF ratio greater than the 95th centile was associated with an increased risk for delivery at <34 + 0 weeks (OR 37.0; 95% CI 3.8–363.9, P < 0.001) (Table 3). Results remained significant after weighting of parameters in multiple regression analysis with an adjusted OR (34.6; 95% CI 1.2–1026.2, P = 0.041).

Discussion

Main findings

Patients with an abnormal UtADV showed distinct foetal and maternal morbidity, with a high prevalence of PE, IUGR, and hypertensive disorders without proteinuria. However, only one of four patients with abnormal UtADV developed PE. Additional measurement of the sFLT-1/PlGF ratio was able to decrease the FPR from 76.0% to 10.5%. A negative test result below the 95th gestational age-specific centile showed an NPV of 89.5%. The NPV was further improved if only patients with early onset PE (94.7%) or those who required delivery at <34 weeks (97.4%) were considered. Therefore, women with a normal sFLT-1/PlGF ratio were at low risk for development of one of these severe clinical courses. Additional measurement of sFLT-1 and PlGF would be helpful to reassure the majority of patients with an abnormal UtADV. An increased angiogenic ratio was associated with an increased risk of a severe hypertensive disorder and concomitant preterm delivery. Provident corticosteroid treatment of these women should be considered.

Interpretation

There are different approaches for prediction of PE. Doppler ultrasound of the uterine arteries during the second trimester is the best-known single predictor for PE, with a detection rate up to 80% for early-onset disease [14, 2426]. However, the majority of patients with abnormal UtADV will never develop PE, which suggests the need to improve the predictive accuracy. Therefore, use of a well-established and widely performed test for identification of a high risk collective and its combination with measurement of angiogenic factors for improvement of specificity is a practically oriented approach. The use of a fully automated diagnostic system that analyses sFLT-1 and PlGF guarantees a high test quality. The Elecsys® immunoassays show high precision and reliability, and allow execution of measurements in laboratories for routine clinical diagnosis [21].
Recent studies have reported complex algorithms that allow prediction of PE by integration of miscellaneous risk factors, including analysis of angiogenic factors between 11–13 weeks of gestation [2729]. Identification of high-risk patients during the first trimester offers the advantage of a preventive therapy with low-dose aspirin, which more than halves the risk of severe PE [30]. However, the ability of prediction or exclusion of PE during the second trimester may also be useful because it can facilitate decision making as to whether intensified patient’s care or corticosteroid treatment for foetal lung maturation will be necessary. There is also increasing evidence for effective preventive treatment strategies in high-risk patients during the second trimester [3133]. Various (nested) case–control studies have examined the differences in maternal angiogenic protein concentrations during the second trimester preceding the occurrence of PE [8, 3441]. The usefulness of circulating sFLT-1 and PlGF as a test to predict PE in healthy pregnant women was recently proven in a comprehensive prospective cohort study by McElrath and colleagues [11]. In a cohort of 2243 patients, the plasma levels of angiogenic factors were repeatedly measured at 10, 17, 24, and 35 weeks of gestation. In the study sample, 139 (6.2%) patients developed PE. At 10 weeks of gestation, the authors did not find any significant differences between plasma concentrations of PlGF or sFLT-1, or the sFLT-1/PIGF ratio in patients with PE compared with healthy controls. Differences became significant at 17 weeks for PlGF (P = 0.009) alone, and were highly significant for PlGF and the sFLT-1/PIGF ratio at 24 weeks (P < 0.001). In contrast, differences for sFLT-1 became significant at 35 weeks. At 24 weeks, the highest detection rate (62.2%) was observed for PlGF alone, with an FPR of 38.4% and a PPV of 9.5%. The relative risk for development of PE was 2.46 (95% CI 1.66–-3.65) if a cut-off level of 379 pg/mL was used [11]. Similar results were shown in a further prospective study including 1622 healthy Hispanic women [25]. Maternal plasma concentrations of various angiogenic proteins were measured at 6–15 and 20–25 weeks of gestation. During pregnancy, 3.8% of patients developed PE (n = 62), and 0.6% had early-onset PE (n = 9). In midtrimester, PlGF measurements