Next Article in Journal
Age-Related Modifications of Electroencephalogram Coherence in Mice Models of Alzheimer’s Disease and Amyotrophic Lateral Sclerosis
Previous Article in Journal
PSMA Expression Correlates with Improved Overall Survival and VEGF Expression in Glioblastoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Communication

Maternal Plasma RNA in First Trimester Nullipara for the Prediction of Spontaneous Preterm Birth ≤ 32 Weeks: Validation Study

1
Department of Obstetrics and Gynecology, University of Kansas Medical Center, Kansas City, KS 66160, USA
2
Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA
3
Rosetta Signaling Laboratory LLC, Phoenix, AZ 85018, USA
4
Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
5
Fetal Medicine Research Institute, King’s College Hospital, London SE5 9RS, UK
6
Departments of Anatomy and Physiology & Midwest Institute of Comparative Stem Cell Biology, Kansas State University, Manhattan, KS 66503, USA
*
Author to whom correspondence should be addressed.
Current address: Medical School, Creighton University, Phoenix, AZ 85013, USA.
Current address: Maternal Fetal Medicine, Dignity Health Medical Group, CommonSpirit Health System, 500 W, Thomas Rd., Suite 700, Phoenix, AZ 85013, USA.
Biomedicines 2023, 11(4), 1149; https://doi.org/10.3390/biomedicines11041149
Submission received: 27 February 2023 / Revised: 31 March 2023 / Accepted: 10 April 2023 / Published: 11 April 2023
(This article belongs to the Section Molecular and Translational Medicine)

Abstract

:
The first-trimester prediction of spontaneous preterm birth (sPTB) has been elusive, and current screening is heavily dependent on obstetric history. However, nullipara lack a relevant history and are at higher risk for spontaneous (s)PTB ≤ 32 weeks compared to multipara. No available objective first-trimester screening test has proven a fair predictor of sPTB ≤ 32 weeks. We questioned whether a panel of maternal plasma cell-free (PCF) RNAs (PSME2, NAMPT, APOA1, APOA4, and Hsa-Let-7g) previously validated at 16–20 weeks for the prediction of sPTB ≤ 32 weeks might be useful in first-trimester nullipara. Sixty (60) nulliparous women (40 with sPTB ≤ 32 weeks) who were free of comorbidities were randomly selected from the King’s College Fetal Medicine Research Institute biobank. Total PCF RNA was extracted and the expression of panel RNAs was quantitated by qRT-PCR. The analysis employed, primarily, multiple regression with the main outcome being the prediction of subsequent sPTB ≤ 32 weeks. The test performance was judged by the area under the curve (AUC) using a single threshold cut point with observed detection rates (DRs) at three fixed false positive rates (FPR). The mean gestation was 12.9 ± 0.5 weeks (range 12.0–14.1 weeks). Two RNAs were differentially expressed in women destined for sPTB ≤ 32 weeks: APOA1 (p < 0.001) and PSME2 (p = 0.05). APOA1 testing at 11–14 weeks predicted sPTB ≤ 32 weeks with fair to good accuracy. The best predictive model generated an AUC of 0.79 (95% CI 0.66–0.91) with observed DRs of 41%, 61%, and 79% for FPRs of 10%, 20%, and 30%, including crown–rump length, maternal weight, race, tobacco use, and age.

1. Introduction

Preterm birth (PTB) is the leading cause of perinatal mortality and neonatal morbidity worldwide. Over 15 million PTBs annually lead to more than a million childhood deaths and lifelong sequelae for many survivors [1]. The global failure to reduce the rates of very (32 to 28 weeks) and extreme preterm (<28 weeks) birth reflects a poor understanding of the mechanisms leading to these births, paired with a limited ability to accurately identify women early in pregnancy who are at ‘high risk’ for PTB ≤ 32 weeks. Since the majority of prematurity-related costs are associated with these births [2,3], their accurate prediction and prevention would have maximal patient and societal impact.
Current screening for PTB is heavily dependent on obstetric history, but nullipara lack an obstetric history and are at higher risk for spontaneous (s)PTB ≤ 32 weeks compared to multipara [4]. No available screening test performed under 20 weeks has proven a fair predictor of sPTB ≤ 32 weeks. Smith [5] tested the predictive value of maternal characteristics and second-trimester alpha-fetoprotein and ß-human chorionic gonadotropin in 84,000 nulliparas. No model generated a receiver operating characteristic area under the curve (AUC) above 0.67. Van der Ven [6] tested the predictive ability of transvaginal sonographic cervical length (CL) at 16–21.9 weeks in 5700 nulliparas and 6200 low-risk multiparas. Though CL performed slightly better in nullipara, it remained a poor predictor (sPTB < 37 weeks, AUC 0.61 vs. 0.56; sPTB < 34 weeks, AUC 0.63 vs. 0.58), a finding supported by Rosenbloom [7]. Esplin [8] studied 9400 nulliparas at 16–22 weeks with both CL and cervical fetal fibronectin (fFn). No AUC exceeded 0.53 for sPTB < 37 weeks and 0.61 for sPTB < 32 weeks. While Esplin did not combine the markers at 16–22 weeks, they did at 22–30 weeks; there was no improvement in the accuracy compared to CL alone. Clearly, these tests are used by caregivers, not because of their accuracy, but because there is a testing void.
Prior reports demonstrate a maternal blood test consisting of five plasma cell-free (PCF) RNAs (PSME2, NAMPT, APOA1, APOA4, and Hsa-Let-7g) measured at 16–20 weeks may be a good predictor of sPTB ≤ 32 weeks [9,10]. A prospective cohort validation study revealed specific panel markers had good to excellent predictive accuracy for PTB ≤ 32 weeks, whether due to spontaneous labor (± preterm premature rupture of membranes (PPROM)) or early-onset preeclampsia (EOP) < 34 weeks independent of parity [10]. While the predictive accuracy for sPTB ≤ 32 weeks was encouraging (AUC = 0.83), antenatal screening has moved progressively to the first trimester for EOP and potentially sPTB ≤ 32 weeks. The objective of this study was to test the potential of the five RNA panel to predict sPTB ≤ 32 weeks in the first trimester nullipara.

