Abstract
In brief
Preterm birth is the leading cause of perinatal morbidity and mortality; however, current therapies offer limited efficacy to delay birth and improve neonatal outcomes. This review explores the potential of repurposing drugs with known safety profiles to quench uterine contractions and inflammation, identifying promising agents for clinical trials.
Abstract
Preterm birth is the leading cause of neonatal morbidity and mortality globally. Despite extensive research into the underlying pathophysiology, rates of preterm birth have not significantly reduced. Currently, preterm labour management is based on optimising neonatal outcomes. Treatment involves administering drugs (tocolytics) to suppress uterine contractions to allow sufficient time for transfer to an appropriate facility and administration of antenatal corticosteroids for fetal lung maturation. Current tocolytics are limited as they are associated with adverse maternal and fetal effects and only delay delivery for a short period. There has been a serious lack of therapeutic development for preterm birth, and new approaches to protect against or delay preterm birth are urgently needed. Repurposing drugs for the prevention of preterm birth presents as a promising approach by reducing the time and costs associated with pharmaceutical drug development. In this review, we explore the evidence for the potential of therapies, specifically proton pump inhibitors, tumour necrosis factor inhibitors, prostaglandin receptor antagonists, aspirin, and statins, to be repurposed as preventatives and/or treatments for preterm birth. Importantly, many of these innovative approaches being explored have good safety profiles in pregnancy. We also review how delivery of these drugs can be enhanced, either through targeted delivery systems or via combination therapy approaches. We aim to present innovative strategies capable of targeting multiple aspects of the complex pathophysiology that underlie preterm birth. There is an urgent unmet need for preterm birth therapeutic development, and these strategies hold great promise for improving neonatal outcomes.
Introduction
Preterm birth is defined as delivery before 37 completed weeks of gestation (World Health Organisation 2015). It is the leading cause of neonatal morbidity and mortality globally and is considered the most important determinant of poor outcomes for babies, in terms of both survival and long-term quality of life (World Health Organisation 2015). The most recent global data show that 15.22 million babies were born preterm in 2019, accounting for 0.66 million neonatal deaths (Cao et al. 2022). Despite extensive research and implementation of improved perinatal care, the overall rates of prematurity are not significantly reducing. While there was a small decline in preterm birth rates between 2019 and 2020 from 10.2% to 10.1% in the United States, the rates remain high particularly among non-Hispanic Black women where the rate was almost double compared to the rate among non-Hispanic white women (14.4% compared with 9.1%, respectively) (Hamilton et al. 2021).
The majority of preterm births occur spontaneously, without indication, but can also be initiated by the healthcare team based on medical complications of the pregnant patient, placenta, or fetus (overview in Fig. 1) (Vogel et al. 2018). Spontaneous preterm birth can be caused by cervical incompetency (premature dilation/ripening of cervix), preterm pre-labour rupture of membranes (PPROM), or spontaneous labour (Fig. 1). Cervical incompetency, characterised by a short cervix at mid-trimester (cervical length <25mm before 24 weeks), is a useful predictive marker for preterm birth risk, although it occurs at a low incidence, which limits the utility of screening (Glover & Manuck 2018). Risk factors for spontaneous preterm birth include multiple gestation, history of preterm birth, short cervix length, smoking, low socioeconomic status, and maternal age (World Health Organisation 2015, Glover & Manuck 2018). Maternal medical conditions during pregnancy such as diabetes, high blood pressure, or other pregnancy complications also increase the risk of preterm labour. Addressing modifiable risk factors can reduce the risk of preterm birth and should be implemented during care. However, it needs to be noted that many preterm births occur without these risk factors. The idiopathic nature of spontaneous preterm labour undoubtedly contributes to our resounding inability to identify pregnancies at high risk of preterm labour.
Even with increased surveillance and tailored treatment for those at high risk, there is relatively little that can be done to mitigate preterm birth, in terms of not only preventing pregnancies from going into preterm labour but also delaying birth for patients in established preterm labour. In preterm labour, management is based on optimising neonatal outcomes. Tocolytics are primarily used to allow sufficient time for transfer to an appropriate tertiary facility and administration of corticosteroids for fetal lung maturation and magnesium sulphate for fetal neuroprotection. However, therapeutics that can safely prolong gestation for longer periods are urgently needed.
This review will discuss the current therapies in use for the management of preterm labour and prevention of preterm birth and will review the evidence that exists for emerging novel therapeutics. The focus will be on therapies that have a known safety profile in pregnancy and could be repurposed for their potential tocolytic properties.
Pathophysiology of preterm birth
The processes and pathways in both normal term and preterm labour are not completely understood. However, it is widely accepted that the onset of labour is characterised by the switch of the uterus from a quiescent to contractile state accompanied by a shift from anti-inflammatory to pro-inflammatory signalling. The three main components of the common pathway of parturition are (i) an increase in myometrial contractility, (ii) remodelling and ripening of the cervix, and (iii) activation and weakening of the fetal membranes and decidua (Gotsch et al. 2009). These processes occur physiologically at term labour via inflammatory processes, but preterm labour is driven by a pathological level of inflammation, with inflammatory responses occurring at a heightened magnitude (Romero et al. 2006, Gotsch et al. 2009).
The inflammatory response in term and preterm labour is characterised by the expression and release of pro-inflammatory cytokines (i.e. interleukin (IL)1B, IL6, and tumour necrosis factor (TNF)), chemokines (CXC motif chemokine ligand-8 and monocyte chemoattractant protein-1), and prostaglandin (PG) E2 and PGF2a by the myometrium and the fetal membranes and is enhanced by infiltrating leukocytes to these tissues (Sivarajasingam et al. 2016). Progesterone is key in maintaining quiescence by repressing the expression of these chemokines, cytokines, and contraction-associated proteins such as oxytocin receptors, gap junction protein connexin-43, and PG receptors (Wu & DeMayo 2017). Immune cells, particularly macrophages and T-regulatory cells, are also important in maintaining maternal–fetal immune homeostasis (Gomez-Lopez et al. 2021).
The myometrium is the muscular layer of the uterus, primarily composed of smooth muscle cells. Myometrial contractility is based on the interaction between myosin and actin in myometrial smooth muscle cells, driven by calcium-dependent myosin light-chain kinase activity (Lopez Bernal 2003). The key mediators of myometrial activation are cytokines, PGs, and oxytocin, which modulate myometrial contractions through their downstream signalling (Fig. 2 and 3). The downstream effect is increased calcium mobilisation within myometrial cells, leading to increased myometrial contractility (Gotsch et al. 2009).
Cervix remodelling – encompassed by cervical softening, ripening, and dilation – is driven by pro-inflammatory cytokines through increased expression of matrix metalloproteinases (MMPs), cyclo-oxygenase-2 (COX-2), and PGE2, leading to softening and dilation of the cervix (Holt et al. 2011). Similarly, increases in MMPs, collagenases, and PGs and reductions in tissue MMP inhibitors result in the activation, weakening, and rupture of fetal membranes (Fig. 2). These processes are highly controlled during term labour, and investigation of how these processes may differ in preterm labour is paramount.
Infection-driven and sterile inflammation in preterm birth
Preterm birth is considered a syndrome involving multiple causal and associated aetiologies, but the most well-established cause is inflammation of the amniotic cavity known as ‘intra-amniotic inflammation’ (Romero et al. 2006, 2014, Goldenberg et al. 2008). Intra-amniotic inflammation can occur in the absence of microbes, known as sterile inflammation, or be driven by infection (chorioamnionitis). The established and novel mechanisms of the immunobiology of preterm labour and birth are detailed in a recent review by Gomez-Lopez and colleagues (Gomez-Lopez et al. 2022). Briefly, sterile inflammation is triggered when danger signals called damage-associated molecular patterns (DAMPs) activate pattern-recognition receptors in the myometrium or fetal membranes to induce an acute innate immune response. DAMPs are endogenous molecules released because of cellular necrosis, stress, or ischaemia (Nadeau-Vallee et al. 2016). This is characterised by elevated inflammatory mediators including IL6 and infiltration of leukocytes (neutrophils, monocytes, macrophages, and T cells) into gestational tissues. Intra-amniotic infections are typically associated with bacterial microorganisms ascending from the lower genital tract and are strongly associated with spontaneous preterm labour (Romero et al. 2001). Some infections can be subclinical, making it difficult to detect and diagnose (Romero et al. 2001). In cases of infection during pregnancy, pathogens are recognised by their microbial molecules called pathogen-associated molecular patterns (PAMPs) by pattern-recognition receptors such as toll-like receptors on gestational tissues and stimulate the release of pro-inflammatory chemokines and cytokines. This leads to increases in PG and MMP production which ultimately activates the parturition pathways, as described above (Romero et al. 2001). While antibiotics are used for intrauterine and systemic infections during pregnancy, they are inefficient in preventing preterm birth in cases of preterm labour with intact membranes (Olson et al. 2008), suggesting labour induced by infection arises via inflammatory pathways that cannot be slowed by treating the infection (Nadeau-Vallee et al. 2016). Therefore, specifically treating the inflammation that precedes and causes preterm birth presents as a very promising target for intervention.