alone showed a better test performance (P < 0.001) than sFLT-1 (P = 0.5) or the sFLT-1/PIGF ratio (P = 0.006). However, the detection rate for PlGF was only 51.6%, with an FPR of 23.6% and a PPV of 8%. A reasonable cut-off value was 215 pg/mL, resulting in an OR of 3.8 (95% CI 2.2–6.7). The detection rate for early-onset PE was 100%, and was therefore better, with an FPR of only 4.2% at a cut-off level of 126 pg/mL [25]. The authors of both studies concluded that angiogenic protein concentrations alone, measured in early or even late pregnancy, were of limited utility for prediction of PE. For prediction of PE, its low incidence and concomitant low predictive value are problematic. Therefore, identification of high-risk patients may be an alternative, more suitable approach. In a prospective cohort study of 3348 patients, Espinoza and colleagues examined the relationship between UtADV and plasma concentrations of PlGF and sFLT-1 between 22 and 26 weeks of gestation [24]. In this study, they showed that abnormal UtADV and maternal PlGF were independent explanatory variables for the occurrence of PE. For prediction of PE, the FPR of abnormal UtADV alone was low (10.4%). Abnormal UtADV had a better test performance than maternal PlGF for identification of a high-risk collective, with ORs for all types of PE and early-onset PE of 4.3 (95% CI 2.82–6.66) and 24.1 (95% CI 9.61–60.44) compared with PlGF (cut-off, 280 pg/mL), with corresponding ORs of 2.6 (95% CI 1.57–3.94) and 5.5 (95% CI 1.98–15.08). The combination of both parameters increased the OR for early-onset PE to 43.8 (95% CI 18.48–103.9). The PPV for early-onset PE was improved from 5.4% for abnormal UtADV and 1.4% for PlGF alone to 12.7% if both factors were combined with a positive likelihood ratio of 18.48 (95% CI 13.07–26.13). This study [24] impressively showed that PlGF significantly contributed to the prediction of patients who were going to develop early-onset PE, and thus confirmed the results of a smaller study by Stepan and colleagues [42]. The supplemental table compares the test characteristics of the sFLT-1/PlGF ratio of various studies (Additional file 1: Table S1). The prospective cohort study by Moore Simas and colleagues included only patients with at least one risk factor for PE in their medical history [43]. In this study, the determination of sFLT-1 showed a good predictive accuracy for early-onset PE, with an AUC value of 0.90 (95% CI 0.78–1.0) between 22 and 26 weeks of gestation. The sFLT-1/PlGF ratio was even better with an AUC value of 0.97 (95% CI 0.91–1.0). Similar results of other studies, including our own data, showed an increase in specificity if the sFLT-1/PlGF ratio instead of PlGF alone was used for prediction of early-onset PE. However, compared with a decrease in PlGF, an increase in sFLT-1 in patients with PE is a late event, with only a narrow time frame between diagnosis and clinical manifestation of PE [8, 11].
Although the end-point of our study was the development of PE, we additionally analysed the correlation between angiogenic factors and the occurrence of other adverse outcome parameters, such as IUGR and delivery at <34 weeks. The group of induced deliveries showed great overlap with the PE cases and could be regarded as a surrogate for a severe clinical course. Therefore, identification of these patients is of high clinical relevance. With an AUC of 0.88, we observed a satisfactory test performance for prediction of cases with a delivery at <34 weeks of gestation. Although PPV was only 50.1%, we found an NPV of 97.4%. In patients with abnormal UtADV and a normal angiogenic ratio, the need for delivery at <34 weeks following hypertensive pregnancy disorders is highly unlikely. With a sensitivity of 71% and a specificity of 76%, Stepan and colleagues also reported the capability of the sFLT-1/PlGF ratio for prediction of preterm delivery [42]. Additionally, in a prospective study of patients with suspected early-onset PE, the sFLT-1/PlGF ratio showed prognostic relevance [44]. Patients with a ratio ≥85 had a significantly increased risk for delivery within the next 2 weeks, with a hazard ratio of 15.2 (95% CI 8.0–28.7).
In our study, angiogenic factors were not suitable for prediction of hypertension without proteinuria or for normotensive IUGR, although in IUGR, the levels of PlGF were significantly lower compared with inconspicuous controls. Differences reported in other studies were also only moderate, and not helpful for diagnosis or prediction of these adverse pregnancy outcome parameters [12, 41, 42].