2. Materials and Methods

In collaboration with the Fetal Medicine Research Institute, King’s College Hospital, London, UK, 60 first-trimester women prospectively enrolled in the Institute’s IRB-approved biobank for future research were randomly selected by one of the authors (AS) subject to the following conditions: nulliparous, healthy, and a pregnancy ending either at term or with sPTB ≤ 32 weeks. There were 20 controls and 40 cases. The racial distribution was the same for cases and controls: 50% self-described White and Black. Women who developed preeclampsia or had known comorbidities were excluded from this study. Gestational age was based on the first-trimester crown–rump length (CRL).

2.1. Molecular Tests

RNA was extracted from 500 µL of EDTA plasma using a proprietary method (Rosetta Signaling Laboratory, Phoenix, AZ, USA). The mean total RNA extracted was 26.7 ± 9.5 µg/mL (median 18.7 µg/mL; range 8.0–76.3 µg/mL). RNA yield was assessed with a Nano spectrometer (NanoDrop Technologies, Wilmington, DE, USA) and integrity confirmed by the Agilent Bio-analyzer (Agilent, Santa Clara, CA, USA). RNA expression was quantified by qRT-PCR, as described previously [9]. All laboratory members were blind to pregnancy outcome.

2.2. qRT-PCR Assays

mRNA RT: RNA samples were diluted with a master mix including dNTP mix, Omniscript Reverse Transcriptase, and Random Primer (Invitrogen, Carlsbad, CA, USA). mRNA was converted into cDNA at 37 °C for 60 min, per manufacturer instructions.
miRNA RT: miRs were polyadenylated using the Invitrogen NCode miRNA First-Strand cDNA Synthesis Kit (ThermoFisher, Waltham, MA, USA). The polyadenylated microRNA was reverse-transcribed to generate the first strand of cDNA per manufacturer’s instructions.
Preamplification and qPCR: Multiplex qPCR reactions used SYBR green and the ViiA 7 Real-Time PCR System (ThermoFisher, Waltham, MA, USA). The primers were custom-designed and synthesized by Integrated DNA Technologies (IDT, Coralville, IA, USA) [9]. The probe sets included primers for one of the five potential markers (PSME2, NAMPT, APOA1, APOA4, and Hsa-Let-7g) plus normalization and spike genes, so all three RNAs were run in the same well. One-µL RT samples were prepared for the preamplification mix reaction and underwent twelve cycles. Two µL of preamplification cDNA samples were diluted into 10µL PCR reaction mix, followed by RT PCR using SYBR Green Supermix (ThermoFisher, Waltham, MA, USA). Threshold cycles (Ct values) of qPCR reactions were extracted using QuantStudio™ Software V1.3 (Applied Biosystems, Foster City, CA, USA). Deltadelta CT method was used to calculate the expression of RNAs and then normalized.

2.3. Statistics

All statistical analyses were performed by one author (HSC) using SAS 9.4 (SAS Institute, Cary, NC, USA). Initial studies of laboratory tests are often modest in size, and metrics such as sensitivity, specificity, and negative/positive predictive values are highly influenced by disease prevalence. In contrast, the AUC is less affected by disease prevalence and provides an aggregate measure of performance across all classification thresholds and is one of the most useful parameters to evaluate a predictive model [11]. An AUC between 0.90 and 1.00 is considered excellent, one between 0.80 and 0.89 is good, 0.70–0.79 is fair, 0.60–0.69 is poor, and 0.50–0.59 is a failure [12]. We used AUC with detection rates (DRs) derived for three fixed false positive rates (FPRs), since an acceptable FPR could vary if the cost of misclassifications was part of model selection [13].
Gaussian modeling was not possible with the sample size, so a variety of linear regressions were tested with and without log base 2 transformation of expression and conversion to multiples of the median (MoM) [14]. The maternal characteristics available included maternal (MA) and gestational ages (GA), CRL, maternal weight (MW), race, and tobacco use. AUC was calculated for optimized models using a single threshold cut point and observed DRs determined for 3 fixed FPRs of 10, 20, and 30%.