Current clinical preterm birth therapeutics (tocolytics)
Tocolytics are drugs used to delay preterm delivery by relaxing uterine myometrial contractions. They are used to prolong pregnancy to provide time to administer antenatal corticosteroids to accelerate fetal lung maturation and transfer patients to a specialised neonatal unit. Both actions have been shown to reduce neonatal morbidity and mortality (Vogel et al. 2018). However, there is no strong evidence that any tocolytic improves neonatal outcomes such as respiratory distress syndrome or death (Haas et al. 2012). Tocolytics are primarily used in cases of spontaneous preterm labour. The use of tocolysis in PPROM is not completely supported due to insufficient evidence to suggest a beneficial role, and any benefits must be weighed against the potential harm associated, particularly to the fetus (Mackeen et al. 2014). Therefore, this review will focus on tocolysis for spontaneous preterm labour with intact membranes.
There are several types of tocolytics which vary in mechanism, cost, safety profile and effectiveness (Haas et al. 2012, Flenady et al. 2014a,b, Reinebrant et al. 2015). Availability varies between countries due to differences in regulatory approvals. The regulatory challenges for preterm birth therapeutic development are discussed in detail in a focused review (Coler et al. 2021).
Calcium channel blockers
Calcium channel blockers (CCBs), such as nifedipine and nicardipine, are conventionally prescribed as anti-hypertensive agents; however, they were investigated and repurposed as tocolytics in the 1980s due to their potent relaxant effect on smooth muscle contractions (Ulmsten et al. 1980). Today, due to significant challenges for development of obstetric medications, including lengthy delays in regulatory approval, these CCBs continue to be used off-label for the treatment of preterm labour. Nifedipine is often considered as the first-line tocolytic therapy for the management of spontaneous preterm labour.
CCBs are non-specific smooth muscle relaxants. They bind to L-type voltage-gated calcium channels in the myometrial cell plasma membrane and inhibit the inward flux of calcium ions required for muscle contractions (Fig. 3). Nifedipine is widely used due to its low cost, availability, favourable safety profile, and capacity for oral administration. Nicardipine has the additional advantage of both oral and i.v. administration; however, i.v. nicardipine has not been shown to be more beneficial in prolonging the duration of pregnancy in comparison to oral nifedipine (Le Ray et al. 2010).
Nifedipine has less adverse effects compared to many other tocolytics (Flenady et al. 2014b). The side effects of nifedipine, including headaches, tachycardia, anxiety, vomiting, and hypotension, are relatively well tolerated and manageable (Flenady et al. 2014b). Additionally, nifedipine does not appear to adversely affect the fetus: not altering fetal movement, heart rate, or blood flow (de Heus et al. 2009).
The latest Cochrane review on the use of CCBs (primarily nifedipine) as tocolytic therapies showed there is insufficient evidence regarding efficacy to improve perinatal mortality or rate of preterm birth (Flenady et al. 2014b). The review included 38 trials of 3550 patients; nifedipine was assessed in 35 of these trials and nicardipine in three. In the two studies that compared nifedipine to placebo or no treatment, there was a delay in delivery and a reduction in birth less than 48 h after trial entry with nifedipine use (Zhang & Liu 2002, Ara & Banu 2008). This supports the use of nifedipine to prolong pregnancies in the short term, to provide time for corticosteroid administration and to transfer to higher level care. However, in terms of the outcome of reducing the incidence of preterm birth, the two trials found conflicting results. The placebo-controlled trial showed nifedipine was no different to placebo (Ara & Banu 2008), while the non-placebo controlled trial showed nifedipine reduced the occurrence of preterm birth (Zhang & Liu 2002). These different findings may be attributable to the fundamental differences between the two trials, particularly regarding doses and blinding to the intervention. Well-designed trials with appropriate primary outcomes and blinding would be needed to truly resolve the contention, and further investigation is needed.
Of note, while nifedipine is primarily used to arrest preterm labour (acute tocolysis), it can also be used to subsequently maintain uterine quiescence (maintenance tocolysis). However, there is very limited evidence supporting the use of nifedipine for maintenance therapy (Naik Gaunekar et al. 2013, Ding et al. 2016) and therefore is not recommended. Thus, although nifedipine is often considered the standard course of treatment for preterm labour, the benefits in improving neonatal survival are minimal, and further investigation is warranted to elucidate why these therapies are limited and how we can improve the development of acute tocolytic therapies.
Oxytocin receptor antagonists
Atosiban is an i.v. medication that acts as an oxytocin receptor antagonist. Unlike other tocolytic agents, atosiban has been developed exclusively for the management of preterm labour. Despite this, it has not been approved by the United States Food and Drug Administration (FDA) but is available for clinical use in Europe.
Atosiban is a peptide analogue of oxytocin and competitively inhibits oxytocin from binding to its membrane-bound receptors in the myometrium and decidua. This prevents the signalling required for the release of intracellular calcium and thus inhibits myometrial contractions (Fig. 3). Atosiban should have limited systemic maternal effects as oxytocin receptors are only highly expressed in the uterus and myoepithelial cells in mammary glands (Fuchs et al. 1984).
As demonstrated in trials, atosiban is associated with fewer maternal adverse effects compared with other tocolytics and therefore is often the preferred tocolytic in high-resource settings but is limited in low-resource settings due to its high cost (Coler et al. 2021). Atosiban can cross the placenta to the fetus but does not have any adverse effects on fetal heart rate, fetal movement, or fetal blood flow (de Heus et al. 2009). Atosiban is currently the only oxytocin receptor antagonist used clinically. Barusiban was tested in phase II clinical trials but did not show any difference to placebo in terms of reducing uterine contractions or proportion of patients delivering within 48 h (Thornton et al. 2009). As atosiban and barusiban are peptide antagonists, their oral bioavailability is poor due to degradation and limited absorption of peptides in the gastrointestinal tract. To counter this issue, non-peptides with high oxytocin receptor selectivity such as retosiban are being investigated (Thornton et al. 2017). Three clinical trials assessing retosiban in the management of preterm birth were evaluated in a systematic review but did not show a significant advantage in prolonging pregnancies compared to placebo (Marchand et al. 2021). However, the favourable safety profile with minimal maternal and fetal adverse effects support further research into this class of drugs.
Betamimetics
Betamimetics, also known as beta-2 adrenergic receptor agonists, are a class of drugs that act on these receptors in myometrial cells. They reduce uterine contractions by binding to the beta-2 adrenergic receptors on the surface of myometrial smooth muscle cells, activating adenylyl cyclase to form cAMP (Fig. 3). In turn, elevated levels of cAMP lead to a decrease in myosin light-chain kinase activity. Therefore, the smooth muscle machinery – myosin and actin – are prevented from interacting to generate contractile force.
Betamimetics were investigated for repurposing as tocolytics in the 1960s after their development as anti-asthma medications. Terbutaline was used off-label (as with most tocolytics) under the US FDA classification of Category B (no evidence of risk in humans) until it was classified as Category C (risk cannot be ruled out) in 2011. While still used in some countries, the use of betamimetics such as terbutaline, ritodrine, and salbutamol has significantly dropped due to serious adverse maternal side effects (such as tachycardia, hyperglycaemia, and chest pain), leading to immediate withdrawal from treatment (Neilson et al. 2014). Additionally, betamimetics are not recommended for long-term tocolysis.
Betamimetics have not been shown to significantly reduce perinatal death compared with placebo (Neilson et al. 2014). While they demonstrate some efficacy in delaying delivery (Flenady et al. 2014a,b, Neilson et al. 2014), they are not often considered as a first-line therapy for preterm birth management due to serious adverse effects.
Non-steroidal anti-inflammatory drugs
Non-steroidal anti-inflammatory drugs (NSAIDs), including indomethacin, have also been utilised as tocolytics. NSAIDs work via inhibition of COX-1 and COX-2 (Fig. 3) (Vane 1971). COX enzymes are responsible for synthesis of PGs, and therefore, inhibition of COX activity leads to reduced PG production. PGs are important in the onset and maintenance of labour (Keirse 1992). During pregnancy, COX-1 and COX-2 are expressed by the myometrium, decidua, and chorioamnion, with significant up-regulation in the expression of COX-2 prior to the onset of labour at both term and preterm (Slater et al. 1999). This suggests that COX-2 mediates increased PG synthesis in the myometrium at term, highlighting the potential to specifically target and inhibit COX-2. PGs cause an increase in free intracellular calcium levels and amplify the activation of myosin light-chain kinase, which leads to muscle contraction (Molnar & Hertelendy 1990). Therefore, inhibiting PG production can directly reduce myometrial contractility (Van den Veyver & Moise 1993, Doret et al. 2002).
While indomethacin was originally shown to cause a reduction in preterm uterine contractions by up to 7 days (Zuckerman et al. 1974), it was subsequently found to be associated with maternal and fetal side effects when administered in later gestation. Indomethacin freely crosses the placenta and can interfere with PG homeostasis in the fetus, resulting in serious neonatal complications including premature closure of the ductus arteriosus or necrotising enterocolitis (Van den Veyver & Moise 1993, Sood et al. 2011). As a result, indomethacin is usually not administered in pregnancies after 32 weeks’ gestation.
Delaying preterm birth with current therapies: what is the consensus?
Clinicians are limited in the choices of effective preterm therapeutics to use. Cochrane reviews have highlighted that there is not any single tocolytic that can reliably and effectively prevent preterm birth. Rather, these current therapies only postpone the imminent birth by up to 48 h, which is not wholly advantageous for cases of early preterm labour. CCBs have demonstrated benefits over other tocolytics (Flenady et al. 2014b). In comparison to betamimetics, CCBs increased prolongation of pregnancy and importantly resulted in fewer maternal adverse effects and cases of severe neonatal morbidity. CCBs also demonstrated benefits compared to oxytocin receptor antagonists (such as atosiban) and magnesium sulphate in terms of briefly delaying birth, but maternal adverse effects were increased compared with atosiban (Flenady et al. 2014b). This finding was confirmed by another meta-analysis demonstrating that atosiban was associated with fewer maternal side effects than nifedipine (Ali et al. 2019). This study indicated there was no difference in the development of neonatal adverse effects between the two drugs (Ali et al. 2019). Interestingly, NSAIDs and CCBs were similar in their ability to briefly prolong pregnancies (Reinebrant et al. 2015). But, with concerns of use in the third trimester, NSAIDs are limited in their application in late preterm labour. Thus, there is no real consensus that has been reached as due consideration of gestation, location, resource settings, and clinical maternal characteristics impact the ultimate choice of therapy to pursue.