Conclusion

In high-risk patients, measurement of angiogenic factors shows a good predictive power for development of PE. The best results can be achieved using the ratio of sFLT-1 and PlGF, with a predictive test accuracy of 84%. In patients with abnormal UtADV, a high FPR of Doppler measurement alone is improved to 10.5%. An increased sFLT-1/PlGF ratio is also predictive for patients who need delivery at <34 + 0 weeks of gestation. The test performed even better for prediction of preterm delivery compared with PE, with a sensitivity and specificity of >80%. In particular, a NPV of 97.4% can help to reassure most patients with an abnormal UtADV.

Acknowledgements

The authors would like to thank Ms Erika Greschkowitz for her assistance in laboratory processing. The study was financed by own resources of the department of Obstetrics and Gynaecology of the University of Rostock.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

JS conceived and designed the study, analysed and interpreted the data, and drafted the manuscript. SU acquired and analysed the patients’ data, and drafted the manuscript. MB acquired patients, performed the ultrasound examination and took blood samples. TK acquired patients, performed the ultrasound examination and took blood samples. MD analysed and interpreted the data, and wrote the manuscript. DUR designed the study and acquired the lab data. TR conceived and designed the study, and wrote the manuscript. All authors have seen and approved the final version of the manuscript.
Literatur
2.
Zurück zum Zitat Gaillard R, Arends LR, Steegers EA, Hofman A, Jaddoe VW: Second- and third-trimester placental hemodynamics and the risks of pregnancy complications: the Generation R Study. Am J Epidemiol. 2013, 177: 743-754.CrossRefPubMed Gaillard R, Arends LR, Steegers EA, Hofman A, Jaddoe VW: Second- and third-trimester placental hemodynamics and the risks of pregnancy complications: the Generation R Study. Am J Epidemiol. 2013, 177: 743-754.CrossRefPubMed
3.
Zurück zum Zitat Zhang WH, Alexander S, Bouvier-Colle MH, Macfarlane A: Incidence of severe pre-eclampsia, postpartum haemorrhage and sepsis as a surrogate marker for severe maternal morbidity in a European population-based study: the MOMS-B survey. BJOG. 2005, 112: 89-96.CrossRefPubMed Zhang WH, Alexander S, Bouvier-Colle MH, Macfarlane A: Incidence of severe pre-eclampsia, postpartum haemorrhage and sepsis as a surrogate marker for severe maternal morbidity in a European population-based study: the MOMS-B survey. BJOG. 2005, 112: 89-96.CrossRefPubMed
4.
Zurück zum Zitat Lisonkova S, Joseph KS: Incidence of preeclampsia: risk factors and outcomes associated with early- versus late-onset disease. Am J Obstet Gynecol. 2013, 209: 544-CrossRefPubMed Lisonkova S, Joseph KS: Incidence of preeclampsia: risk factors and outcomes associated with early- versus late-onset disease. Am J Obstet Gynecol. 2013, 209: 544-CrossRefPubMed
5.
Zurück zum Zitat MacKay AP, Berg CJ, Atrash HK: Pregnancy-related mortality from preeclampsia and eclampsia. Obstet Gynecol. 2001, 97: 533-538.PubMed MacKay AP, Berg CJ, Atrash HK: Pregnancy-related mortality from preeclampsia and eclampsia. Obstet Gynecol. 2001, 97: 533-538.PubMed
6.
Zurück zum Zitat Odegard RA, Vatten LJ, Nilsen ST, Salvesen KA, Austgulen R: Preeclampsia and fetal growth. Obstet Gynecol. 2000, 96: 950-955.PubMed Odegard RA, Vatten LJ, Nilsen ST, Salvesen KA, Austgulen R: Preeclampsia and fetal growth. Obstet Gynecol. 2000, 96: 950-955.PubMed
7.
Zurück zum Zitat Maynard SE, Min JY, Merchan J, Lim KH, Li J, Mondal S, Libermann TA, Morgan JP, Sellke FW, Stillman IE, Epstein FH, Sukhatme VP, Karumanchi SA: Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest. 2003, 111: 649-658.CrossRefPubMedPubMedCentral Maynard SE, Min JY, Merchan J, Lim KH, Li J, Mondal S, Libermann TA, Morgan JP, Sellke FW, Stillman IE, Epstein FH, Sukhatme VP, Karumanchi SA: Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest. 2003, 111: 649-658.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, Schisterman EF, Thadhani R, Sachs BP, Epstein FH, Sibai BM, Sukhatme VP, Karumanchi SA: Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med. 2004, 350: 672-683.CrossRefPubMed Levine RJ, Maynard SE, Qian C, Lim KH, England LJ, Yu KF, Schisterman EF, Thadhani R, Sachs BP, Epstein FH, Sibai BM, Sukhatme VP, Karumanchi SA: Circulating angiogenic factors and the risk of preeclampsia. N Engl J Med. 2004, 350: 672-683.CrossRefPubMed