3. Results

The mean MA (±1SD) was 32.1 ± 1.6 y and the mean GA was 12.9 ± 0.1 weeks (12.0–14.1 weeks). Two controls were sampled at 14.0 and 14.1 weeks; the remaining 18 controls and 40 cases were sampled ≤13.9 weeks. The mean MW was 72.9 ± 15.5 kg. There were no differences between the case and control groups in either the maternal characteristics or the gestation at sampling. There was a trend toward obesity in cases (p = 0.09). Three women smoked tobacco: 1 control and 2 cases. The mean (±SD (range)) GA at delivery was 39.99 ± 0.8 weeks (39.1–41.8 weeks) for controls and 27.9 ± 2.3 weeks (24.1–31.8 weeks) for cases.
The five RNAs comprising the current panel were previously shown to have low expression in controls but overexpressed in cases [9,10], and in the current study, there were subjects in whom one or more of the RNA levels fell below assay detection, and more often than was seen in the 16–20 weeks study [10]. PSME2 was undetectable in 5% (1/20) of controls and 10% (4/40) of cases (Figure 1), NAMPT was undetectable in 60% (12/20) of controls and 52.5% (21/40) of cases, APOA1 was undetectable in 50% (10/20) of controls and 17.5% (7/40) of cases, APOA4 was undetectable in 65% (13/20) of controls and 32.5% (13/40) of cases, and, lastly, Hsa-LET-7g was undetectable in 10% (2/20) of controls and 5% (2/40) of cases. Overall, panel RNA expression was more likely to be undetected in controls than cases (Chi-square with Yates’ correction 6.2073, p = 0.012).
Two of the five panel RNAs, APOA1 and PSME2, were differentially expressed in women destined for sPTB ≤ 32 weeks (Figure 1). One subject in each group had expression levels below detection for both RNA markers (‘no calls’, 3.3% of subjects).
The predictive accuracy of the significant models is listed in Table 1. Converting gene expression to MoMs had a modest impact on the AUCs, while the addition of CRL and some maternal characteristics appeared to enhance predictive accuracy. APOA1 (MoMs) (AUC = 0.73, 95% CI 0.60–0.87) performed better than PSME2 (MoMs) (0.65, 95% CI 0.50–0.80). The combination of APOA1 and PSME2 did not significantly improve AUC over APOA1 alone. The two best models were: APOA1 (MoMs) + MA yielding an AUC = 0.79 (95% CI 0.66–0.91) and DR = 75% with a 30% FPR, and APOA1 (MoMs) + CRL, MA, MW, race, and tobacco use yielding an AUC = 0.79 (95% CI 0.66–0.91) and DR = 79% with a 30% FPR.