Repurposing drugs as novel therapeutics for the prevention of preterm birth
As discussed above, therapeutic development for preterm birth has been very slow. One approach in obstetric therapeutic development is to repurpose medications. Drug repurposing is the process of discovering new applications for existing drugs (Ashburn & Thor 2004). Traditional pharmaceutical development is expensive and time-consuming; repurposing drugs can fast-track the implementation of an existing intervention for a new indication (Ashburn & Thor 2004) by investigating approved drugs with established safety and pharmacokinetic profiles from extensive testing in clinical trials. This is clearly evident with the history of current tocolytics (as discussed above); CCBs, betamimetics, and indomethacin were all originally developed for cardiovascular conditions or pain. Carefully designed randomised clinical trials will be needed to assess whether some of these repurposed agents may also confer long-term protection to preterm labour or have fewer side effects than current therapies.
There are several important pro-inflammatory factors that are implicated in preterm birth pathophysiology, particularly IL1, IL6, and the TLR4 signalling pathways. There are existing biologics and drugs that target many of these key mediators that are being preclinically investigated for repurposing for treatment of preterm birth. IL1 receptor inhibitors rytvela and anakinra, TLR4 antagonist naloxone, and broad-spectrum chemokine inhibitors (mechanisms of action in Fig. 2) are reviewed in detail elsewhere (Robertson et al. 2020, Coler et al. 2021). For that reason, we have elected to discuss targeting other relevant pro-inflammatory signalling agents that have not been reviewed in detail recently. Further, we will focus on repurposed drug development for preterm birth, using therapies known to be safe during pregnancy that have shown potential as preterm therapies in preclinical studies (overviews in Fig. 2 and 3).
Targeting prostaglandin synthesis and action
As described earlier, NSAIDs, specifically indomethacin, have been used as tocolytics due to their ability to block PG production via inhibition of COX enzymes (Fig. 3). As these drugs act primarily as PG synthesis inhibitors, they are only targeting one aspect of inflammatory signalling that is activated in preterm labour. Additionally, the use of indomethacin has proven to be limited with serious neonatal complications associated with its use, as discussed earlier. However, aspirin, another NSAID, has gained interest as a preterm birth therapeutic.
Aspirin (acetylsalicylic acid) is used in the prevention and treatment of cardiovascular disease. Aspirin is an antiplatelet drug with anti-inflammatory properties, exerting its effects by non-selectively inhibiting COX-1, and to a lesser extent COX-2 enzymes in the cyclo-oxygenase pathway, in turn blocking their enzymatic conversion of arachidonic acid into PGs (Fig. 3) (Atallah et al. 2017). COX-1 is a constitutive enzyme that supports beneficial homeostatic functions, whereas COX-2 is an inducible enzyme, which is up-regulated by inflammatory mediators and increased around the time of labour (Slater et al. 1999).
Interest in aspirin as a potential therapy for preterm birth stemmed from research in preeclampsia prevention; meta-analyses and systematic reviews revealed that trials of prophylactic low-dose aspirin during pregnancy for the prevention of preeclampsia were also associated with reductions in preterm birth (Roberge et al. 2013, Henderson et al. 2014). Use of aspirin in pregnancy is generally considered safe and therefore has been proposed to be a preventative for preterm labour. However, aspirin can cause maternal gastrointestinal bleeding and has recently been shown to be associated with increased postpartum bleeding and postpartum hematoma (Hastie et al. 2021). In terms of fetal effects, aspirin can cross the placenta and reach the fetus, inhibiting fetal platelet aggregation (Leonhardt et al. 2003), and may be associated with neonatal intracranial haemorrhage (Hastie et al. 2021). As the side effects seem to be dose dependent, use of low-dose aspirin is being investigated for preterm birth prevention in randomised control trials (RCTs). Two large RCTs, the ASPIRIN study in nulliparous patients (Hoffman et al. 2020) and the subsequent APRIL study in patients with recurrent preterm birth (Landman et al. 2022), found conflicting results on whether low-dose aspirin has beneficial effects on preterm birth prevention. Specifically, the ASPIRIN trial showed a reduction in the incidence of preterm birth and a decrease in perinatal mortality with aspirin as a preventative, whereas the APRIL trial did not. However, it is important to note that the ASPIRIN trial did not distinguish between spontaneous and indicated (provider-initiated) preterm birth. The APRIL trial was focused on preventing preterm birth in a high-risk population with previous spontaneous preterm birth. Therefore, further investigation in both preclinical and clinical trials would be necessary before recommending aspirin as a preterm birth therapeutic.
Another approach for specific targeting of PGs is via the use of PG receptor antagonists. OBE-022 (ebopiprant) acts by targeting the PGF2a receptor (Fig. 3) and has recently emerged as a once-daily, oral, and selective therapy with promising results (Pohl et al. 2018a). Following success in phase I trials (Pohl et al. 2018b, 2019, Taubel et al. 2018), OBE-022 was advanced to the PROLONG trial, a phase IIa trial in pregnancies with spontaneous preterm labour. Proof-of-concept data were recently presented in an update on the PROLONG trial and demonstrated that OBE-022 was well tolerated and efficacious, particularly when combined with atosiban treatment (ObsEva 2021). The study is anticipated to advance to a phase IIb/III adaptive study. By blocking signalling of specific PG receptors (i.e. PGF2a), this therapy avoids the side effects associated with non-selectively inhibiting the formation of all forms of PGs which are required for fetal development (Pohl et al. 2018a). Thus, this more targeted approach theoretically avoids the serious consequences in the fetus that are associated with NSAIDs, and trial results are awaited with anticipation.
Tumour necrosis factor antagonists
TNF is a key pro-inflammatory cytokine that mediates the inflammatory cascades in both term and preterm labour and has been found to be elevated in amniotic fluid in preterm labour (Lyon et al. 2010). Additionally, TNF plays a role in cervical remodelling and membrane rupture, by promoting the production of MMPs in the cervix and amnion (So et al. 1992, Crider et al. 2005). Administration of TNF has been shown to induce preterm labour in murine and non-human primate models (Silver et al. 1997, Sadowsky et al. 2006). Therefore, TNF poses as an candidate target for intervention in preterm labour.
TNF antagonists (also referred to as anti-TNF drugs) are agents that neutralise TNF (Fig. 2), and each varies slightly in mechanism (Tracey et al. 2008). These drugs can be either fusion proteins (such as etanercept) or neutralising monoclonal antibodies (such as infliximab and adalimumab). They are typically administered for the treatment and management of autoimmune and chronic inflammatory conditions, such as rheumatoid arthritis, and can be continued during pregnancy. There are limited reports on the rate of preterm birth amongst people taking these drugs. In a population-based study, TNF antagonists were shown to be associated with increased risks of preterm birth, caesarean section, and small for gestational age babies (Broms et al. 2020). However, these findings may indicate an association related to the underlying chronic inflammatory disease rather than to the agents. In animal models, TNF antagonists have shown to have beneficial effects in reducing inflammatory markers, preterm birth rates, and fetal injury (Holmgren et al. 2008, Shin et al. 2019). In infection-induced (using lipopolysaccharide (LPS)) models of preterm birth in mice, pre-treatment of mice with TNF antagonists has shown conflicting results: Holmgren and colleagues (2008) showed a decrease in the rate of preterm birth and fetal death, while an earlier study by Fidel and colleagues (1997) did not. More recently, Burdet and colleagues demonstrated that etanercept reduced preterm delivery rates by 30% in rats (Burdet et al. 2013). These differences may be due to the differences between the antibodies or the models. Interestingly, inhibition of TNF did not decrease COX-2 expression which was up-regulated by LPS in the mouse model, indicating that TNF may not directly regulate COX-2 expression in response to LPS (Holmgren et al. 2008). This finding highlights the complexities in attempting to target and inhibit single mediators or pathways involved in preterm labour. However, it should be noted that targeted anti-inflammatory biologics such as TNF or IL1 receptor antagonists have advantages over NSAIDs with their specificity, which reduces off-target effects. Given the involvement of TNF in the inflammatory cascade of preterm birth, there is scope to further investigate the ability of currently available TNF inhibitors to reduce inflammation in human gestational tissues, but without trials in humans, the evidence remains limited.
Proton pump inhibitors
Proton pump inhibitors (PPIs) are conventionally used as treatments for gastroesophageal reflux disease. PPIs act on and inhibit the H+/K+ATPase enzymes, which control the acidification of the stomach. The expression of these enzymes has also been found in the uterus in rats although this has not yet been reported in humans (Crowson & Shull 1992, Jaisser et al. 1993). PPIs are considered safe to use in pregnancy, with studies of large numbers of pregnancies exposed to PPIs prior to conception and throughout pregnancy (>5000 exposures), indicating no increase in birth defects or adverse outcomes (Gill et al. 2009, Pasternak & Hviid 2010).