9.
Zurück zum Zitat Reimer T, Rohrmann H, Stubert J, Pecks U, Glocker MO, Richter DU, Gerber B: Angiogenic factors and acute-phase proteins in serum samples of preeclampsia and HELLP patients: a matched-pair analysis. J Matern Fetal Neonatal Med. 2013, 26: 263-269.CrossRefPubMed Reimer T, Rohrmann H, Stubert J, Pecks U, Glocker MO, Richter DU, Gerber B: Angiogenic factors and acute-phase proteins in serum samples of preeclampsia and HELLP patients: a matched-pair analysis. J Matern Fetal Neonatal Med. 2013, 26: 263-269.CrossRefPubMed
10.
Zurück zum Zitat Verlohren S, Galindo A, Schlembach D, Zeisler H, Herraiz I, Moertl MG, Pape J, Dudenhausen JW, Denk B, Stepan H: An automated method for the determination of the sFLT-1/PIGF ratio in the assessment of preeclampsia. Am J Obstet Gynecol. 2010, 202: 161-CrossRefPubMed Verlohren S, Galindo A, Schlembach D, Zeisler H, Herraiz I, Moertl MG, Pape J, Dudenhausen JW, Denk B, Stepan H: An automated method for the determination of the sFLT-1/PIGF ratio in the assessment of preeclampsia. Am J Obstet Gynecol. 2010, 202: 161-CrossRefPubMed
11.
Zurück zum Zitat McElrath TF, Lim KH, Pare E, Rich-Edwards J, Pucci D, Troisi R, Parry S: Longitudinal evaluation of predictive value for preeclampsia of circulating angiogenic factors through pregnancy. Am J Obstet Gynecol. 2012, 207: 407-CrossRefPubMed McElrath TF, Lim KH, Pare E, Rich-Edwards J, Pucci D, Troisi R, Parry S: Longitudinal evaluation of predictive value for preeclampsia of circulating angiogenic factors through pregnancy. Am J Obstet Gynecol. 2012, 207: 407-CrossRefPubMed
12.
Zurück zum Zitat McElrath TF, Lim KH, Pare E, Rich-Edwards J, Pucci D, Troisi R, Parry S: The sFLT-1/PlGF ratio in different types of hypertensive pregnancy disorders and its prognostic potential in preeclamptic patients. Am J Obstet Gynecol. 2012, 206: 58-CrossRef McElrath TF, Lim KH, Pare E, Rich-Edwards J, Pucci D, Troisi R, Parry S: The sFLT-1/PlGF ratio in different types of hypertensive pregnancy disorders and its prognostic potential in preeclamptic patients. Am J Obstet Gynecol. 2012, 206: 58-CrossRef
13.
Zurück zum Zitat Thadhani R, Kisner T, Hagmann H, Bossung V, Noack S, Schaarschmidt W, Jank A, Kribs A, Cornely OA, Kreyssig C, Hemphill L, Rigby AC, Khedkar S, Lindner TH, Mallmann P, Stepan H, Karumanchi SA, Benzing T: Pilot study of extracorporeal removal of soluble fms-like tyrosine kinase 1 in preeclampsia. Circulation. 2011, 124: 940-950.CrossRefPubMed Thadhani R, Kisner T, Hagmann H, Bossung V, Noack S, Schaarschmidt W, Jank A, Kribs A, Cornely OA, Kreyssig C, Hemphill L, Rigby AC, Khedkar S, Lindner TH, Mallmann P, Stepan H, Karumanchi SA, Benzing T: Pilot study of extracorporeal removal of soluble fms-like tyrosine kinase 1 in preeclampsia. Circulation. 2011, 124: 940-950.CrossRefPubMed
14.
Zurück zum Zitat Cnossen JS, Morris RK, ter Riet G, Mol BW, van der Post JA, Coomarasamy A, Zwinderman AH, Robson SC, Bindels PJ, Kleijnen J, Khan KS: Use of uterine artery Doppler ultrasonography to predict pre-eclampsia and intrauterine growth restriction: a systematic review and bivariable meta-analysis. CMAJ. 2008, 178: 701-711.CrossRefPubMedPubMedCentral Cnossen JS, Morris RK, ter Riet G, Mol BW, van der Post JA, Coomarasamy A, Zwinderman AH, Robson SC, Bindels PJ, Kleijnen J, Khan KS: Use of uterine artery Doppler ultrasonography to predict pre-eclampsia and intrauterine growth restriction: a systematic review and bivariable meta-analysis. CMAJ. 2008, 178: 701-711.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Myatt L, Clifton RG, Roberts JM, Spong CY, Hauth JC, Varner MW, Wapner RJ, Thorp JM, Mercer BM, Grobman WA, Ramin SM, Carpenter MW, Samuels P, Sciscione A, Harper M, Tolosa JE, Saade G, Sorokin Y, Anderson GD: The utility of uterine artery Doppler velocimetry in prediction of preeclampsia in a low-risk population. Obstet Gynecol. 2012, 120: 815-822.CrossRefPubMedPubMedCentral Myatt L, Clifton RG, Roberts JM, Spong CY, Hauth JC, Varner MW, Wapner RJ, Thorp JM, Mercer BM, Grobman WA, Ramin SM, Carpenter MW, Samuels P, Sciscione A, Harper M, Tolosa JE, Saade G, Sorokin Y, Anderson GD: The utility of uterine artery Doppler velocimetry in prediction of preeclampsia in a low-risk population. Obstet Gynecol. 2012, 120: 815-822.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Schaffer H: Doppler-Referenzkurven. Dopplersonographie in Geburtshilfe und Gynäkologie. 2000, Heidelberg Berlin New York: Springer Verlag, 292-293. Schaffer H: Doppler-Referenzkurven. Dopplersonographie in Geburtshilfe und Gynäkologie. 2000, Heidelberg Berlin New York: Springer Verlag, 292-293.