4. Discussion

No validated clinical screening test to date has provided even a ‘fair’ predictive accuracy (i.e., AUC > 0.70) for sPTB ≤ 32 weeks in the first trimester of parous, much less nulliparous, women. The current study is the third validation study of this five RNA panel specifically selected based on their relationship to a unique set of myometrial RNAs overexpressed in women delivering <32 weeks [9]. Like the prospective cohort validation study performed at 16–20 weeks [10], only two of the five panel RNAs were predictive, PSME2 and APOA1 at 12–14 weeks, and APOA1 was the best performer. The RNA-only AUC for APOA1 = 0.73 (95% CI 0.60–0.87) provided a DR = 75%. This DR is similar to the RNA-only AUC = 0.76 (95% CI 0.65–0.87) achieved in a prospective cohort study at 16–20 weeks [10], suggesting the overall accuracy of the RNA panel for sPTB ≤ 32 weeks is unaffected by parity and may be useful in the first trimester. Combining APOA1 with CRL, MW, MA, race, and tobacco use achieved an AUC = 0.79 (95% CI 0.66–0.91) for sPTB ≤ 32 weeks with a DR = 79%. This is similar to the AUC = 0.83 (95% CI 0.74–0.92) achieved in the 16–20-week samples with a DR = 77% when race and a history of prior PTB were combined [10]. The fact that the majority of women destined for sPTB ≤ 32 weeks can be identified by 12–14 weeks suggests that their risk is already ‘set’ in the absence of therapeutic intervention. It also suggests sPTB ≤ 32 weeks is not a syndrome with numerous causes that may begin in the second or third trimesters.
While medical history, CL, and fFN are used clinically to identify women at risk for sPTB, studies suggest the popularity of these tests is not based on their predictive accuracy, as all three have repeatedly been shown to be clinically poor tests [4,5,6]. Rather, caregivers and patients alike have nothing better to use. The risk prediction by maternal characteristics/history performs worse in nullipara compared to multipara, to the point of not being clinically useful for the prediction of sPTB > 32 weeks [15]. In the current study, the available maternal characteristics were limited but still seemingly associated with an AUC improvement. It is possible a larger sample size coupled with a more robust list of characteristics including prepregnancy-related hypertension, diabetes, or a family history of preterm birth would raise the first trimester AUC > 0.80 for sPTB ≤ 32 weeks. Dude [16] noted that a similar percentage of women with a short CL delivered prematurely, independent of parity, suggesting premature CL shortening is not impacted by prior pregnancy. Additionally, after prior studies of two different populations in which the five RNA panel at 16–20 weeks provided reproducible accuracy superior to conventional screening methods, this current study that was confined to first-trimester nullipara suggests plasma RNA testing has the potential to be a first-trimester screening tool.
Others have sought biomarkers for sPTB in asymptomatic women [17,18,19,20] but only Cook [21] and Hromadnikova [22] focused on the first trimester. Cook sampled women 12 to 21 weeks 6 d. There were 13 cases used for discovery but only 10 cases used for validation. Differentially expressed miRNA for sPTB < 34 weeks was sought using the Nanostring nCounter miRNA assay and Q rtPCR. In the 12–14 weeks 6 d group, Cook identified five differentially expressed miRNA. Another four miRNAs were differentially expressed at 15–21 weeks 6 d. In the validation phase, all 9 miRNAs were differentially expressed from the earliest gestation forward. The AUCs for individual miRNAs in the validation study ranged from 0.82 to 0.87 for the four best markers. This laboratory has not yet published a follow-up validation study. Hromadnikova [22] studied maternal peripheral leucocyte RNA but limited their investigation to 29 cardiovascular disease-associated microRNAs and sPTB < 37 weeks. Their best model predictive of sPTB < 37 weeks yielded an AUC = 0.81 that consisted of six miRNAs. The addition of MA and serum PAPP-A did not improve performance. The validation study has yet to be published.
There is reason to believe one or more RNAs comprising the current five RNA panel will be predictive of preeclampsia and/or EOP in the first trimester, just as occurred at 16–20 weeks [10]. Del Vecchio [23] described a first-trimester discovery study using RNAseq on plasma mRNA from 17 women in the late first trimester, 9 with a normal delivery at term, 5 with preeclampsia, and 3 with gestational hypertension. They identified 170 differentially expressed RNAs in the 8 women who developed pregnancy-related hypertension and selected 5 for logistic regression modeling on the same dataset used for discovery. The result was AUC = 0.86. One of their selected markers was NAMPT, which in the second trimester is a predictor of preeclampsia and EOP [10]. Their remaining PCF RNAs were MMP8, SRPK1, S100A9, and S100A8. No validation study has been published to date. In the current first-trimester validation study and in our prior second-trimester cohort validation study [10], the informative marker RNAs for sPTB ≤ 32 weeks achieved similar AUCs, but only PSME2 was predictive of sPTB ≤ 32 weeks in both the first and second trimesters. Yet, in the first trimester, it did not add to the predictive accuracy of APOA1 and may be a sample size issue. Not being able to detect the expression of NAMPT cannot be explained by a laboratory failure, as APOA4 had the highest undetectable rates in the first trimester.
The clinical value of a screening test would be increased exponentially if there was an effective intervention available. Prophylactic progesterone is recommended in several scenarios. Additionally, while some professional organizations consider vaginal progesterone the standard of care for certain indications [24], there is clearly no international consensus on effectiveness [25,26]. Perhaps efficacy could be improved by a more accurate selection of at-risk women, as occurred with low-dose aspirin and EOP [27]. It was recently shown [28] that the rate of sPTB < 32 weeks in women deficient in DHA could be halved by DHA supplementation beginning in the early second trimester. Supplementation had no impact on sPTB rates in women with normal DHA levels. The ability to objectively identify a high-risk pool of women sPTB ≤ 32 weeks in the first trimester would facilitate future therapeutic trials, whether that be a current or new therapy. However, whether the prevention therapy is progesterone, DHA [28,29,30], cervical cerclage, or pessary, it is clear that new therapeutic approaches are needed. In addition, since the financial costs of sPTB ≤ 32 weeks represent the majority of healthcare costs in year one of life, even a small decrease in the rate of sPTB ≤ 32 weeks would likely be cost-effective [31].
As encouraging as the association of differentially expressed PCF RNAs is with pregnancy pathology, there is a clear need for large, real-world prospective studies to confirm the reproducibility of RNA markers among laboratories, and their predictive accuracy when other pathologies are present.
Interestingly, APOA1 at 16–20 weeks is a good predictor of EOP and not sPTB ≤ 32 weeks [10]. These differences are of clinical and biologic importance. First, they highlight the need for accurate dating. Second, they suggest these differentially expressed PCF RNAs that likely originate from the placenta [9] may alter their cell target as the placenta develops, perhaps by being packaged in a different transporter. The impact of gestation on informative RNAs for sPTB ≤ 32 weeks is consistent with our hypothesis as to why RNAs that are rigorously selected to predict sPTB ≤ 32 weeks could prove to be better predictors of EOP at other gestations [10]. In silica, the five PCF RNAs comprising the current panel have the potential to enhance intracellular calcium in smooth muscle, and in vitro, the overexpression of APOA4 in immortalized human pregnant myometrial cells increases both intracellular calcium and cell contraction frequency [9]. This suggests that one of the potential effects of these overexpressed RNAs is interference with myometrial quiescence.
PCF RNAs rarely circulate naked; rather, they are associated with transporters such as extracellular vesicles, carrier proteins such as Argonaute 2 [32], or high-density lipoproteins [33]. These transporters slow RNA degradation and provide a targeting mechanism for RNA uptake by specific cells. sPTB and EOP share abnormal smooth muscle responsiveness. We have hypothesized sPTB occurs when marker RNAs are within transporters targeting myometrial smooth muscle, while EOP occurs when marker RNAs are within transporters targeting vascular smooth muscle (and/or endothelium) [10]. We have shown that increased APOA1 is predictive of sPTB ≤ 32 weeks at 12 weeks, but also of EOP at 16–20 weeks. Thus, the maternal disease phenotype may depend as much on the RNA transporter as it does on the contained RNA. In support of this hypothesis, Yoffe et al. [34] described a first-trimester study of small noncoding RNAs using Next Generation Sequencing seeking differentially expressed RNAs that were predictive of EOP. They identified two of the same differentially expressed miRs that we identified in 2nd-trimester women [9], miR Let-7g and 99b. However, at 16–20 weeks, these were predictors of sPTB ≤ 32 weeks and not EOP, as Yoffe found at 11–13 weeks.
The present validation study has several strengths. First, it is the third successful validation study of the five PCF RNA panel. Second, the samples were well documented and randomly chosen by an otherwise uninvolved party from a large biobank in a country previously not used for the testing of the panel. Third, the sample size, though limited, is similar to or larger than similar efforts from other laboratories. A fourth strength of the study is the use of a plasma RNA extraction method that increases total RNA yield from the nanograms extracted by existing commercial kits to microgram levels [35]. No PCF RNA test can be reproducible if the RNA extraction is not.
The study also has limitations. First, subject selection occurred a decade ago, early in our experience with the plasma transcriptome. We intentionally excluded women who either developed preeclampsia or had other medical comorbidities, out of concern comorbid conditions in a limited sample size could have significant but nonidentifiable effects. As a result, we have no information about the applicability of the five PCF RNA panel in the first trimester to preeclampsia. Second, the case-control nature of the study engenders a high prevalence of cases and creates the potential for a biased sample selection. The insulated sample selection process from a very large biobank should have minimized sample selection bias. Further, the use of AUC rather than sensitivity, specificity, etc., helps offset the impact of high disease prevalence. A third potential weakness is a reliance on regression analysis, as overfitting could exaggerate the AUCs. However, the changing expression of the marker RNAs as gestation advances prevented us from testing first-trimester expression with AUC curves generated from our larger second-trimester samples. Lastly, there was a limited number of maternal characteristics available for inclusion in the regression models. However, this particular weakness would have acted to lower the composite AUC rather than falsely elevate it. The inclusion of additional characteristics such as socioeconomic status, prepregnancy diseases (e.g., diabetes) and a family history of a prior preterm birth in the future may enhance accuracy [36,37].