PPIs are currently under examination as a therapy for preeclampsia. Preclinically, our team demonstrated that PPIs have anti-inflammatory and cytoprotective actions on the dysfunctional placenta and endothelium, with additional vasorelaxant properties on whole vessels (Onda et al. 2017), as well as anti-hypertensive effects in mouse models of preeclampsia (Gu et al. 2022). The effectiveness of PPIs to prevent preeclampsia has been conflicting in clinical trials (Saleh et al. 2017, Cluver et al. 2018, Atwa et al. 2021, Neuman et al. 2022), and further investigation around timing, dosage, and route of administration is required.
Regardless of this fact, the PPIs have demonstrated properties that suggest they may be candidate preterm birth therapeutics. Emerging investigation in preclinical studies for this application is underway. An early study has shown that omeprazole can reduce spontaneous and calcium-induced myometrial contractions in in vitro models testing myometrium collected from pregnancy (Yildirim et al. 2001). More recently, a variety of PPIs were examined in human myometrial tissue bath experiments. Pantoprazole demonstrated the most effective relaxant action, greater than nifedipine and other tocolytics used as comparators (Terranova et al. 2014).
Lansoprazole was also uncovered as a strong potential therapeutic candidate for preterm birth via computational identification methods using transcriptomic data (Le et al. 2020). The authors further validated this finding with a pilot study of a mouse model of LPS-induced preterm birth, which showed reduction in fetal loss with lansoprazole treatment.
PPIs are hypothesised to have effects on the myometrium through pathways other than directly influencing the H+/K+ATPase. Whilst the specific mechanisms are not clear, it is thought that pantoprazole may interfere with the regulation of intracellular calcium concentrations (Fig. 3) (Terranova et al. 2014). In animal models, omeprazole reduced smooth muscle tone of the trachea (Rhoden et al. 1996) and lower oesophageal sphincter (Welsh et al. 2014). These papers suggest PPIs may exert muscle relaxant effects via inhibiting the Rho/Rho-associated protein kinase (ROCK) pathways (Fig. 3). The Rho/ROCK pathway in myometrial cells is known to be involved in uterine contractility, and inhibition of this pathway promotes muscle relaxation (Lartey & Lopez Bernal 2009). The ability of PPIs to potently reduce myometrial contractions and the safety profile of these drugs support the further investigation into repurposing PPIs as promising candidates for preterm labour suppression.
Statins
Statins are a class of drugs that potently inhibit cholesterol production and are used as a preventative/treatment for cardiovascular disease. They inhibit HMG-CoA reductase, the rate-limiting enzyme in the cholesterol biosynthesis pathway. However, statins have also received interest as candidate drugs for the treatment and prevention of preeclampsia. Importantly, statins have anti-inflammatory and cytoprotective properties and can inhibit vascular smooth muscle contractions (Perez-Guerrero et al. 2005). Pravastatin has been shown preclinically to have beneficial effects on placental and systemic vascular disease by up-regulating antioxidant pathways, reducing the production of anti-angiogenic factors and improving endothelial migration and adhesion (Brownfoot et al. 2016, de Alwis et al. 2020). For these reasons, pravastatin has been assessed in several clinical trials (Costantine et al. 2016, 2021, Ahmed et al. 2020).
Furthermore, these anti-inflammatory properties make the statins desirable candidates for the prevention and/or treatment of preterm birth and are now being examined in preclinical studies. Direct anti-inflammatory effects of pravastatin and simvastatin on pregnant mouse cervix and uterus were examined in an LPS-induced mouse model of preterm birth. Pre-treatment with either of these statins was associated with a decrease in the expression and production of pro-inflammatory cytokines in the cervix and uterus of mice (Basraon et al. 2012). Interestingly, simvastatin emerged as the stronger anti-inflammatory compared with pravastatin as it reduced a greater number of cytokines. The anti-inflammatory effects of simvastatin have been reiterated in another preclinical study (Boyle et al. 2019).
Further to anti-inflammatory actions, statins were demonstrated to prevent cervical remodelling and inhibit myometrial contractions in mouse and human tissues and prevent preterm birth in a mouse model (Gonzalez et al. 2014, Boyle et al. 2019). Gonzalez and colleagues (2014) proposed that statins may be exerting these actions by activation of the heme oxygenase/carbon monoxide (HO-1/CO) pathway. HO-1 has previously been reported to diminish myometrial contractility by increasing the production of CO in myometrial cells, which in turn acts as a messenger to activate soluble guanylate cyclase activity, thus increasing the production of cyclic GMP (Fig. 3) (Acevedo & Ahmed 1998). This leads to smooth muscle relaxation. Additionally, Boyle and colleagues (2019) demonstrated that simvastatin could inhibit the Rho/ROCK pathway which, as described earlier, is a pathway associated with muscle contraction (Fig. 3). In terms of preventing cervical remodelling, pravastatin achieved this by (i) decreasing MMP activity and (ii) inhibiting complement activation (Fig. 2) (Gonzalez et al. 2014). The regulation of complement activation by the statins in animal models of other pregnancy complications such as preeclampsia has been previously reported, thus highlighting that this may be a pathway worth targeting (Kumasawa et al. 2011, Singh et al. 2011).
Collectively, these preclinical findings have convincingly presented the statins as promising candidates. As such, pravastatin has been recently taken into the clinical trial phase. The PIPIN trial in the UK is investigating the feasibility of pravastatin as a treatment for preterm birth (Whitaker et al. 2019). So far, no adverse effects have been reported in the participants who received pravastatin.
Currently, statins are classified as Category X for use in pregnancy by the US FDA, meaning that ‘studies in animals or humans have demonstrated fetal abnormalities and the risks involved in use of the drug in pregnant women clearly outweigh potential benefits’, and the UK NICE guidelines advise to avoid their use in pregnancy. While not approved for therapeutic use in pregnancy yet, there is now substantial evidence (Zarek & Koren 2014) to refute the original study (Edison & Muenke 2004) that claimed that statins were teratogenic.
Further to this, as pravastatin is a hydrophilic drug, it should not pass easily through the placenta to the fetus (Hamelin & Turgeon 1998). This is supported with evidence that pravastatin is not detectable (Costantine et al. 2016), or at very low concentrations, in the umbilical cord at birth (Ahmed et al. 2020, Costantine et al. 2021). Moreover, clinical trials assessing pravastatin have not shown any adverse fetal effects (Costantine et al. 2021). Therefore, there is momentum with supporting evidence to indicate that the statins are safe in pregnancy or that any risks may outweigh the benefits of the treatment for preeclampsia or preterm birth.
Enhanced activity and efficacy – combination therapeutic approaches
Another approach to enhance therapeutics may be combination therapy approaches, whereby drugs with complementary actions that act via different mechanisms can be dually administered (Vogel et al. 2014). These drugs could have additive or synergistic effects. It is postulated that combination therapy can enhance the overall effectiveness of the treatment, reducing dosage and frequency, thus averting adverse effects (Vogel et al. 2014). The current tocolytic approach to suppress uterine contractions is unlikely to achieve more than delaying birth for a few days, unless the underlying inflammatory drivers, which can also cause fetal inflammatory injury, are additionally dampened. Therefore, when considering combination therapies, both inflammation and myometrial contractions should both be targeted. The combination of a CCB (to directly inhibit uterine contractions) and indomethacin (to reduce inflammation) is an example of this approach. A recent RCT found that combination therapy of nifedipine and the anti-inflammatory NSAID indomethacin between 26 and 34 weeks’ gestation was a more effective tocolytic therapy than monotherapy of either drug (Kashanian et al. 2020). Combination therapy of nifedipine and indomethacin increased the proportion of pregnancies where contractions were inhibited for 48 h and for 7 days. Additionally, gestational age at birth and neonatal weight were significantly increased by combination treatment compared with either drug alone. There is currently a single-hospital RCT being conducted with Sudanese participants diagnosed with threatened preterm labour between 25 and 34 weeks investigating the nifedipine/indomethacin combination compared with nifedipine monotherapy (Ibrahim et al. 2021). Optimistically, further well-designed trials will reveal any clear advantages for combination therapy. Synergistic effects between therapies should also be investigated in preclinical models of preterm labour, to determine how the targeted pathways may interact and whether adverse impacts on the fetus may result. Importantly in cases of infection, or suspicion of infection, antibiotics will also need to be administered, and therefore any contraindications need to be considered to ensure drug activity is not disrupted.
Targeted drug delivery to enhance therapeutic action on myometrial contractions
While many of these repurposed therapies demonstrate promise, potential off-target effects are still of concern when administering any drug in pregnancy, as they may inadvertently harm the developing fetus, as was seen in the Dutch STRIDER trial with sildenafil citrate (Groom et al. 2018). However, targeted delivery systems such as the use of advanced targeted nanoparticles may prevent these off-target effects by allowing delivery of therapeutics to specific cells, organs, or tissues (Pritchard et al. 2021). Targeted myometrial drug delivery using nanoparticles has recently been reviewed in thorough detail (Coler et al. 2021, Pritchard et al. 2021) and will be briefly reviewed here. Tocolytics can be delivered in nanocarrier systems such as nanoliposomes. Refuerzo and colleagues (2015) first examined the selectivity and efficacy of indomethacin packaged in liposomes. Encapsulated indomethacin resulted in a 7.6-fold reduction in the amount of drug that transferred to the fetus compared with systemic administration. However, the encapsulated drug did not differ in efficacy compared with the non-packaged drug (Refuerzo et al. 2015). To improve specificity, targeted liposomes were developed. The oxytocin receptor is highly expressed by myometrial cells during pregnancy and is specific to the uterus and breast tissue (Fuchs et al. 1984), thus providing a means of targeting therapeutics to the myometrium. In a subsequent study, the addition of an oxytocin receptor antagonist to the liposomal surface increased selective targeting to the uterus and resulted in a 15% reduction in preterm birth in a mouse model (Refuerzo et al. 2016). The benefit of targeting to the oxytocin receptor to deliver known tocolytics, including nifedipine, salbutamol, and indomethacin, has also been shown (Paul et al. 2017). This study elegantly demonstrated the specificity of the liposomes in binding to the oxytocin receptor in mouse models of preterm birth. In pregnant mice, transplacental passage of the liposomes to the fetus was not observed, but there was some localisation to the mammary tissue (Paul et al. 2017). So far, nanoparticle-based targeted delivery systems for preterm birth have not reached clinical trials; however, the preclinical data are very promising and the flexibility to deliver different classes of drugs makes this delivery system an exciting innovation.