17.
Zurück zum Zitat Bhide A, Acharya G, Bilardo CM, Brezinka C, Cafici D, Hernandez-Andrade E, Kalache K, Kingdom J, Kiserud T, Lee W, Lees C, Leung KY, Malinger G, Mari G, Prefumo F, Sepulveda W, Trudinger B: ISUOG practice guidelines: use of Doppler ultrasonography in obstetrics. Ultrasound Obstet Gynecol. 2013, 41: 233-239.CrossRefPubMed Bhide A, Acharya G, Bilardo CM, Brezinka C, Cafici D, Hernandez-Andrade E, Kalache K, Kingdom J, Kiserud T, Lee W, Lees C, Leung KY, Malinger G, Mari G, Prefumo F, Sepulveda W, Trudinger B: ISUOG practice guidelines: use of Doppler ultrasonography in obstetrics. Ultrasound Obstet Gynecol. 2013, 41: 233-239.CrossRefPubMed
18.
Zurück zum Zitat Report of the National High Blood Pressure Education Program Working Group on High Blood Pressure in Pregnancy. Am J Obstet Gynecol. 2000, 183: S1-S22. Report of the National High Blood Pressure Education Program Working Group on High Blood Pressure in Pregnancy. Am J Obstet Gynecol. 2000, 183: S1-S22.
19.
Zurück zum Zitat von Dadelszen P, Magee LA, Roberts JM: Subclassification of preeclampsia. Hypertens Pregnancy. 2003, 22: 143-148.CrossRefPubMed von Dadelszen P, Magee LA, Roberts JM: Subclassification of preeclampsia. Hypertens Pregnancy. 2003, 22: 143-148.CrossRefPubMed
20.
Zurück zum Zitat Voigt M, Rochow N, Hesse V, Olbertz D, Schneider KT, Jorch G: Short communication about percentile values of body measures of newborn babies. Z Geburtshilfe Neonatol. 2010, 214: 24-29.CrossRefPubMed Voigt M, Rochow N, Hesse V, Olbertz D, Schneider KT, Jorch G: Short communication about percentile values of body measures of newborn babies. Z Geburtshilfe Neonatol. 2010, 214: 24-29.CrossRefPubMed
21.
Zurück zum Zitat Schiettecatte J, Russcher H, Anckaert E, Mees M, Leeser B, Tirelli AS, Fiedler GM, Luthe H, Denk B, Smitz J: Multicenter evaluation of the first automated Elecsys sFLT-1 and PlGF assays in normal pregnancies and preeclampsia. Clin Biochem. 2010, 43: 768-770.CrossRefPubMed Schiettecatte J, Russcher H, Anckaert E, Mees M, Leeser B, Tirelli AS, Fiedler GM, Luthe H, Denk B, Smitz J: Multicenter evaluation of the first automated Elecsys sFLT-1 and PlGF assays in normal pregnancies and preeclampsia. Clin Biochem. 2010, 43: 768-770.CrossRefPubMed
22.
Zurück zum Zitat Ohkuchi A, Hirashima C, Suzuki H, Takahashi K, Yoshida M, Matsubara S, Suzuki M: Evaluation of a new and automated electrochemiluminescence immunoassay for plasma sFLT-1 and PlGF levels in women with preeclampsia. Hypertens Res. 2010, 33: 422-427.CrossRefPubMed Ohkuchi A, Hirashima C, Suzuki H, Takahashi K, Yoshida M, Matsubara S, Suzuki M: Evaluation of a new and automated electrochemiluminescence immunoassay for plasma sFLT-1 and PlGF levels in women with preeclampsia. Hypertens Res. 2010, 33: 422-427.CrossRefPubMed
23.
Zurück zum Zitat Faul F, Erdfelder E, Buchner A, Lang AG: Statistical power analyses using G*Power 3.1: tests for correlation and regression analyses. Behav Res Methods. 2009, 41: 1149-1160.CrossRefPubMed Faul F, Erdfelder E, Buchner A, Lang AG: Statistical power analyses using G*Power 3.1: tests for correlation and regression analyses. Behav Res Methods. 2009, 41: 1149-1160.CrossRefPubMed
24.
Zurück zum Zitat Espinoza J, Kusanovic JP, Bahado-Singh R, Gervasi MT, Romero R, Lee W, Vaisbuch E, Mazaki-Tovi S, Mittal P, Gotsch F, Erez O, Gomez R, Yeo L, Hassan SS: Should bilateral uterine artery notching be used in the risk assessment for preeclampsia, small-for-gestational-age, and gestational hypertension?. J Ultrasound Med. 2010, 29: 1103-1115.PubMedPubMedCentral Espinoza J, Kusanovic JP, Bahado-Singh R, Gervasi MT, Romero R, Lee W, Vaisbuch E, Mazaki-Tovi S, Mittal P, Gotsch F, Erez O, Gomez R, Yeo L, Hassan SS: Should bilateral uterine artery notching be used in the risk assessment for preeclampsia, small-for-gestational-age, and gestational hypertension?. J Ultrasound Med. 2010, 29: 1103-1115.PubMedPubMedCentral