5. Conclusions

Including the present study, there are now five validation studies [9,10,18,20] of varying size, conducted from 12.0 to 24 weeks, reaching similar conclusions: PCF RNAs combined with maternal characteristics identify with good accuracy those women who are destined for sPTB ≤ 32 weeks (labor ± PPROM), months in advance of the event. Expanded first-trimester testing will reveal whether the current markers are also useful for the prediction of other pregnancy disorders such as EOP/preeclampsia, as they were in the second trimester. This confirmation will make a first-trimester test available for the three most common causes of PTB ≤ 32 weeks and open the door to precision medicine in obstetrics (Figure 2).

6. Patents

EU Patent No. 2646554 and US Patent No. 10,954,564.

Author Contributions

Conceptualization, C.P.W. and Y.D.; validation, C.P.W., H.Z. and Y.D.; formal analysis, C.P.W., H.C., M.L.W.; C.P.W., H.Z. and Y.D.; patient enrollment, A.S. and K.H.N.; data curation, H.Z.; writing—original draft preparation, C.P.W.; writing—review and editing, C.P.W., H.C., M.L.W., H.Z., A.S. and K.H.N.; supervision, Y.D.; project administration, C.P.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The biobank materials were collected and the research described was performed under one or more of the following IRB protocol approval numbers: King’s College Hospital, London, UK: REC reference # 02-03-033 (Approval 12 August 2009); KUMC Research Institute IRB—#10452 (Approval 15 June 2006).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

All data is available on request to the authors.

Acknowledgments

The authors are grateful to the thousands of pregnant women who provided biologic samples to the Fetal Medicine Foundation to support such research. The authors also thank Ellen Weiss for the design and production of Figure 2.

Conflicts of Interest

Carl P. Weiner: Founder of Rosetta Signaling Laboratory, which controls the intellectual property related to plasma nucleic acid extraction and the RNA markers described, including EU Patent No. 2646554, US patent No. 10,954,564, and European and US trademarks on the name FutureBIRTH™ (Serial No. 87/110459). Helen Zhou: Reports no related conflicts of interest. Howard S. Cuckle: Reports no related conflicts of interest. Argyro Synglaki: Reports no related conflicts of interest. Kypros Nicolaides: Reports no related conflicts of interest. Mark L. Weiss: Reports no related conflicts of interest. Yafeng Dong: Co-founder of Rosetta Signaling Laboratory, which controls the intellectual property related to plasma nucleic acid extraction and the RNA markers described including EU Patent No. 2646554, US patent No. 10,954,564, and European and US trademarks on the name FutureBIRTH™ (Serial No. 87/110459).