Future perspectives
Repurposing existing drugs brings significant advantages to the therapeutic development pipeline. The known safety profiles, established mechanisms, and previous extensive testing make it quicker, cheaper, and easier to implement these therapies in trials. However, it must be considered that this approach is limited to existing and available drugs. The cost of therapies must also be considered in therapeutic development, as low- and middle-income populations are where preterm birth rates are highest.
A multifaceted approach is essential for therapeutic development, where human in vitro work, animal models, computational analysis, and evidence from epidemiological studies can inform each other and guide which drugs to investigate further in clinical trials. In addition, clinical trials must be carefully designed to maximise their impact. Meta-analyses of preterm birth tocolytics continually call for better designed trials and standardised reporting of outcomes, and this must be adopted by the field.
Innovation in therapeutic development for preterm birth will involve a combination of repurposing therapies (e.g. proton pump inhibitors (PPIs), statins, and aspirin), targeted delivery approaches (e.g. nanoliposome encapsulation), and inhibition of specific key pathways (e.g. TNF inhibitors and PG receptor antagonists). The potential that some of these drugs have shown in clinical trials for the prevention of preeclampsia is exciting for the area of preterm birth therapies. We propose that efforts should be focused on therapies that target both the underlying inflammation and downstream myometrial contractions.
Conclusion
The rates of preterm birth remain high, and despite research focused towards understanding the pathophysiology of preterm birth and development of new therapeutics, there has not been a substantial reduction in this rate. Current tocolytics are highly limited in their effectiveness and only provide a short delay in delivery. As preterm birth is a syndrome of heterogeneous aetiologies, therapeutic development is challenging. Further research aimed at elucidating the mechanisms of preterm birth pathophysiology is essential for the generation of predictive, screening, and therapeutic tools. Repurposing therapies poses as an exciting approach for the path forward in innovative therapeutic development for preterm birth. Importantly, preterm birth prevention encapsulates more than medical intervention with tocolysis. Preventative public health measures are also crucial to tackling the underlying causes and risk factors of preterm birth.
Declaration of interest
The authors declare that there are no conflicts of interest. Natalie Hannan is on the editorial board of Reproduction. Natalie was not involved in the review or editorial process for this paper, on which she is listed as an author.
Funding
This study was supported by an ARC Future Fellowship to NJH (FT210100193) and a University of Melbourne postgraduate research scholarship (Felix Meyer Scholarship in Obstetrics & Gynaecology) to BMA.
Author contribution statement
BMA and NJH planned the scope, wrote and edited the manuscript, planned/prepared figures, and contributed guidance to other contributing authors. NKB and NDA contributed to writing and editing of the manuscript. TKL contributed to final edits and version of the manuscript. All authors have read and agreed to publish the present version of the manuscript.
References
Acevedo CH & Ahmed A 1998 Hemeoxygenase-1 inhibits human myometrial contractility via carbon monoxide and is upregulated by progesterone during pregnancy. Journal of Clinical Investigation 101 949–955. (https://doi.org/10.1172/JCI927)
Ahmed A, Williams DJ, Cheed V, Middleton LJ, Ahmad S, Wang K, Vince AT, Hewett P, Spencer K & Khan KS et al.2020 Pravastatin for early-onset pre-eclampsia: a randomised, blinded, placebo-controlled trial. BJOG 127 478–488. (https://doi.org/10.1111/1471-0528.16013)
Ali AA, Sayed AK, El Sherif L, Loutfi GO, Ahmed AMM, Mohamed HB, Anwar AT, Taha AS, Yahia RM & Elgebaly A et al.2019 Systematic review and meta-analysis of randomized controlled trials of atosiban versus nifedipine for inhibition of preterm labor. International Journal of Gynaecology and Obstetrics 145 139–148. (https://doi.org/10.1002/ijgo.12793)
Ara I & Banu H 2008 A prospective randomised trial of nifedipine versus placebo in preterm labour. Bangladesh Journal of Obstetrics and Gynaecology 23 61–64. (https://doi.org/10.3329/bjog.v23i2.4962)
Ashburn TT & Thor KB 2004 Drug repositioning: identifying and developing new uses for existing drugs. Nature Reviews. Drug Discovery 3 673–683. (https://doi.org/10.1038/nrd1468)
Atallah A, Lecarpentier E, Goffinet F, Doret-Dion M, Gaucherand P & Tsatsaris V 2017 Aspirin for prevention of preeclampsia. Drugs 77 1819–1831. (https://doi.org/10.1007/s40265-017-0823-0)
Atwa K, Ibrahim Z, Elshaer M, Taha O & Aboelroose A 2021 Role of esomeprazole in early preeclampsia: a randomized controlled trial. Austin Journal of Obstetrics and Gynecology 8 1189. (https://doi.org/10.26420/austiniobstetgynecol.2021.1189)
Basraon SK, Menon R, Makhlouf M, Longo M, Hankins GD, Saade GR & Costantine MM 2012 Can statins reduce the inflammatory response associated with preterm birth in an animal model? American Journal of Obstetrics and Gynecology 207 224.e1–224.e7. (https://doi.org/10.1016/j.ajog.2012.06.020)
Boyle AK, Rinaldi SF, Rossi AG, Saunders PTK & Norman JE 2019 Repurposing simvastatin as a therapy for preterm labor: evidence from preclinical models. FASEB Journal 33 2743–2758. (https://doi.org/10.1096/fj.201801104R)
Broms G, Kieler H, Ekbom A, Gissler M, Hellgren K, Lahesmaa-Korpinen AM, Pedersen L, Schmitt-Egenolf M, Sorensen HT & Granath F 2020 Anti-TNF treatment during pregnancy and birth outcomes: a population-based study from Denmark, Finland, and Sweden. Pharmacoepidemiology and Drug Safety 29 316–327. (https://doi.org/10.1002/pds.4930)
Brownfoot FC, Tong S, Hannan NJ, Hastie R, Cannon P & Kaitu'u-Lino TJ 2016 Effects of simvastatin, rosuvastatin and pravastatin on soluble fms-like tyrosine kinase 1 (sFlt-1) and soluble endoglin (sENG) secretion from human umbilical vein endothelial cells, primary trophoblast cells and placenta. BMC Pregnancy and Childbirth 16 117. (https://doi.org/10.1186/s12884-016-0902-3)
Burdet J, Sacerdoti F, Cella M, Franchi AM & Ibarra C 2013 Role of TNF-alpha in the mechanisms responsible for preterm delivery induced by Stx2 in rats. British Journal of Pharmacology 168 946–953. (https://doi.org/10.1111/j.1476-5381.2012.02239.x)
Cao G, Liu J & Liu M 2022 Global, regional, and national incidence and mortality of neonatal preterm birth, 1990–2019. JAMA Pediatrics 176 787–796. (https://doi.org/10.1001/jamapediatrics.2022.1622)
Cluver CA, Hannan NJ, Van Papendorp E, Hiscock R, Beard S, Mol BW, Theron GB, Hall DR, Decloedt EH & Stander M et al.2018 Esomeprazole to treat women with preterm preeclampsia: a randomized placebo controlled trial. American Journal of Obstetrics and Gynecology 219 388.e1–388.e17. (https://doi.org/10.1016/j.ajog.2018.07.019)
Coler BS, Shynlova O, Boros-Rausch A, Lye S, Mccartney S, Leimert KB, Xu W, Chemtob S, Olson D & Li M et al.2021 Landscape of preterm birth therapeutics and a path forward. Journal of Clinical Medicine 10. (https://doi.org/10.3390/jcm10132912)
Costantine MM, Cleary K, Hebert MF, Ahmed MS, Brown LM, Ren Z, Easterling TR, Haas DM, Haneline LS & Caritis SN et al.2016 Safety and pharmacokinetics of pravastatin used for the prevention of preeclampsia in high-risk pregnant women: a pilot randomized controlled trial. American Journal of Obstetrics and Gynecology 214 720.e721–720.e717. (https://doi.org/ 10.1016/j.ajog.2015.12.038)
Costantine MM, West H, Wisner KL, Caritis S, Clark S, Venkataramanan R, Stika CS, Rytting E, Wang X & Ahmed MS et al.2021 A randomized pilot clinical trial of pravastatin versus placebo in pregnant patients at high risk of preeclampsia. American Journal of Obstetrics and Gynecology 225 666.e661–666.e615. (https://doi.org/10.1016/j.ajog.2021.05.018)
Crider KS, Whitehead N & Buus RM 2005 Genetic variation associated with preterm birth: a HuGE review. Genetics in Medicine 7 593–604. (https://doi.org/10.1097/01.gim.0000187223.69947.db)