25.
Zurück zum Zitat Kusanovic JP, Romero R, Chaiworapongsa T, Erez O, Mittal P, Vaisbuch E, Mazaki-Tovi S, Gotsch F, Edwin SS, Gomez R, Yeo L, Conde-Agudelo A, Hassan SS: A prospective cohort study of the value of maternal plasma concentrations of angiogenic and anti-angiogenic factors in early pregnancy and midtrimester in the identification of patients destined to develop preeclampsia. J Matern Fetal Neonatal Med. 2009, 22: 1021-1038.CrossRefPubMedPubMedCentral Kusanovic JP, Romero R, Chaiworapongsa T, Erez O, Mittal P, Vaisbuch E, Mazaki-Tovi S, Gotsch F, Edwin SS, Gomez R, Yeo L, Conde-Agudelo A, Hassan SS: A prospective cohort study of the value of maternal plasma concentrations of angiogenic and anti-angiogenic factors in early pregnancy and midtrimester in the identification of patients destined to develop preeclampsia. J Matern Fetal Neonatal Med. 2009, 22: 1021-1038.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Velauthar L, Plana MN, Kalidindi M, Zamora J, Thilaganathan B, Illanes SE, Khan KS, Aquilina J, Thangaratinam S: Uterine artery Doppler in the first trimester as a risk factor for adverse pregnancy outcomes: A meta-analysis involving 55,974 women. Ultrasound Obstet Gynecol. 2013, 43: 500-507.CrossRef Velauthar L, Plana MN, Kalidindi M, Zamora J, Thilaganathan B, Illanes SE, Khan KS, Aquilina J, Thangaratinam S: Uterine artery Doppler in the first trimester as a risk factor for adverse pregnancy outcomes: A meta-analysis involving 55,974 women. Ultrasound Obstet Gynecol. 2013, 43: 500-507.CrossRef
27.
Zurück zum Zitat Akolekar R, Syngelaki A, Poon L, Wright D, Nicolaides KH: Competing risks model in early screening for preeclampsia by biophysical and biochemical markers. Fetal Diagn Ther. 2013, 33: 8-15.CrossRefPubMed Akolekar R, Syngelaki A, Poon L, Wright D, Nicolaides KH: Competing risks model in early screening for preeclampsia by biophysical and biochemical markers. Fetal Diagn Ther. 2013, 33: 8-15.CrossRefPubMed
28.
Zurück zum Zitat Myers JE, Kenny LC, McCowan LM, Chan EH, Dekker GA, Poston L, Simpson NA, North RA: Angiogenic factors combined with clinical risk factors to predict preterm pre-eclampsia in nulliparous women: a predictive test accuracy study. BJOG. 2013, 120: 1215-1223.CrossRefPubMed Myers JE, Kenny LC, McCowan LM, Chan EH, Dekker GA, Poston L, Simpson NA, North RA: Angiogenic factors combined with clinical risk factors to predict preterm pre-eclampsia in nulliparous women: a predictive test accuracy study. BJOG. 2013, 120: 1215-1223.CrossRefPubMed
29.
Zurück zum Zitat Scazzocchio E, Figueras F, Crispi F, Meler E, Masoller N, Mula R, Gratacos E: Performance of a first-trimester screening of preeclampsia in a routine care low-risk setting. Am J Obstet Gynecol. 2013, 208: 203-CrossRefPubMed Scazzocchio E, Figueras F, Crispi F, Meler E, Masoller N, Mula R, Gratacos E: Performance of a first-trimester screening of preeclampsia in a routine care low-risk setting. Am J Obstet Gynecol. 2013, 208: 203-CrossRefPubMed
30.
Zurück zum Zitat Roberge S, Giguere Y, Villa P, Nicolaides K, Vainio M, Forest JC, von Dadelszen P, Vaiman D, Tapp S, Bujold E: Early administration of low-dose aspirin for the prevention of severe and mild preeclampsia: a systematic review and meta-analysis. Am J Perinatol. 2013, 29: 551-556. Roberge S, Giguere Y, Villa P, Nicolaides K, Vainio M, Forest JC, von Dadelszen P, Vaiman D, Tapp S, Bujold E: Early administration of low-dose aspirin for the prevention of severe and mild preeclampsia: a systematic review and meta-analysis. Am J Perinatol. 2013, 29: 551-556.
31.
Zurück zum Zitat Schleussner E, Lehmann T, Kahler C, Schneider U, Schlembach D, Groten T: Impact of the nitric oxide-donor pentaerythrityl-tetranitrate on perinatal outcome in risk pregnancies: a prospective, randomized, double-blinded trial. J Perinat Med. 2014, 42: 507-514.CrossRefPubMed Schleussner E, Lehmann T, Kahler C, Schneider U, Schlembach D, Groten T: Impact of the nitric oxide-donor pentaerythrityl-tetranitrate on perinatal outcome in risk pregnancies: a prospective, randomized, double-blinded trial. J Perinat Med. 2014, 42: 507-514.CrossRefPubMed