References

  1. Chawanpaiboon, S.; Vogel, J.P.; Moller, A.-B.; Lumbiganon, P.; Petzold, M.; Hogan, D.; Landoulsi, S.; Jampathong, N.; Kongwattanakul, K.; Laopaiboon, M.; et al. Global, regional, and national estimates of levels of preterm birth in 2014: A systematic review and modelling analysis. Lancet Glob. Health 2019, 7, e37–e46. [Google Scholar] [CrossRef] [Green Version]
  2. Kaempf, J.W.; Tomlinson, M.; Arduza, C.; Anderson, S.; Campbell, B.; Ferguson, L.A.; Zabari, M.; Stewart, V.T. Medical staff guidelines for periviability pregnancy counseling and medical treat. Pediatrics 2006, 117, 22–29. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Murray, C.J.L.; Vos, T.; Lozano, R.; Naghavi, M.; Flaxman, A.D.; Michaud, C.; Ezzati, M.; Shibuya, K.; Salomon, J.A.; Abdalla, S.; et al. Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990–2010: A systematic analysis for the Global Burden of Disease Study 2010. Lancet 2012, 380, 2197–2223. [Google Scholar] [CrossRef] [PubMed]
  4. Koullali, B.; Van Zijl, M.D.; Kazemier, B.M.; Oudijk, M.A.; Mol, B.W.J.; Pajkrt, E.; Ravelli, A.C.J. The association between parity and spontaneous preterm birth: A population based study. BMC Pregnancy Childbirth 2020, 20, 233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Smith, G.; Shah, I.; White, I.; Pell, J.; Crossley, J.; Dobbie, R.; Smith, G. Maternal and biochemical predictors of antepartum stillbirth among nulliparous women in relation to gestational age of fetal death. BJOG Int. J. Obstet. Gynaecol. 2007, 114, 705–714. [Google Scholar] [CrossRef]
  6. Van Der Ven, J.; Van Os, M.A.; Kazemier, B.M.; Kleinrouweler, E.; Verhoeven, C.J.; De Miranda, E.; van Wassenaer-Leemhuis, A.G.; Kuiper, P.N.; Porath, M.; Willekes, C.; et al. The capacity of mid-pregnancy cervical length to predict preterm birth in low-risk women: A national cohort study. Acta. Obs. Gynecol. Scand. 2015, 94, 1223–1234. [Google Scholar] [CrossRef] [Green Version]
  7. Rosenbloom, J.I.; Raghuraman, N.; Temming, L.A.; Stout, M.J.; Tuuli, M.G.; Dicke, J.M.; Macones, G.A.; Cahill, A.G. Predictive Value of Midtrimester Universal Cervical Length Screening Based on Parity. J. Ultrasound Med. 2020, 39, 147–154. [Google Scholar] [CrossRef]
  8. Esplin, M.S.; Elovitz, M.; Iams, J.D.; Parker, C.B.; Wapner, R.; Grobman, W.A.; Simhan, H.N.; Wing, D.A.; Haas, D.M.; Silver, R.M.; et al. Predictive Accuracy of Serial Transvaginal Cervical Lengths and Quantitative Vaginal Fetal Fibronectin Levels for Spontaneous Preterm Birth Among Nulliparous Women. JAMA 2017, 317, 1047–1056. [Google Scholar] [CrossRef] [PubMed]
  9. Weiner, C.P.; Dong, Y.; Zhou, H.; Cuckle, H.; Ramsey, R.; Egerman, R.; Buhimschi, I.; Buhimschi, C. Early pregnancy prediction of spontaneous preterm birth before 32 completed weeks of pregnancy using plasma RNA: Transcriptome discovery and initial validation of an RNA panel of markers. BJOG 2021, 128, 1870–1880. [Google Scholar] [CrossRef]
  10. Weiner, C.P.; Cuckle, H.; Weiss, M.L.; Buhimschi, I.A.; Dong, Y.; Zhou, H.; Ramsey, R.; Egerman, R.; Buhimschi, C.S. Evaluation of a Maternal Plasma RNA Panel Predicting Spontaneous Preterm Birth and Its Expansion to the Prediction of Preeclampsia. Diagnostics 2022, 12, 1327. [Google Scholar] [CrossRef]
  11. Mandrekar, I.N. Biostatistics for Clinicians: Receiver Operating Characteristic Curve in Diagnostic Test Assessment. J. Thoracic. Onc. 2010, 5, 1315–1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. El Khouli, R.H.; Macura, K.J.; Barker, P.B.; Habba, M.R.; Jacobs, M.A.; Bluemke, D.A. Relationship of temporal resolution to diagnostic performance for dynamic contrast enhanced MRI of the breast. JMRI 2009, 30, 999–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Lu, H.; Xu, Y.; Ye, M.; Yan, K.; Gao, Z.; Jin, Q. Learning misclassification costs for imbalanced classification on gene expression data. BMC Bioinformatics. 2019, 20, 681. [Google Scholar] [CrossRef] [PubMed]
  14. Burnham, K.P.; Anderson, D.R. Model Selection and Multimodel Inference: A practical Information-Theoretic Approach, 2nd ed.; Springer: Berlin/Heidelberg, Germany, 2002. [Google Scholar]
  15. Luo, W.; Huning, E.Y.-S.; Tran, T.; Phung, D.; Venkatesh, S. Screening for post 32-week preterm birth risk: How helpful is routine perinatal data collection? Heliyon 2016, 2, e00119. [Google Scholar] [CrossRef] [Green Version]
  16. Dude, C.M.; Levine, L.D.; Schwartz, N. The Impact of Previous Obstetric History on the Risk of Spontaneous Preterm Birth in Women with a Sonographic Short Cervix. Am. J. Perinatol. 2020, 37, 1189–1194. [Google Scholar] [CrossRef]
  17. Markenson, G.R.; Saade, G.R.; Laurent, L.C.; Heyborne, K.D.; Coonrod, D.V.; Schoen, C.N.; Baxter, J.K.; Haas, D.M.; Longo, S.; Grobman, W.A.; et al. Performance of a proteomic preterm delivery predictor in a large inde-pendent prospective cohort. Am. J. Obstet. Gynecol. MFM 2020, 2, 100140. [Google Scholar] [CrossRef]