Crowson MS & Shull GE 1992 Isolation and characterization of a cDNA encoding the putative distal colon H+,K(+)-ATPase. Similarity of deduced amino acid sequence to gastric H+,K(+)-ATPase and Na+,K(+)-ATPase and mRNA expression in distal colon, kidney, and uterus. Journal of Biological Chemistry 267 13740–13748. (https://doi.org/10.1016/S0021-9258(1842276-4)
de Alwis N, Beard S, Mangwiro YT, Binder NK, Kaitu'u-Lino TJ, Brownfoot FC, Tong S & Hannan NJ 2020 Pravastatin as the statin of choice for reducing pre-eclampsia-associated endothelial dysfunction. Pregnancy Hypertension 20 83–91. (https://doi.org/10.1016/j.preghy.2020.03.004)
de Heus R, Mulder EJ, Derks JB & Visser GH 2009 The effects of the tocolytics atosiban and nifedipine on fetal movements, heart rate and blood flow. Journal of Maternal-Fetal and Neonatal Medicine 22 485–490. (https://doi.org/10.1080/14767050802702349)
Ding MX, Luo X, Zhang XM, Bai B, Sun JX & Qi HB 2016 Progesterone and nifedipine for maintenance tocolysis after arrested preterm labor: a systematic review and meta-analysis of randomized controlled trial. Taiwanese Journal of Obstetrics and Gynecology 55 399–404. (https://doi.org/10.1016/j.tjog.2015.07.005)
Doret M, Mellier G, Benchaib M, Piacenza JM, Gharib C & Pasquier JC 2002 In vitro study of tocolytic effect of rofecoxib, a specific cyclo-oxygenase 2 inhibitor. Comparison and combination with other tocolytic agents. BJOG 109 983–988. (https://doi.org/10.1111/j.1471-0528.2002.01518.x)
Edison RJ & Muenke M 2004 Central nervous system and limb anomalies in case reports of first-trimester statin exposure. New England Journal of Medicine 350 1579–1582. (https://doi.org/10.1056/NEJM200404083501524)
Fidel PL Jr, Romero R, Cutright J, Wolf N, Gomez R, Araneda H, Ramirez M & Yoon BH 1997 Treatment with the interleukin-I receptor antagonist and soluble tumor necrosis factor receptor Fc fusion protein does not prevent endotoxin-induced preterm parturition in mice. Journal of the Society for Gynecologic Investigation 4 22–26. (https://doi.org/10.1177/107155769700400104)
Flenady V, Reinebrant HE, Liley HG, Tambimuttu EG & Papatsonis DN 2014a Oxytocin receptor antagonists for inhibiting preterm labour. Cochrane Database of Systematic Reviews 6 CD004452. (https://doi.org/10.1002/14651858.CD004452.pub3)
Flenady V, Wojcieszek AM, Papatsonis DN, Stock OM, Murray L, Jardine LA & Carbonne B 2014b Calcium channel blockers for inhibiting preterm labour and birth. Cochrane Database of Systematic Reviews 6 CD002255. (https://doi.org/10.1002/14651858.CD002255.pub2)
Fuchs AR, Fuchs F, Husslein P & Soloff MS 1984 Oxytocin receptors in the human uterus during pregnancy and parturition. American Journal of Obstetrics and Gynecology 150 734–741. (https://doi.org/10.1016/0002-9378(8490677-x)
Gill SK, O'Brien L, Einarson TR & Koren G 2009 The safety of proton pump inhibitors (PPIs) in pregnancy: a meta-analysis. American Journal of Gastroenterology 104 1541–1545. (https://doi.org/10.1038/ajg.2009.122)
Glover AV & Manuck TA 2018 Screening for spontaneous preterm birth and resultant therapies to reduce neonatal morbidity and mortality: a review. Seminars in Fetal and Neonatal Medicine 23 126–132. (https://doi.org/10.1016/j.siny.2017.11.007)
Goldenberg RL, Culhane JF, Iams JD & Romero R 2008 Epidemiology and causes of preterm birth. Lancet 371 75–84. (https://doi.org/10.1016/S0140-6736(0860074-4)
Gomez-Lopez N, Galaz J, Miller D, Farias-Jofre M, Liu Z, Arenas-Hernandez M, Garcia-Flores V, Shaffer Z, Greenberg JM & Theis KR et al.2022 The immunobiology of preterm labor and birth: intra-amniotic inflammation or breakdown of maternal-fetal homeostasis. Reproduction 164 R11–R45. Published online ahead of print. (https://doi.org/10.1530/REP-22-0046)
Gomez-Lopez N, Garcia-Flores V, Chin PY, Groome HM, Bijland MT, Diener KR, Romero R & Robertson SA 2021 Macrophages exert homeostatic actions in pregnancy to protect against preterm birth and fetal inflammatory injury. JCI Insight 6 e146089-146081–e146089-146022. (https://doi.org/10.1172/jci.insight.146089)
Gonzalez JM, Pedroni SM & Girardi G 2014 Statins prevent cervical remodeling, myometrial contractions and preterm labor through a mechanism that involves hemoxygenase-1 and complement inhibition. Molecular Human Reproduction 20 579–589. (https://doi.org/10.1093/molehr/gau019)
Gotsch F, Gotsch F, Romero R, Erez O, Vaisbuch E, Kusanovic JP, Mazaki-Tovi S, Kim SK, Hassan S & Yeo L 2009 The preterm parturition syndrome and its implications for understanding the biology, risk assessment, diagnosis, treatment and prevention of preterm birth. Journal of Maternal-Fetal and Neonatal Medicine 22(Supplement 2) 5–23. (https://doi.org/10.1080/14767050902860690)
Groom KM, Ganzevoort W, Alfirevic Z, Lim K, Papageorghiou AT & Strider Consortium 2018 Clinicians should stop prescribing sildenafil for fetal growth restriction (FGR): comment from the STRIDER Consortium. Ultrasound in Obstetrics and Gynecology 52 295–296. (https://doi.org/10.1002/uog.19186)
Gu S, Zhou C, Pei J, Wu Y, Wan S, Zhao X, Hu J & Hua X 2022 Esomeprazole inhibits hypoxia/endothelial dysfunction-induced autophagy in preeclampsia. Cell and Tissue Research 388 181–194. (https://doi.org/10.1007/s00441-022-03587-z)
Haas DM, Caldwell DM, Kirkpatrick P, Mcintosh JJ & Welton NJ 2012 Tocolytic therapy for preterm delivery: systematic review and network meta-analysis. BMJ 345 e6226. (https://doi.org/10.1136/bmj.e6226)
Hamelin BA & Turgeon J 1998 Hydrophilicity/lipophilicity: relevance for the pharmacology and clinical effects of HMG-CoA Reductase Inhibitors. Trends in Pharmacological Sciences 19 26–37. (https://doi.org/10.1016/s0165-6147(9701147-4)
Hamilton BE, Martin JA & Osterman MJ 2021 Births: provisional data for 2020. In Vital Statistics Rapid Release: Hyattsville, MD: National Center for Health Statistics. (https://doi.org/10.15620/cdc:104993)
Hastie R, Tong S, Wikstrom AK, Sandstrom A, Hesselman S & Bergman L 2021 Aspirin use during pregnancy and the risk of bleeding complications: a Swedish population-based cohort study. American Journal of Obstetrics and Gynecology 224 95.e1–95.e12. (https://doi.org/10.1016/j.ajog.2020.07.023)
Henderson JT, O'connor E & Whitlock EP 2014 Low-dose aspirin for prevention of morbidity and mortality from preeclampsia. Annals of Internal Medicine 161 613–614. (https://doi.org/10.7326/L14-5020-5)
Hoffman MK, Goudar SS, Kodkany BS, Metgud M, Somannavar M, Okitawutshu J, Lokangaka A, Tshefu A, Bose CL & Mwapule A et al.2020 Low-dose aspirin for the prevention of preterm delivery in nulliparous women with a singleton pregnancy (Aspirin): a randomised, double-blind, placebo-controlled trial. Lancet 395 285–293. (https://doi.org/10.1016/S0140-6736(1932973-3)
Holmgren C, Esplin MS, Hamblin S, Molenda M, Simonsen S & Silver R 2008 Evaluation of the use of anti-TNF-alpha in an LPS-induced murine model. Journal of Reproductive Immunology 78 134–139. (https://doi.org/10.1016/j.jri.2007.11.003)
Holt R, Timmons BC, Akgul Y, Akins ML & Mahendroo M 2011 The molecular mechanisms of cervical ripening differ between term and preterm birth. Endocrinology 152 1036–1046. (https://doi.org/10.1210/en.2010-1105)
Ibrahim MH, Elfaki T, Elhassan EM, Abdelrahiem SK & Adam I 2021 The effectiveness of nifedipine/indomethacin combination therapy and nifedipine monotherapy for postponing preterm birth (25–34 weeks of gestation) in Sudanese women: a randomized clinical trial study protocol. BMC Pregnancy and Childbirth 21 457. (https://doi.org/10.1186/s12884-021-03951-x)
Jaisser F, Coutry N, Farman N, Binder HJ & Rossier BC 1993 A putative H(+)-K(+)-ATPase is selectively expressed in surface epithelial cells of rat distal colon. American Journal of Physiology 265 C1080–C1089. (https://doi.org/10.1152/ajpcell.1993.265.4.C1080)
Kashanian M, Shirvani S, Sheikhansari N & Javanmanesh F 2020 A comparative study on the efficacy of nifedipine and indomethacin for prevention of preterm birth as monotherapy and combination therapy: a randomized clinical trial. Journal of Maternal-Fetal and Neonatal Medicine 33 3215–3220. (https://doi.org/10.1080/14767058.2019.1570117)
Keirse MJ 1992 Therapeutic uses of prostaglandins. Bailliere's Clinical Obstetrics and Gynaecology 6 787–808. (https://doi.org/10.1016/s0950-3552(0580189-0)
Kumasawa K, Ikawa M, Kidoya H, Hasuwa H, Saito-Fujita T, Morioka Y, Takakura N, Kimura T & Okabe M 2011 Pravastatin induces placental growth factor (PGF) and ameliorates preeclampsia in a mouse model. Proceedings of the National Academy of Sciences of the United States of America 108 1451–1455. (https://doi.org/10.1073/pnas.1011293108)
Landman AJEMC, De Boer MA, Visser L, Nijman TAJ, Hemels MAC, Naaktgeboren CN, Van Der Weide MC, Mol BW, Van Laar JOEH & Papatsonis DNM et al.2022 Evaluation of low-dose aspirin in the prevention of recurrent spontaneous preterm labour (the APRIL study): A multicentre, randomised, double-blinded, placebo-controlled trial. PLOS Medicine 19 e1003892. (https://doi.org/10.1371/journal.pmed.1003892)
Lartey J & Lopez Bernal A 2009 RHO protein regulation of contraction in the human uterus. Reproduction 138 407–424. (https://doi.org/10.1530/REP-09-0160)
Le BL, Iwatani S, Wong RJ, Stevenson DK & Sirota M 2020 Computational discovery of therapeutic candidates for preventing preterm birth. JCI Insight 5 e133761. (https://doi.org/10.1172/jci.insight.133761)
Le Ray C, Maillard F, Carbonne B, Verspyck E & Cabrol D Goffinet F & Groupe Evaprima 2010 Nifedipine or Nicardipine in management of threatened preterm delivery: an observational population-based study. Journal de Gynécologie, Obstétrique et Biologie de la Reproduction 39 490–4 97. (https://doi.org/10.1016/j.jgyn.2010.04.004)
Leonhardt A, Bernert S, Watzer B, Schmitz-Ziegler G & Seyberth HW 2003 Low-dose aspirin in pregnancy: maternal and neonatal aspirin concentrations and neonatal prostanoid formation. Pediatrics 111 e77–e81. (https://doi.org/10.1542/peds.111.1.e77)
Lopez Bernal A 2003 Mechanisms of labour--biochemical aspects. BJOG 110(Supplement 20) 39–45. (https://doi.org/10.1046/j.1471-0528.2003.00023.x)
Lyon D, Cheng CY, Howland L, Rattican D, Jallo N, Pickler R, Brown L & Mcgrath J 2010 Integrated review of cytokines in maternal, cord, and newborn blood: Part I—Associations with preterm birth. Biological Research for Nursing 11 371–376. (https://doi.org/10.1177/1099800409344620)
Mackeen AD, Seibel-Seamon J, Muhammad J, Baxter JK & Berghella V 2014 Tocolytics for preterm premature rupture of membranes. Cochrane Database of Systematic Reviews 2 CD007062. (https://doi.org/10.1002/14651858.CD007062.pub3)
Marchand G, Blumrick R, Ruuska AD, Ware K, Masoud AT, King A, Ruther S, Brazil G, Cieminski K & Calteux N et al.2021 Novel oxytocin receptor antagonists for tocolysis: a systematic review and meta-analysis of the available data on the efficacy, safety, and tolerability of retosiban. Current Medical Research and Opinion 37 1677–1688. (https://doi.org/10.1080/03007995.2021.1944076)
Molnar M & Hertelendy F 1990 Regulation of intracellular free calcium in human myometrial cells by prostaglandin F2 alpha: comparison with oxytocin. Journal of Clinical Endocrinology and Metabolism 71 1243–1250. (https://doi.org/10.1210/jcem-71-5-1243)
Nadeau-Vallee M, Obari D, Palacios J, Brien MÈ, Duval C, Chemtob S & Girard S 2016 Sterile inflammation and pregnancy complications: a review. Reproduction 152 R277–R292. (https://doi.org/10.1530/REP-16-0453)
Naik Gaunekar N, Raman P, Bain E & Crowther CA 2013 Maintenance therapy with calcium channel blockers for preventing preterm birth after threatened preterm labour. Cochrane Database of Systematic Reviews 10 CD004071. (https://doi.org/10.1002/14651858.CD004071.pub3)
Neilson JP, West HM & Dowswell T 2014 Betamimetics for inhibiting preterm labour. Cochrane Database of Systematic Reviews 2 CD004352. (https://doi.org/10.1002/14651858.CD004352.pub3)
Neuman RI, Baars MD, Saleh L, Broekhuizen M, Nieboer D, Cornette J, Schoenmakers S, Verhoeven M, Koch BCP & Russcher H et al.2022 Omeprazole administration in preterm preeclampsia: a randomized controlled trial to study its effect on sFlt-1 (soluble Fms-like tyrosine Kinase-1), PlGF (placental growth factor), and ET-1 (endothelin-1). Hypertension 79 1297–1307. (https://doi.org/10.1161/HYPERTENSIONAHA.122.19070)
ObsEva 2021 ObsEva Presents PROLONG Phase 2a Proof-of-Concept Data on Ebopiprant (OBE022) for the treatment of Spontaneous preterm labor at the RCOG Virtual World Congress 2021. [online]. Available at: https://www.globenewswire.com/news-release/2021/06/10/2244873/0/en/ObsEva-Presents-PROLONG-Phase-2a-Proof-of-Concept-Data-on-Ebopiprant-OBE022-for-the-Treatment-of-Spontaneous-Preterm-Labor-at-the-RCOG-Virtual-World-Congress-2021.html
Olson DM, Christiaens I, Gracie S, Yamamoto Y & Mitchell BF 2008 Emerging tocolytics: challenges in designing and testing drugs to delay preterm delivery and prolong pregnancy. Expert Opinion on Emerging Drugs 13 695–707. (https://doi.org/10.1517/14728210802568764)
Onda K, Tong S, Beard S, Binder N, Muto M, Senadheera SN, Parry L, Dilworth M, Renshall L & Brownfoot F et al.2017 Proton pump inhibitors decrease soluble fms-like tyrosine Kinase-1 and soluble endoglin secretion, decrease hypertension, and rescue endothelial dysfunction. Hypertension 69 457–468. (https://doi.org/10.1161/HYPERTENSIONAHA.116.08408)
Pasternak B & Hviid A 2010 Use of proton-pump inhibitors in early pregnancy and the risk of birth defects. New England Journal of Medicine 363 2114–2123. (https://doi.org/10.1056/NEJMoa1002689)
Paul JW, Hua S, Ilicic M, Tolosa JM, Butler T, Robertson S & Smith R 2017 Drug delivery to the human and mouse uterus using immunoliposomes targeted to the oxytocin receptor. American Journal of Obstetrics and Gynecology 216 283.e1–283.e14. (https://doi.org/10.1016/j.ajog.2016.08.027)
Perez-Guerrero C, Marquez-Martin A, Herrera MD, Marhuenda E & Alvarez De Sotomayor M 2005 Regulation of vascular tone from spontaneously hypertensive rats by the HMG-CoA reductase inhibitor, simvastatin. Pharmacology 74 209–215. (https://doi.org/10.1159/000085957)
Pohl O, Chollet A, Kim SH, Riaposova L, Spezia F, Gervais F, Guillaume P, Lluel P, Meen M & Lemaux F et al.2018a OBE022, an oral and selective prostaglandin F2alpha receptor antagonist as an effective and safe modality for the treatment of preterm labor. Journal of Pharmacology and Experimental Therapeutics 366 349–364. (https://doi.org/10.1124/jpet.118.247668)
Pohl O, Marchand L, Gotteland JP, Coates S, Taubel J & Lorch U 2018b Pharmacokinetics, safety and tolerability of OBE022, a selective prostaglandin F2alpha receptor antagonist tocolytic: a first-in-human trial in healthy postmenopausal women. British Journal of Clinical Pharmacology 84 1839–1855. (https://doi.org/10.1111/bcp.13622)
Pohl O, Marchand L, Gotteland JP, Coates S, Taubel J & Lorch U 2019 Coadministration of the prostaglandin F2alpha receptor antagonist preterm labour drug candidate OBE022 with magnesium sulfate, atosiban, nifedipine and betamethasone. British Journal of Clinical Pharmacology 85 1516–1527. (https://doi.org/10.1111/bcp.13925)
Pritchard N, Kaitu'u-Lino T, Harris L, Tong S & Hannan N 2021 Nanoparticles in pregnancy: the next frontier in reproductive therapeutics. Human Reproduction Update 27 280–304. (https://doi.org/10.1093/humupd/dmaa049)
Refuerzo JS, Alexander JF, Leonard F, Leon M, Longo M & Godin B 2015 Liposomes: a nanoscale drug carrying system to prevent indomethacin passage to the fetus in a pregnant mouse model. American Journal of Obstetrics and Gynecology 212 508.e1–508.e7. (https://doi.org/10.1016/j.ajog.2015.02.006)
Refuerzo JS, Leonard F, Bulayeva N, Gorenstein D, Chiossi G, Ontiveros A, Longo M & Godin B 2016 Uterus-targeted liposomes for preterm labor management: studies in pregnant mice. Scientific Reports 6 34710. (https://doi.org/10.1038/srep34710)
Reinebrant HE, Pileggi-Castro C, Romero CL, Dos Santos RA, Kumar S, Souza JP & Flenady V 2015 Cyclo-oxygenase (COX) inhibitors for treating preterm labour. Cochrane Database of Systematic Reviews 6 CD001992. (https://doi.org/10.1002/14651858.CD001992.pub3)
Rhoden KJ, Tallini G & Douglas JS 1996 H+-K+ ATPase inhibitors cause relaxation of guinea pig and human airway smooth muscle in vitro. Journal of Pharmacology and Experimental Therapeutics 276 897–903.
Roberge S, Nicolaides KH, Demers S, Villa P & Bujold E 2013 Prevention of perinatal death and adverse perinatal outcome using low-dose aspirin: a meta-analysis. Ultrasound in Obstetrics and Gynecology 41 491–499. (https://doi.org/10.1002/uog.12421)
Robertson SA, Hutchinson MR, Rice KC, Chin PY, Moldenhauer LM, Stark MJ, Olson DM & Keelan JA 2020 Targeting Toll-like receptor-4 to tackle preterm birth and fetal inflammatory injury. Clinical and Translational Immunology 9 e1121. (https://doi.org/10.1002/cti2.1121)
Romero R, Dey SK & Fisher SJ 2014 Preterm labor: one syndrome, many causes. Science 345 760–765. (https://doi.org/10.1126/science.1251816)
Romero R, Espinoza J, Kusanovic JP, Gotsch F, Hassan S, Erez O, Chaiworapongsa T & Mazor M 2006 The preterm parturition syndrome. BJOG 113(Supplement 3) 17–42. (https://doi.org/10.1111/j.1471-0528.2006.01120.x)
Romero R, Gomez R, Chaiworapongsa T, Conoscenti G, Kim JC & Kim YM 2001 The role of infection in preterm labour and delivery. Paediatric and Perinatal Epidemiology 15(Supplement 2) 41–56. (https://doi.org/10.1046/j.1365-3016.2001.00007.x)
Sadowsky DW, Adams KM, Gravett MG, Witkin SS & Novy MJ 2006 Preterm labor is induced by intraamniotic infusions of interleukin-1beta and tumor necrosis factor-alpha but not by interleukin-6 or interleukin-8 in a nonhuman primate model. American Journal of Obstetrics and Gynecology 195 1578–1589. (https://doi.org/10.1016/j.ajog.2006.06.072)
Saleh L, Samantar R, Garrelds IM, Van den Meiracker AH, Visser W & Danser AHJ 2017 Low soluble Fms-like tyrosine Kinase-1, endoglin, and endothelin-1 levels in women with confirmed or suspected preeclampsia using proton pump inhibitors. Hypertension 70 594–600. (https://doi.org/10.1161/HYPERTENSIONAHA.117.09741)
Shin SH, Kim EK, Lee KY & Kim HS 2019 TNF-alpha antagonist attenuates systemic lipopolysaccharide-induced brain white matter injury in neonatal rats. BMC Neuroscience 20 45. (https://doi.org/10.1186/s12868-019-0529-1)
Silver RM, Edwin SS, Umar F, Dudley DJ, Branch DW & Mitchell MD 1997 Bacterial lipopolysaccharide-mediated murine fetal death: the role of interleukin-1. American Journal of Obstetrics and Gynecology 176 544–549. (https://doi.org/10.1016/s0002-9378(9770545-3)
Singh J, Ahmed A & Girardi G 2011 Role of complement component C1q in the onset of preeclampsia in mice. Hypertension 58 716–724. (https://doi.org/10.1161/HYPERTENSIONAHA.111.175919)
Sivarajasingam SP, Imami N & Johnson MR 2016 Myometrial cytokines and their role in the onset of labour. Journal of Endocrinology 231 R101–R119. (https://doi.org/10.1530/JOE-16-0157)
Slater DM, Dennes WJ, Campa JS, Poston L & Bennett PR 1999 Expression of cyclo-oxygenase types-1 and -2 in human myometrium throughout pregnancy. Molecular Human Reproduction 5 880–884. (https://doi.org/10.1093/molehr/5.9.880)
So T, Ito A, Sato T, Mori Y & Hirakawa S 1992 Tumor necrosis factor-alpha stimulates the biosynthesis of matrix metalloproteinases and plasminogen activator in cultured human chorionic cells. Biology of Reproduction 46 772–778. (https://doi.org/10.1095/biolreprod46.5.772)
Sood BG, Lulic-Botica M, Holzhausen KA, Pruder S, Kellogg H, Salari V & Thomas R 2011 The risk of necrotizing enterocolitis after indomethacin tocolysis. Pediatrics 128 e54–e62. (https://doi.org/10.1542/peds.2011-0265)
Taubel J, Lorch U, Coates S, Fernandes S, Foley P, Ferber G, Gotteland JP & Pohl O 2018 Confirmation of the cardiac safety of PGF2alpha receptor antagonist OBE022 in a first-in-human study in healthy subjects, using intensive ECG assessments. Clinical Pharmacology in Drug Development 7 889–900. (https://doi.org/10.1002/cpdd.447)
Terranova C, Petrella C, Improta G, Severini C, Signore F, Damiani P, Plotti F, Scarpignato C & Angioli R 2014 Relaxant effect of proton pump inhibitors on in vitro myometrium from pregnant women. European Journal of Pharmaceutical Sciences 52 125–131. (https://doi.org/10.1016/j.ejps.2013.10.007)
Thornton S, Goodwin TM, Greisen G, Hedegaard M & Arce JC 2009 The effect of Barusiban, a selective oxytocin antagonist, in threatened preterm labor at late gestational age: a randomized, double-blind, placebo-controlled trial. American Journal of Obstetrics and Gynecology 200 627.e1–627.10. (https://doi.org/10.1016/j.ajog.2009.01.015)
Thornton S, Valenzuela G, Baidoo C, Fossler MJ, Montague TH, Clayton L, Powell M, Snidow J, Stier B & Soergel D 2017 Treatment of spontaneous preterm labour with retosiban: a phase II pilot dose-ranging study. British Journal of Clinical Pharmacology 83 2283–2291. (https://doi.org/10.1111/bcp.13336)
Tracey D, Klareskog L, Sasso EH, Salfeld JG & Tak PP 2008 Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacology and Therapeutics 117 244–279. (https://doi.org/10.1016/j.pharmthera.2007.10.001)
Ulmsten U, Andersson KE & Wingerup L 1980 Treatment of premature labor with the calcium antagonist nifedipine. Archives of Gynecology 229 1–5. (https://doi.org/10.1007/BF02109822)
Van den Veyver IB & Moise KJ Jr 1993 Prostaglandin synthetase inhibitors in pregnancy. Obstetrical and Gynecological Survey 48 493–502. (https://doi.org/10.1097/00006254-199307000-00026)
Vane JR 1971 Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nature: New Biology 231 232–235. (https://doi.org/10.1038/newbio231232a0)
Vogel JP, Chawanpaiboon S, Moller AB, Watananirun K, Bonet M & Lumbiganon P 2018 The global epidemiology of preterm birth. Best Practice and Research. Clinical Obstetrics and Gynaecology 52 3–12. (https://doi.org/10.1016/j.bpobgyn.2018.04.003)
Vogel JP, Nardin JM, Dowswell T, West HM & Oladapo OT 2014 Combination of tocolytic agents for inhibiting preterm labour. Cochrane Database of Systematic Reviews 7 CD006169. (https://doi.org/10.1002/14651858.CD006169.pub2)
Welsh C, Kasirer MY, Pan J, Shifrin Y & Belik J 2014 Pantoprazole decreases gastroesophageal muscle tone in newborn rats via rho-kinase inhibition. American Journal of Physiology: Gastrointestinal and Liver Physiology 307 G390–G396. (https://doi.org/10.1152/ajpgi.00005.2014)
Whitaker EMH, Whyte SA, Norrie J, Andrew R, Williams D, Stock SJ & Norman JE 2019 PIPIN: a feasibility trial of pravastatin for the prevention of preterm birth in women trial methods and protocol. Reproductive Sciences 26 320A. (https://doi.org/10.1177/1933719119834079)
World Health Organisation 2015 WHO Recommendations on Interventions to Improve Preterm Birth Outcomes: Geneva: World Health Organisation.
Wu SP & Demayo FJ 2017 Progesterone receptor signaling in uterine myometrial physiology and preterm birth. Current Topics in Developmental Biology 125 171–190. (https://doi.org/10.1016/bs.ctdb.2017.03.001)
Yildirim K, Sarioglu Y, Kaya T, Cetin A & Yildirim S 2001 Inhibitor effect of omeprazole in isolated human myometrial smooth muscle. Life Sciences 69 435–442. (https://doi.org/10.1016/s0024-3205(0101135-3)
Zarek J & Koren G 2014 The fetal safety of statins: a systematic review and meta-analysis. Journal of Obstetrics and Gynaecology Canada. Journal d'Obstétrique et Gynécologie du Canada 36 506–509. (https://doi.org/10.1016/S1701-2163(1530565-X)
Zhang X & Liu M 2002 Clinical observations on the prevention and treatment of premature labor with nifedipine. Hua Xi Yi Ke Da Xue Xue Bao [Journal of West China University of Medical Sciences] 33 288–290.
Zuckerman H, Reiss U & Rubinstein I 1974 Inhibition of human premature labor by indomethacin. Obstetrics and Gynecology 44 787–792. (https://doi.org/10.1097/00006254-197505000-00009)