32.
Zurück zum Zitat Becker R, Keller T, Kiesewetter H, Fangerau H, Bittner U: Individual risk assessment of adverse pregnancy outcome by multivariate regression analysis may serve as basis for drug intervention studies: retrospective analysis of 426 high-risk patients including ethical aspects. Arch Gynecol Obstet. 2013, 288: 41-48.CrossRefPubMedPubMedCentral Becker R, Keller T, Kiesewetter H, Fangerau H, Bittner U: Individual risk assessment of adverse pregnancy outcome by multivariate regression analysis may serve as basis for drug intervention studies: retrospective analysis of 426 high-risk patients including ethical aspects. Arch Gynecol Obstet. 2013, 288: 41-48.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Belizan JM, Villar J, Gonzalez L, Campodonico L, Bergel E: Calcium supplementation to prevent hypertensive disorders of pregnancy. N Engl J Med. 1991, 325: 1399-1405.CrossRefPubMed Belizan JM, Villar J, Gonzalez L, Campodonico L, Bergel E: Calcium supplementation to prevent hypertensive disorders of pregnancy. N Engl J Med. 1991, 325: 1399-1405.CrossRefPubMed
34.
Zurück zum Zitat Tidwell SC, Ho HN, Chiu WH, Torry RJ, Torry DS: Low maternal serum levels of placenta growth factor as an antecedent of clinical preeclampsia. Am J Obstet Gynecol. 2001, 184: 1267-1272.CrossRefPubMed Tidwell SC, Ho HN, Chiu WH, Torry RJ, Torry DS: Low maternal serum levels of placenta growth factor as an antecedent of clinical preeclampsia. Am J Obstet Gynecol. 2001, 184: 1267-1272.CrossRefPubMed
35.
Zurück zum Zitat Villa PM, Hamalainen E, Maki A, Raikkonen K, Pesonen AK, Taipale P, Kajantie E, Laivuori H: Vasoactive agents for the prediction of early- and late-onset preeclampsia in a high-risk cohort. BMC Pregnancy Childbirth. 2013, 13: 110-CrossRefPubMedPubMedCentral Villa PM, Hamalainen E, Maki A, Raikkonen K, Pesonen AK, Taipale P, Kajantie E, Laivuori H: Vasoactive agents for the prediction of early- and late-onset preeclampsia in a high-risk cohort. BMC Pregnancy Childbirth. 2013, 13: 110-CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Krauss T, Pauer HU, Augustin HG: Prospective analysis of placenta growth factor (PlGF) concentrations in the plasma of women with normal pregnancy and pregnancies complicated by preeclampsia. Hypertens Pregnancy. 2004, 23: 101-111.CrossRefPubMed Krauss T, Pauer HU, Augustin HG: Prospective analysis of placenta growth factor (PlGF) concentrations in the plasma of women with normal pregnancy and pregnancies complicated by preeclampsia. Hypertens Pregnancy. 2004, 23: 101-111.CrossRefPubMed
37.
Zurück zum Zitat Hassan MF, Rund NM, Salama AH: An Elevated Maternal Plasma Soluble fms-Like Tyrosine Kinase-1 to Placental Growth Factor Ratio at Midtrimester Is a Useful Predictor for Preeclampsia. Obstet Gynecol Int. 2013, 2013: 202346-CrossRefPubMedPubMedCentral Hassan MF, Rund NM, Salama AH: An Elevated Maternal Plasma Soluble fms-Like Tyrosine Kinase-1 to Placental Growth Factor Ratio at Midtrimester Is a Useful Predictor for Preeclampsia. Obstet Gynecol Int. 2013, 2013: 202346-CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Unal ER, Robinson CJ, Johnson DD, Chang EY: Second-trimester angiogenic factors as biomarkers for future-onset preeclampsia. Am J Obstet Gynecol. 2007, 197: 211-214.CrossRefPubMed Unal ER, Robinson CJ, Johnson DD, Chang EY: Second-trimester angiogenic factors as biomarkers for future-onset preeclampsia. Am J Obstet Gynecol. 2007, 197: 211-214.CrossRefPubMed
39.
Zurück zum Zitat Tjoa ML, van Vugt JM, Mulders MA, Schutgens RB, Oudejans CB, van Wijk IJ: Plasma placenta growth factor levels in midtrimester pregnancies. Obstet Gynecol. 2001, 98: 600-607.PubMed Tjoa ML, van Vugt JM, Mulders MA, Schutgens RB, Oudejans CB, van Wijk IJ: Plasma placenta growth factor levels in midtrimester pregnancies. Obstet Gynecol. 2001, 98: 600-607.PubMed
40.
Zurück zum Zitat Vatten LJ, Eskild A, Nilsen TI, Jeansson S, Jenum PA, Staff AC: Changes in circulating level of angiogenic factors from the first to second trimester as predictors of preeclampsia. Am J Obstet Gynecol. 2007, 196: 239-6.CrossRefPubMed Vatten LJ, Eskild A, Nilsen TI, Jeansson S, Jenum PA, Staff AC: Changes in circulating level of angiogenic factors from the first to second trimester as predictors of preeclampsia. Am J Obstet Gynecol. 2007, 196: 239-6.CrossRefPubMed
41.
Zurück zum Zitat Romero R, Nien JK, Espinoza J, Todem D, Fu W, Chung H, Kusanovic JP, Gotsch F, Erez O, Mazaki-Tovi S, Gomez R, Edwin S, Chaiworapongsa T, Levine RJ, Karumanchi SA: A longitudinal study of angiogenic (placental growth factor) and anti-angiogenic (soluble endoglin and soluble vascular endothelial growth factor receptor-1) factors in normal pregnancy and patients destined to develop preeclampsia and deliver a small for gestational age neonate. J Matern Fetal Neonatal Med. 2008, 21: 9-23.CrossRefPubMedPubMedCentral Romero R, Nien JK, Espinoza J, Todem D, Fu W, Chung H, Kusanovic JP, Gotsch F, Erez O, Mazaki-Tovi S, Gomez R, Edwin S, Chaiworapongsa T, Levine RJ, Karumanchi SA: A longitudinal study of angiogenic (placental growth factor) and anti-angiogenic (soluble endoglin and soluble vascular endothelial growth factor receptor-1) factors in normal pregnancy and patients destined to develop preeclampsia and deliver a small for gestational age neonate. J Matern Fetal Neonatal Med. 2008, 21: 9-23.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Stepan H, Unversucht A, Wessel N, Faber R: Predictive value of maternal angiogenic factors in second trimester pregnancies with abnormal uterine perfusion. Hypertension. 2007, 49: 818-824.CrossRefPubMed Stepan H, Unversucht A, Wessel N, Faber R: Predictive value of maternal angiogenic factors in second trimester pregnancies with abnormal uterine perfusion. Hypertension. 2007, 49: 818-824.CrossRefPubMed
43.
Zurück zum Zitat Moore Simas TA, Crawford SL, Solitro MJ, Frost SC, Meyer BA, Maynard SE: Angiogenic factors for the prediction of preeclampsia in high-risk women. Am J Obstet Gynecol. 2007, 197: 244-248.CrossRefPubMed Moore Simas TA, Crawford SL, Solitro MJ, Frost SC, Meyer BA, Maynard SE: Angiogenic factors for the prediction of preeclampsia in high-risk women. Am J Obstet Gynecol. 2007, 197: 244-248.CrossRefPubMed
44.
Zurück zum Zitat Rana S, Powe CE, Salahuddin S, Verlohren S, Perschel FH, Levine RJ, Lim KH, Wenger JB, Thadhani R, Karumanchi SA: Angiogenic factors and the risk of adverse outcomes in women with suspected preeclampsia. Circulation. 2012, 125: 911-919.CrossRefPubMedPubMedCentral Rana S, Powe CE, Salahuddin S, Verlohren S, Perschel FH, Levine RJ, Lim KH, Wenger JB, Thadhani R, Karumanchi SA: Angiogenic factors and the risk of adverse outcomes in women with suspected preeclampsia. Circulation. 2012, 125: 911-919.CrossRefPubMedPubMedCentral
Metadaten
Titel
Prediction of preeclampsia and induced delivery at <34 weeks gestation by sFLT-1 and PlGF in patients with abnormal midtrimester uterine Doppler velocimetry: a prospective cohort analysis
verfasst von
Johannes Stubert
Stefanie Ullmann
Michael Bolz
Thomas Külz
Max Dieterich
Dagmar-Ulrike Richter
Toralf Reimer
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
BMC Pregnancy and Childbirth / Ausgabe 1/2014
Elektronische ISSN: 1471-2393
DOI
https://doi.org/10.1186/1471-2393-14-292

Weitere Artikel der Ausgabe 1/2014

BMC Pregnancy and Childbirth 1/2014 Zur Ausgabe

Hirsutismus bei PCOS: Laser- und Lichttherapien helfen

26.04.2024 Hirsutismus Nachrichten

Laser- und Lichtbehandlungen können bei Frauen mit polyzystischem Ovarialsyndrom (PCOS) den übermäßigen Haarwuchs verringern und das Wohlbefinden verbessern – bei alleiniger Anwendung oder in Kombination mit Medikamenten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Weniger postpartale Depressionen nach Esketamin-Einmalgabe

Bislang gibt es kein Medikament zur Prävention von Wochenbettdepressionen. Das Injektionsanästhetikum Esketamin könnte womöglich diese Lücke füllen.

Bei RSV-Impfung vor 60. Lebensjahr über Off-Label-Gebrauch aufklären!

22.04.2024 DGIM 2024 Kongressbericht

Durch die Häufung nach der COVID-19-Pandemie sind Infektionen mit dem Respiratorischen Synzytial-Virus (RSV) in den Fokus gerückt. Fachgesellschaften empfehlen eine Impfung inzwischen nicht nur für Säuglinge und Kleinkinder.

Update Gynäkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert – ganz bequem per eMail.