  18. Ngo, T.T.M.; Moufarrej, M.N.; Rasmussen, M.-L.H.; Camunas-Soler, J.; Pan, W.; Okamoto, J.; Neff, N.F.; Liu, K.; Wong, R.J.; Downes, K.; et al. Noninvasive blood tests for fetal development predict gestational age and preterm delivery. Science 2018, 360, 1133–1136. [Google Scholar] [CrossRef] [Green Version]
  19. Jehan, F.; Sazawal, S.; Baqui, A.H.; Nisar, M.I.; Dhingra, U.; Khanam, R.; Ilyas, M.; Dutta, A.; Mitra, D.K.; Mehmood, U.; et al. Multiomics Characterization of Preterm Birth in Low- and Middle-Income Countries. JAMA Netw. Open 2020, 3, e2029655. [Google Scholar] [CrossRef]
  20. Camunas-Soler, J.; Gee, E.P.; Reddy, M.; Mi, J.D.; Thao, M.; Brundage, T.; Siddiqui, F.; Hezelgrave, N.L.; Shennan, A.H.; Namsaraev, E.; et al. Predictive RNA profiles for early and very early spontaneous preterm birth. Am. J. Obstet. Gynecol. 2022, 227, 72.e1–72.e16. [Google Scholar] [CrossRef]
  21. Cook, J.; Bennett, P.R.; Kim, S.H.; Teoh, T.G.; Sykes, L.; Kindinger, L.M.; Garrett, A.; Binkhamis, R.; MacIntyre, D.A.; Terzidou, V. First Trimester Circulating MicroRNA Biomarkers Predictive of Subsequent Preterm Delivery and Cervical Shortening. Sci. Rep. 2019, 9, 5861. [Google Scholar] [CrossRef] [Green Version]
  22. Hromadnikova, I.; Kotlabova, K.; Krofta, L. First Trimester Prediction of Preterm Delivery in the Absence of Other Pregnan-cy-Related Complications Using Cardiovascular-Disease Associated MicroRNA Biomarkers. Int. J. Mol. Sci. 2022, 23, 3951. [Google Scholar] [CrossRef]
  23. Del Vecchio, G.; Li, Q.; Li, W.; Thamotharan, S.; Tosevska, A.; Morselli, M.; Sung, K.; Janzen, C.; Zhou, X.; Pellegrini, M.; et al. Cell-free DNA Methylation and Transcriptomic Signature Prediction of Pregnancies with Adverse Outcomes. Epigenetics 2021, 16, 642–661. [Google Scholar] [CrossRef]
  24. Jain, V.; McDonald, S.D.; Mundle, W.R.; Farine, D. Guideline No. 398: Progesterone for Prevention of Spontaneous Preterm Birth. J. Obstet. Gynaecol. Can. 2020, 42, 806–812. [Google Scholar] [CrossRef]
  25. National Guideline Alliance Hosted by the Royal College of Obstetricians and Gynaecologists (UK). Preterm Labour and Birth: [A] Evidence Review for Clinical Effectiveness of Prophylactic Progesterone in Preventing Preterm Labour; National Institute for Health and Care Excellence (UK): London, UK, 2019. [Google Scholar]
  26. Norman, J.E. Progesterone and preterm birth. Int. J. Gynaecol. Obstet. 2020, 150, 24–30. [Google Scholar] [CrossRef]
  27. Poon, L.C.; Wright, D.; Rolnik, D.L.; Syngelaki, A.; Delgado, J.L.; Tsokaki, T.; Leipold, G.; Akolekar, R.; Shearing, S.; De Stefani, L.; et al. Aspirin for Evidence-Based Preeclampsia Prevention trial: Effect of aspirin in prevention of preterm preeclampsia in subgroups of women according to their characteristics and medical and obstetrical history. Am. J. Obstet. Gynecol. 2017, 217, 585.e1–585.e5. [Google Scholar] [CrossRef] [Green Version]
  28. Carlson, S.E.; Gajewski, B.J.; Valentine, C.J.; Kerling, E.H.; Weiner, C.P.; Cackovic, M.; Buhimschi, C.S.; Rogers, L.K.; Sands, S.A.; Brown, A.R.; et al. Higher dose docosahexaenoic acid supplementation during pregnancy reduces early preterm birth: A randomised, double-blind, adaptive-design superiority trial. EClinicalMedicine 2021, 36, 100905. [Google Scholar] [CrossRef]
  29. Sun, L.; Li, Y.; Xie, W.; Xue, X. Association between omega-3 fatty acid supplementation and lower risk of preterm delivery: A systematic review and meta-analysis. J. Matern. Neonatal Med. 2022, 35, 2294–2303. [Google Scholar] [CrossRef] [PubMed]
  30. Simmonds, L.A.; Sullivan, T.R.; Skubisz, M.; Middleton, P.F.; Best, K.P.; Yelland, L.N.; Quinlivan, J.; Zhou, S.J.; Liu, G.; McPhee, A.J.; et al. Omega-3 fatty acid supplementation in pregnancy—Baseline omega-3 status and early preterm birth: Exploratory analysis of a randomised controlled trial. BJOG: Int. J. Obstet. Gynaecol. 2020, 127, 975–981. [Google Scholar] [CrossRef] [PubMed]
  31. Lee, R.H.; Weiner, C.P. Robust Cost Efficacy of a Novel, Validated Screening Test at 12 to 20 Weeks for the Prediction Preterm Birth (PTB) ≦ 32 Weeks in Singletons. Gynecol. Obstet. Clin. Medicine 2021, 1, 107–111. [Google Scholar] [CrossRef]
  32. Arroyo, J.D.; Chevillet, J.R.; Kroh, E.M.; Ruf, I.K.; Pritchard, C.C.; Gibson, D.F.; Mitchell, P.S.; Bennett, C.F.; Pogosova-Agadjanyan, E.L.; Stirewalt, D.L.; et al. Argonaute2 complexes carry a population of circulating microRNAs independent of vesicles in human plasma. Proc. Natl. Acad. Sci. USA 2011, 108, 5003–5008. [Google Scholar] [CrossRef] [Green Version]
  33. Vickers, K.C.; Palmisano, B.T.; Shoucri, B.M.; Shamburek, R.D.; Remaley, A.T. MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat. Cell Biol. 2011, 13, 423–433. [Google Scholar] [CrossRef] [Green Version]
  34. Yoffe, L.; Gilam, A.; Yaron, O.; Polsky, A.; Farberov, L.; Syngelaki, A.; Nicolaides, K.; Hod, M.; Shomron, N. Early Detection of Preeclampsia Using Circulating Small non-coding RNA. Sci. Rep. 2018, 8, 3401. [Google Scholar] [CrossRef] [Green Version]
  35. Filant, J.; Nejad, P.; Paul, A.; Simonson, B.; Srinivasan, S.; Zhang, X.; Balaj, L.; Das, S.; Gandhi, R.; Laurent, L.C.; et al. Isolation of Extracellular RNA from Serum/Plasma. Methods Mol Biol. 2018, 1740, 43–57. [Google Scholar] [CrossRef] [PubMed]
  36. Goldenberg, R.L.; Culhane, J.F.; Iams, J.D.; Romero, R. Epidemiology and causes of preterm birth. Lancet 2008, 371, 75–84. [Google Scholar] [CrossRef] [PubMed]
  37. Esplin, M.S.; O’Brien, E.; Fraser, A.; Kerber, R.A.; Clark, E.; Simonsen, S.E.; Holmgren, C.; Mineau, G.P.; Varner, M.W. Estimating Recurrence of Spontaneous Preterm Delivery. Obstet. Gynecol. 2008, 112, 516–523. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Expression Levels in Plasma Cell-Free PSME2 and APOA1—cases and controls. Boxplots showing differentially expressed RNAs, e.g., PSME2 (with maternal age), APOA1, and APOA1 MoM. The boxes indicate the median and first and third quartiles, the lines demark the 10th and 90th percentile of the distributions, and the notches in the bars indicate median ±1.57 * interquartile range/Sqrt (n). The overlying circles indicate individual patient’s data, and these circles are coded by race. Nonoverlapping notches are “strong evidence” the medians differ. The folding of the box back towards the notch indicates a skewed distribution. Any sample outside whiskers might be an outlier. Footnote: Graphs created in R using ggplot2 library and the geom_boxplot() method (contact authors for the script).
Figure 1. Expression Levels in Plasma Cell-Free PSME2 and APOA1—cases and controls. Boxplots showing differentially expressed RNAs, e.g., PSME2 (with maternal age), APOA1, and APOA1 MoM. The boxes indicate the median and first and third quartiles, the lines demark the 10th and 90th percentile of the distributions, and the notches in the bars indicate median ±1.57 * interquartile range/Sqrt (n). The overlying circles indicate individual patient’s data, and these circles are coded by race. Nonoverlapping notches are “strong evidence” the medians differ. The folding of the box back towards the notch indicates a skewed distribution. Any sample outside whiskers might be an outlier. Footnote: Graphs created in R using ggplot2 library and the geom_boxplot() method (contact authors for the script).
Biomedicines 11 01149 g001
Figure 2. Dawn of Obstetric Personalized Medicine. Women may soon have the opportunity to choose screening based on a single plasma sample of cell-free RNA as early as the first trimester using a group of already validated tests and then initiate targeted therapy as desired.
Figure 2. Dawn of Obstetric Personalized Medicine. Women may soon have the opportunity to choose screening based on a single plasma sample of cell-free RNA as early as the first trimester using a group of already validated tests and then initiate targeted therapy as desired.
Biomedicines 11 01149 g002
Table 1. Performance of the Differentially Expressed RNAs in First Trimester Nullipara.
Table 1. Performance of the Differentially Expressed RNAs in First Trimester Nullipara.
12-Week RNA PanelAUCp *95% CIDetection Rates of sPTB ≤ 32 Weeks
10% FPR20% FPR30% FPR
PSME20.650.050.50–0.80183563
PSME2 (MoMs)0.650.060.50–0.80183864
PSME2 + MA0.69<0.020.54–0.83264555
PSME2 (MoMs) + MA0.67<0.020.53–0.82244058
PSME2 + CRL, MA, MW, race, tobacco0.71<0.0050.57–0.85323958
PSME2 (MoMs) + CRL, MA, MW, race, tobacco0.70<0.0050.56–0.84323955
APOA10.72<0.0010.59–0.85455065
APOA1 (MoMs)0.73<0.0010.60–0.87455065
APOA1 + MA0.77<0.00010.64–0.90536970
APOA1 (MoMs) + MA0.79<0.00010.66–0.91526175
APOA1 + CRL, weight, race, tobacco, MA0.77<0.00010.64–0.90487275
APOA1 (MoMs) + CRL, MA, MW, race, tobacco0.79<0.00010.66–0.91416179
PSME2 and APOA10.72<0.0020.58–0.86415059
PSME2 and APOA1 (MoMs)0.73<0.0020.59–0.87404861
PSME2 and APOA1 + MA0.78<0.00010.65–0.90506474
PSME2 and APOA1 (MoMs) + MA0.78<0.00010.66–0.91456578
PSME2 and APOA1 + CRL, MA, MW, race, tobacco0.78<0.00010.65–0.90486672
PSME2 and APOA1 (MoMs) + CRL, MA, MW, race, tobacco0.78<0.00010.66–0.91416178
Key: AUC—area under the curve; FPR—fixed false positive rate; MA—maternal age; MW—maternal weight; CRL—crown–rump length; MoM—multiple of the median; tobacco—tobacco use. * Compared with 0.50.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Weiner, C.P.; Zhou, H.; Cuckle, H.; Syngelaki, A.; Nicolaides, K.H.; Weiss, M.L.; Dong, Y. Maternal Plasma RNA in First Trimester Nullipara for the Prediction of Spontaneous Preterm Birth ≤ 32 Weeks: Validation Study. Biomedicines 2023, 11, 1149. https://doi.org/10.3390/biomedicines11041149

AMA Style

Weiner CP, Zhou H, Cuckle H, Syngelaki A, Nicolaides KH, Weiss ML, Dong Y. Maternal Plasma RNA in First Trimester Nullipara for the Prediction of Spontaneous Preterm Birth ≤ 32 Weeks: Validation Study. Biomedicines. 2023; 11(4):1149. https://doi.org/10.3390/biomedicines11041149

Chicago/Turabian Style

Weiner, Carl P., Helen Zhou, Howard Cuckle, Argyro Syngelaki, Kypros H. Nicolaides, Mark L. Weiss, and Yafeng Dong. 2023. "Maternal Plasma RNA in First Trimester Nullipara for the Prediction of Spontaneous Preterm Birth ≤ 32 Weeks: Validation Study" Biomedicines 11, no. 4: 1149. https://doi.org/10.3390/biomedicines11041149

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop