Abstract
Patent ductus arteriosus (PDA) is the most common cardiovascular condition diagnosed in premature infants. Acetaminophen was first proposed as a potential treatment for PDA in 2011. Since that time acetaminophen use among extremely preterm neonates has increased substantially. The limited available data demonstrate that acetaminophen reduces PDA without evident hepatotoxicity. These findings have led some to suggest that acetaminophen is a safe and effective therapy for PDA closure. However, the lack of apparent hepatoxicity is predictable. Acetaminophen induced cellular injury is due to CYP2E1 derived metabolites; and hepatocyte CYP2E1 expression is low in the fetal and neonatal period. Here, we review preclinical and clinical data that support the hypothesis that the lung, which expresses high levels of CYP2E1 during fetal and early postnatal development, may be particularly susceptible to acetaminophen induced toxicity. Despite these emerging data, the true potential pulmonary risks and benefits of acetaminophen for PDA closure are largely unknown. The available clinical studies in are marked by significant weakness including low sample sizes and minimal evaluation of extremely preterm infants who are typically at highest risk of pulmonary morbidity. We propose that studies interrogating mechanisms linking developmentally regulated, cell-specific CYP2E1 expression and acetaminophen-induced toxicity as well as robust assessment of pulmonary outcomes in large trials that evaluate the safety and efficacy of acetaminophen in extremely preterm infants are needed.
Introduction
Patent ductus arteriosus (PDA) is the most common cardiovascular condition diagnosed in premature infants [1]. The incidence of PDA is inversely related to gestational age at birth and can be detected by echocardiography at two months of age in over 50% of infants born less than 26 weeks’ gestation [2]. In hopes of avoiding serious morbidities associated with PDA, including bronchopulmonary dysplasia (BPD) and necrotizing enterocolitis, clinicians have sought safe and effective ways to close the PDA via pharmacological or procedural intervention [3]. Since the 1970s, non-steroidal anti-inflammatory drugs (NSAIDs) have been the most common medications used to treat the PDA [1, 4]. These drugs inhibit cyclooxygenases, which decrease prostaglandin production and facilitate ductal constriction. Unfortunately, randomized trials have not demonstrated consistent respiratory and long-term neurologic benefit with these medications and evidence indicates they are associated with unwanted adverse drug effects that may negate the potential benefits conferred by earlier ductal closure [3].
Not surprisingly, there has been interest in identifying safer, alternative treatments for PDA. A case series published in 2011 reported the “incidental observation” that a persistent, hemodynamically significant PDA closed after acetaminophen was administered “for unrelated reason” to 17-day-old infant born at 26 weeks’ gestational age [5]. In total, the authors described a similar relationship in 5 preterm neonates [5]. Since this initial case series, the use of acetaminophen to treat the PDA has increased substantially and has been identified as a “promising alternative to indomethacin and ibuprofen with a better safety profile.” [6] However, pre-clinical and epidemiological data raise concern that the use of acetaminophen to treat PDA may carry risks and that the assumption of drug safety may be premature. In this commentary, we review recent clinical data characterizing the use and outcomes associated with acetaminophen administration in premature infants and highlight the results of pre-clinical studies that point towards potentially important but under evaluated mechanisms of adverse pulmonary drug effects with acetaminophen use in extremely preterm infants.
Increasing frequency of acetaminophen use in preterm infants
Since early reports temporally linking acetaminophen administration with closure of the PDA, acetaminophen has become one of the most frequently prescribed medications in neonatal intensive care units (NICU) worldwide. In NICUs managed by the Pediatrix Medical Group, acetaminophen had the eighth highest absolute increase in use relative to other common medications between the years 2010 and 2017 [7]. In NICUs in the United Kingdom (UK), acetaminophen demonstrated the fifth highest absolute increase in use during the same time frame [8]. In French Level 3 neonatal wards in 2017–18, only vitamin supplementation exceeded acetaminophen use, with approximately 65% of infants born less than 27 week’s gestation receiving acetaminophen therapy [9]. In the international PDA-TOLERATE trial, acetaminophen was chosen by clinicians as the first line therapy in 26% of extremely preterm infants who were randomized to early drug treatment [10]. Data from an Italian multicenter report indicate acetaminophen was used to treat PDA in 24% of infants born at 25–28 weeks’ gestational age and 47% of those born at 23–24 weeks’ gestational age [11]. Lastly, survey of clinicians in Australia, New Zealand, and the UK suggest that 70–80% of neonatologists in those countries have prescribed acetaminophen to treat PDA [12, 13].
Is acetaminophen effective for PDA closure?
Multiple small, randomized trials have evaluated the efficacy and tolerability of acetaminophen for PDA closure in preterm or low-birth-weight infants. A Cochrane systematic review published in 2022 summarized the data from 27 studies that enrolled a total of 2278 infants [14]. Multiple comparisons were conducted including acetaminophen versus ibuprofen, acetaminophen versus indomethacin, and acetaminophen versus placebo or no intervention. Of these comparisons, acetaminophen versus placebo or no intervention included the fewest number of infants (3 studies, 240 infants). When compared to placebo/no intervention, infants who received acetaminophen were less likely to have a PDA detected by post-treatment echocardiography (16.8% vs. 61.1%, relative risk 0.27, 95% CI 0.18–0.42). Acetaminophen had similar efficacy for preventing subsequent PDA when compared to ibuprofen (18 trials, 1535 infants; rates of treatment failure: 30.3% vs 30.1; RR 1.02, 95% CI 0.88–1.18) and indomethacin (4 trials, 380 infants; rates of treatment failure 29.8% vs. 29.7%; RR 1.02, 95% CI 0.78–1.33). The authors concluded that the available data provided moderate-certainty evidence that acetaminophen and ibuprofen have similar effectiveness for PDA closure, but low-certainty evidence for the other comparisons.
The mechanism of action of acetaminophen to achieve PDA closure is not fully understood. Parts of its activity mimic that of COX-2 selective inhibitors indicating that acetaminophen, similar to NSAIDs, may decrease prostaglandin production [6, 15]. With the increasing use of acetaminophen in premature infants and growing evidence that it may achieve similar rates of PDA closure, it is essential to consider whether acetaminophen is truly a safer alternative.
Is acetaminophen safe to use for PDA closure?
At present, it is unclear whether acetaminophen provides a more favorable safety profile than alternative treatment options. Several limitations in the current data contribute to this uncertainty. Firstly, the available individual trials have been small; the largest reported outcomes for only 200 participants [14]. Secondly, most trials included relatively few infants born less than 28 weeks of gestation, the population that is most likely to be treated for a PDA and potentially most vulnerable to adverse drug effects. Lastly, there is inconsistent reporting of key neonatal outcomes, particularly related to organ systems that might be vulnerable to acetaminophen toxicity. The sections that follow discuss pre-clinical and clinical studies that raise important safety considerations with acetaminophen use in preterm infants.
Is hepatotoxicity the most relevant safety measure in neonates?
The risk of hepatoxicity with excess acetaminophen exposure in children and adults is well-established. In the pediatric population as a whole, acetaminophen overdose is a leading cause of hepatic failure [16,17,18]. Likely motivated by this known association, trials of acetaminophen therapy for PDA closure in preterm infants have frequently assessed hepatic injury by monitoring serum alanine and aspartate transaminases [14]. However, unlike in older children, acetaminophen-induced hepatotoxicity is rare in the neonatal period [19, 20]. As per one recent report, “clinical evidence shows a low or absent hepatic toxicity in neonates, suggesting the existence of a large therapeutic serum concentration range for paracetamol.” [21] Understanding the mechanisms underlying acetaminophen-induced hepatoxicity helps explain why this injury is uncommon in newborns.
Acetaminophen toxicity is dependent on cell-type specific expression of the xenobiotic enzyme CYP2E1 (Fig. 1). In adults, hepatic glucuronyl transferases and sulfotransferases convert 80–90% of acetaminophen into non-toxic metabolites [21,22,23,24]. Pericentral hepatocytes express CYP2E1 and convert the remaining small percentage of acetaminophen into the toxic metabolite N-acetyl-p-benzoquinone imine (NAPQI). Available glutathione stores readily inactivate this toxic metabolite. However, when these normal metabolizing pathways are saturated and glutathione is consumed, accumulating NAPQI binds to various proteins resulting in mitochondrial dysfunction and cell death. In adults, pericentral hepatocytes express the highest level of CYP2E1, which explains why these cells are the most sensitive to acetaminophen overdose and the zone-specific pattern of resulting hepatotoxicity (Fig. 2) [25, 26].
This is a representative H&E stained hepatic section from an acetaminophen exposed (2, 8 and 24 h; 280 mg/kg, IP) adult male ICR mouse. Areas of necrosis and sinusoidal dilution are located around the central vein (CV) while hepatocytes around the portal triad (PT) are spared. Internal scale bar 100 μm.
The finding of low hepatic expression of CYP2E1 in the fetus and neonate is consistent across multiple species including humans [27,28,29,30,31]. In preclinical studies, neonatal animals demonstrate resistance to acetaminophen-induced hepatic injury [32, 33]. In a similar manner, hepatotoxicity is rarely reported in human neonates following supratherapeutic acetaminophen exposures [19]. While these findings indicate acetaminophen is unlikely to result in hepatotoxicity when used in the neonatal period, they also demonstrate that hepatic expression of the enzyme responsible for acetaminophen-induced cell injury – CYP2E1 - is dynamic during fetal and postnatal development. Thorough examination of whether other cell types express CYP2E1, and whether that expression is dynamic during development may help identify cell populations at risk of acetaminophen-induced injury and inform clinical assessment of acetaminophen safety.
Pulmonary CYP2E1 expression – a potential mechanism of acetaminophen toxicity in the neonate?
In addition to expression within the liver, it is well recognized that CYP2E1 is present within the pulmonary epithelium, club cells, and macrophages of the adult lung [34, 35]. These findings suggest a mechanistic framework underlying the observation that lung injury is a consistent, co-occurring finding with hepatotoxic acetaminophen exposures in rodents [19, 20, 36,37,38,39,40,41]. Moreover, there is evidence that pulmonary injury occurs independent of hepatic injury. Following toxic acetaminophen exposure, glutathione depletion and acetaminophen-adduct accumulation occurs in the adult murine lung [36, 41,42,43,44]. Direct administration of acetaminophen or intratracheal NAPQI injures alveolar epithelial cells in culture and in the in-vivo lung, respectively [45,46,47]. Furthermore, pulmonary injury occurs even when hepatic injury is mitigated [38,39,40, 48, 49]. These observations had previously been limited to adult rodents following hepatotoxic exposures. We recently demonstrated that the adult murine lung is susceptible to acetaminophen-induced injury even in the absence of hepatotoxicity [48]. Potentially more relevant to preterm infants, we have now shown that the murine lung in the early alveolar stage of development (equivalent to late preterm or term gestation in human infants) is similarly susceptible to injury with acetaminophen exposures that do not cause hepatotoxicity [50].
Until recently, the pulmonary expression profile of CYP2E1 during lung development was not known. However, new data demonstrate dynamic, developmentally regulated and pulmonary cell type specific expression of CYP2E1. Data from the Molecular Atlas of Lung Development Program (LungMAP) indicate that CYP2E1 expression exhibits two peaks during murine lung development: one at embryonic day 18 during the saccular stage of lung development, and one at postnatal day 14, during the second phase of murine lung alveologenesis [51]. These data have been confirmed in a recent single-cell atlas of mouse lung development and in a single-cell analysis of mouse lung fibroblasts [52, 53]. Of note, lung mesenchymal myofibroblasts characterized by Pdgfra (platelet derived growth factor receptor alpha) expression [54,55,56] are potentially susceptible to APAP exposure based on their expression of CYP2E1. These cells are abundant in the developing murine lung (Fig. 3) and multiple studies have demonstrated that disruption of Pdgfrα signaling or myofibroblast function during lung development results in abnormal alveolar formation characterized by simplification and loss of surface area for gas exchange, a common histological phenotype observed in preterm infants with BPD [54, 57,58,59,60]. Lastly, data from the lungs of human neonates born at 30 weeks’ gestational age also demonstrate lung mesenchymal cell expression of CYP2E1 expression [61,62,63]. Collectively, these suggest a potential mechanism by which acetaminophen use could contribute to lung injury in preterm infants. However, it is unknown whether acetaminophen exposures that occur during periods marked by high mesenchymal myofibroblast specific CYP2E1 expression result in cellular dysfunction and abnormal lung development in humans.
Is there clinical evidence of adverse effects of acetaminophen exposures during human development?
Although large-scale use of acetaminophen in the preterm population is a relatively new phenomenon, there has been long-standing use of this therapy in other potentially at-risk populations. In recent years, closer examination of the potential adverse effects of acetaminophen exposure have prompted renewed interest in studying the safety of this drug therapy. For instance, acetaminophen use during pregnancy became commonplace in the 1970s and 80 s and now occurs in 60–70% of pregnancies in US [64]. Clinical studies in the 1970s of in utero exposures confirmed a lack of teratogenicity [65], and use quickly became regarded as safe [66] and “recommended, without reservation, in standard therapeutic doses during pregnancy.” [67] Unfortunately, preclinical studies examining the impact of acetaminophen exposures during pregnancy on fetal development at that time were sparse, and limited to the assessment of the placenta and fetal “growth” (i.e., weight) [68], and fetal hepatic glutathione consumption [69]. However, emerging clinical data from large epidemiological registries have raised concern that perinatal exposure to acetaminophen may be harmful to the developing offspring. Owing to observations such as this as well as the potential links between acetaminophen and endocrinologic and neurologic morbidity in offspring, one expert panel recommended that women should be cautioned at the beginning of pregnancy to avoid acetaminophen unless it is “medically indicated” and that the lowest effective dose should be used for the shortest possible time [70]. The precautionary conclusions reached by these authors, which are based on observational data and lack mechanistic relationships have been criticized by multiple groups, including the Society of Obstetricians and Gynaecologists of Canada [71], the American College of Obstetricians and Gynecologists [72], and the European Network of Teratology Information Services [73]. Greater research in this area will support evidence-based use of acetaminophen in pregnant mothers.
The possibility of adverse respiratory effects with acetaminophen exposure during fetal development and in early childhood has been previously explored. The first clinical evidence that routine acetaminophen use may affect pulmonary outcomes emerged in the early 2000s. During this period, Shaheen and colleagues published a series of manuscripts that demonstrated significant adverse associations between acetaminophen exposure and the development of asthma in children and adults [74,75,76]. A recent metanalysis comprised of 13 studies and over 1 million subjects found that “prenatal paracetamol exposure could increase the risk of child asthma (OR = 1.19; 95% CI, 1.12–1.27).” [77] Similarly, early childhood acetaminophen exposure has been linked to the development of asthma [78]. Whether this association represents a causal relationship continues to be debated, in part because pre-clinical studies have not yet linked acetaminophen exposure to an allergic or asthmatic phenotype [79]. Notably, these studies were motivated by pre-clinical observations which showed that acetaminophen depletes pulmonary glutathione stores and thus potentially increases susceptibility to oxidant and inflammatory stress [36, 41,42,43,44]. This demonstration of pre-clinical findings supporting subsequent clinical work provides a useful example that may inform future studies on the safety of acetaminophen in extremely preterm infants.
Is there clinical evidence from randomized trials that acetaminophen for PDA adversely impacts lung development or function in preterm infants?
It is largely unknown whether acetaminophen use contributes to poor pulmonary outcomes in preterm infants. BPD is the most commonly reported pulmonary outcome in trials evaluating acetaminophen to treat PDA. However, the inclusion of this outcome has been inconsistent and might have been motivated more by hopes of observed benefit from ductal closure rather than concerns related to drug toxicity. Among the 27 trials included in the 2022 Cochrane review on acetaminophen for PDA closure in preterm infants, only 15 reported BPD as an outcome and only 8 specified which definition or diagnostic criteria were used for BPD (Table 1). Moreover, only 5 of the 15 trials that reported BPD as an outcome enrolled subjects born less than 28 weeks’ gestational age [80,81,82,83,84]. As a result, the risk of BPD following acetaminophen therapy for PDA has been assessed in randomized trials enrolling fewer than 125 subjects born extremely preterm. None of the few trials that included long-term follow-up assessed pulmonary outcomes [85]. This small amount of data precludes any clear conclusions regarding the potential risk of adverse pulmonary effects with acetaminophen exposure.
Do data from observational studies raise concern that acetaminophen for PDA may adversely impact lung development or function?
Case series and cohort studies examining acetaminophen use have included larger numbers of high-risk extremely preterm infants. Unfortunately, similar to the trial data, the majority of these studies did not report pulmonary outcomes and many are underpowered to detect potential harm [19]. Among those that evaluated the incidence of BPD, some suggested that acetaminophen may be associated with increased risk of BPD, observing statistically significant differences or large albeit non-significant increases in BPD rates [86,87,88]. However, these findings conflict with other observational studies which found no association between acetaminophen therapy and BPD [89,90,91]. Additionally, data from the Experiences in Timing and Choices for Ductal Closure in Patent Ductus Arteriosus (INTERPDA) Study Online Registry (Turkey) demonstrated significant differences in rates of death or BPD in infants treated with ibuprofen (IV/PO) or APAP (IV/PO), with the highest rates recorded in the group exposed to IV APAP (p < 0.04) [92]. Higher rates of BPD have also been observed following administration of multiple rounds of continuous intravenous (IV) infusion (54.4%) compared to IV bolus (26.1%, p < 0.001) [93]. As with all observational data, these studies may be subject to bias and unmeasured confounding. Nonetheless, the possible increased risk of BPD observed in some of these reports is hypothesis generating and supports further unbiased investigation of pulmonary outcomes following acetaminophen therapy.
Do data from studies comparing different acetaminophen treatment regimens for PDA suggest potential harm with increased exposure?
A single center randomized non-inferiority trial conducted in infants born less than 30 weeks’ gestational age (n = 102) compared outcomes among those who received conventional (15 mg/kg q6 ×5 days) versus low dose (10 mg/kg q6 ×3 days) acetaminophen [94]. Rates of treatment failure (continued PDA) were similar between the groups, but BPD at 36 weeks’ PMA was more common among those assigned to conventional versus low dose therapy (46% vs. 35%) [94]. Although this finding was not statistically significant (p = 0.28) [94], a true risk difference of 11% would be consistent with clinically significant harm and suggest a worrisome dose-response relationship. Higher rates of BPD have also been observed following administration of multiple rounds of continuous intravenous (IV) infusion compared to IV bolus [93]. Of note, the infants who received continuous IV infusion, despite receiving a significantly higher cumulative dose of acetaminophen, had lower rates of PDA closure [93]. The extent to which more frequent treatment failure or increased acetaminophen exposure may have contributed to the higher rates of BPD is unknown.
Similar hypothesis generating data have been reported with IV comparted to enteral dosing. In contrast to enteral dosing, IV administration avoids hepatic first-pass metabolism and may increase direct exposure of pulmonary CYPE1 expressing cells to APAP. If a consistent signal for harm with IV vs. PO exposure is observed, further work is justified to interrogate this potential relationship. One study that was underpowered to detect potential harm demonstrated that rates of BPD were 85% after IV therapy compared to 73% with enteral therapy, but the difference was not statistically significant (p = 0.67) [95]. Regardless, these few dose and route comparison studies highlight the need for more robust pharmacokinetic and pharmacodynamic analyses of acetaminophen in extremely preterm infants. Lastly, a recent report indicated acetaminophen exposure may acutely increase pulmonary vascular resistance in premature infants [96]. This finding requires confirmation in future studies, but raises the possibility of additional mechanistic contributors that may affect cardiopulmonary function in extremely preterm infants..
Remaining questions: issues unique to acetaminophen exposures in the NICU
Neonatologists have brought acetaminophen to the NICU and increased exposures among high-risk infants despite the absence of robust safety data. Several factors unique to the preterm population that potentially increase the toxicity of acetaminophen exposures have thus far not been considered. Many, if not all, neonates receiving acetaminophen to treat PDA are simultaneously treated with supplemental oxygen and are at risk for associated oxidative stress [97]. This exposure may increase mitochondrial dysfunction and deplete the already limited pulmonary glutathione stores present in the preterm lung [98]. Whether these factors act to increase sensitivity to NAPQI mitochondrial toxicity, and limit the ability to detoxify CYP2E1-derived acetaminophen metabolites is unknown.
The clinical implications of acetaminophen metabolism that are unique to the preterm population remain relatively unexplored. Acetaminophen metabolites in neonates born less than 32 weeks’ gestational age exposed to 10, 15, or 20 mg/kg IV dosing have been reported [99]. In these neonates, levels of CYP2E1-derived APAP metabolites were significantly higher than in similarly exposed adults [99]. Elevated levels of the same CYP2E1-derived metabolites have been detected in neonates following supratherapeutic exposure to acetaminophen [(200 mg/kg × 4; [100] 600 mg/kg × 1 [101]] who did not develop detectable hepatotoxicity. In the absence of clinically obvious hepatotoxicity, these findings may be indicative of extra-hepatic CYP2E1 expression, acetaminophen metabolism and potential cellular toxicity is unknown. Whether these metabolites are derived from developmentally regulated, cell-type specific expression in the brain, kidney, or lung remains to be determined.
Conclusions
Acetaminophen exposures in the high risk, extremely preterm population are rapidly increasing. This practice change may carry unanticipated adverse effects. The available randomized trials and observational studies examining the safety and efficacy of acetaminophen to treat the PDA are marked by significant weakness including low sample sizes, potential bias, and lack of representation of the smallest and sickest infants at the highest risk of pulmonary morbidity. Although some observational data raise concern for potential increase in risk of BPD with acetaminophen therapy, it is unclear whether this represents a true causal relationship or is attributable to unmeasured confounding. While this review has been focused on acetaminophen for PDA, its use as an analgesic for preterm infants is also increasing and the pulmonary implications of these exposures similarly deserve further study. Indeed, a recent a large single center report found that that rates of grade 3 BPD increased significantly following implementation of routine IV acetaminophen administration to treat pain and discomfort associated with NICU care [102].
Preclinical data that support the safety of acetaminophen are lacking. The absence of observed hepatoxicity with acetaminophen use in preterm infants is reassuring, but is predictable given the results from preclinical studies and what is known about developmentally regulated hepatic CYP2E1 expression. Concerningly, however, the preclinical data point to the lungs of preterm infants as a particular site of potential acetaminophen induced injury. Moreover, the outcome of BPD, which is one of the few pulmonary outcomes that has been assessed after acetaminophen therapy in extremely preterm infants, provides incomplete insight into potential adverse drug effects predicted by the preclinical models. Studies interrogating potential mechanisms linking cell-specific CYP2E1 expression and acetaminophen-induced toxicity and robust assessment of pulmonary outcomes measured both before and after when BPD is diagnosed must be performed in large scale trials evaluating the safety and efficacy of acetaminophen in extremely preterm infants. Such efforts will ensure a thoughtful and thorough research approach to understanding the safety and efficacy of acetaminophen in our most vulnerable patients. Fortunately, large clinical trials of acetaminophen for PDA closure in high-risk preterm neonates are ongoing. These studies will hopefully provide meaningful data on the risks and benefits of this increasingly used but incompletely studied therapy [103, 104].
References
Backes CH, Hill KD, Shelton EL, Slaughter JL, Lewis TR, Weisz DE, et al. Patent ductus arteriosus: a contemporary perspective for the pediatric and adult cardiac care provider. J Am Heart Assoc. 2022;11:e025784.
Semberova J, Sirc J, Miletin J, Kucera J, Berka I, Sebkova S, et al. Spontaneous closure of patent ductus arteriosus in infants ≤1500 g. Pediatrics. 2017;140:e20164258.
Benitz WE. Committee on Fetus and Newborn, American Academy of Pediatrics. Patent ductus arteriosus in preterm infants. Pediatrics. 2016;137. https://doi.org/10.1542/peds.2015-3730.
Nelson RJ, Thibeault DW, Emmanouilides GC, Lippmann M. Improving the results of ligation of patent ductus arteriosus in small preterm infants. J Thorac Cardiovasc Surg. 1976;71:169–78.
Hammerman C, Bin-Nun A, Markovitch E, Schimmel MS, Kaplan M, Fink D. Ductal closure with paracetamol: a surprising new approach to patent ductus arteriosus treatment. Pediatrics. 2011;128:e1618–e1621.
Manalastas M, Zaheer F, Nicoski P, Weiss MG, Amin S. Acetaminophen therapy for persistent patent ductus arteriosus. NeoReviews. 2021;22:e320–e331.
Stark A, Smith PB, Hornik CP, Zimmerman KO, Hornik CD, Pradeep S, et al. Medication use in the neonatal intensive care unit and changes from 2010 to 2018. J Pediatr. 2022;240:66–71.e4.
Al-Turkait A, Szatkowski L, Choonara I, Ojha S. Drug utilisation in neonatal units in England and Wales: a national cohort study. Eur J Clin Pharm. 2022;78:669–77.
Gouyon B, Martin-Mons S, Iacobelli S, Razafimahefa H, Kermorvant-Duchemin E, Brat R, et al. Characteristics of prescription in 29 Level 3 Neonatal Wards over a 2-year period (2017-8). An inventory for future research. PLOS One. 2019;14:e0222667.
Liebowitz M, Kaempf J, Erdeve O, Bulbul A, Håkansson S, Lindqvist J, et al. Comparative effectiveness of drugs used to constrict the patent ductus arteriosus: a secondary analysis of the PDA-TOLERATE trial (NCT01958320). J Perinatol. 2019;39:599–607.
Dani C, Mosca F, Cresi F, Lago P, Lista G, Laforgia N, et al. Patent ductus arteriosus in preterm infants born at 23–24 weeks’ gestation: Should we pay more attention? Early Hum Dev. 2019;135:16–22.
Mukherjee A, Jadhav V, Gupta A. Off-label use of paracetamol in managing patent ductus arteriosus across neonatal intensive care units in the UK. Arch Dis Child Fetal Neonatal Ed. 2021;106:113–4.
Dowd LA, Wheeler BJ, Al-Sallami HS, Broadbent RS, Edmonds LK, Medlicott NJ. Paracetamol treatment for patent ductus arteriosus: practice and attitudes in Australia and New Zealand. J Matern Fetal Neonatal Med. 2019;32:3039–44.
Jasani B, Mitra S, Shah PS. Paracetamol (acetaminophen) for patent ductus arteriosus in preterm or low birth weight infants. Cochrane Database Syst Rev. 2022, https://doi.org/10.1002/14651858.CD010061.pub5.
Jasani B, Weisz DE, McNamara PJ. Evidence-based use of acetaminophen for hemodynamically significant ductus arteriosus in preterm infants. Semin Perinatol. 2018;42:243–52.
Squires RH. Acute liver failure in children. Semin Liver Dis. 2008;28:153–66.
Lee WS, McKiernan P, Kelly DA. Etiology, outcome and prognostic indicators of childhood fulminant hepatic failure in the United Kingdom. J Pediatr Gastroenterol Nutr. 2005;40:575–81.
Squires RH, Shneider BL, Bucuvalas J, Alonso E, Sokol RJ, Narkewicz MR, et al. Acute liver failure in children: the first 348 patients in the pediatric acute liver failure study group. J Pediatr. 2006;148:652–658.e2.
Wright CJ. Acetaminophen and the developing lung: could there be lifelong consequences? J Pediatr. 2021;235:264–276.e1.
McCulley DJ, Jensen EA, Sucre JMS, McKenna S, Sherlock LG, Dobrinskikh E, et al. Racing against time: leveraging preclinical models to understand pulmonary susceptibility to perinatal acetaminophen exposures. Am J Physiol Lung Cell Mol Physiol. 2022;323:L1–L13.
Pacifici GM, Allegaert K. Clinical pharmacology of paracetamol in neonates: a review. Curr Ther Res. 2015;77:24–30.
McGill MR, Jaeschke H. Metabolism and disposition of acetaminophen: recent advances in relation to hepatotoxicity and diagnosis. Pharm Res. 2013;30:2174–87.
Ramachandran A, Jaeschke H. Acetaminophen toxicity: novel insights into mechanisms and future perspectives. Gene Expr. 2018;18:19–30.
Yoon E, Babar A, Choudhary M, Kutner M, Pyrsopoulos N. Acetaminophen-induced hepatotoxicity: a comprehensive update. J Clin Transl Hepatol. 2016;4:131–42.
Sandoval J, Orlicky DJ, Allawzi A, Butler B, Ju C, Phan CT, et al. Toxic acetaminophen exposure induces distal lung ER stress, proinflammatory signaling, and emphysematous changes in the adult murine lung. Oxid Med Cell Longev. 2019;2019:1–15.
Lee WM. Acetaminophen (APAP) hepatotoxicity—Isn’t it time for APAP to go away? J Hepatol. 2017;67:1324–31.
Robinson JF, Hamilton EG, Lam J, Chen H, Woodruff TJ. Differences in cytochrome p450 enzyme expression and activity in fetal and adult tissues. Placenta. 2020;100:35–44.
Sadler NC, Nandhikonda P, Webb-Robertson B-J, Ansong C, Anderson LN, Smith JN, et al. Hepatic cytochrome p450 activity, abundance, and expression throughout human development. Drug Metab Dispos. 2016;44:984–91.
Hines RN. The ontogeny of drug metabolism enzymes and implications for adverse drug events. Pharm Ther. 2008;118:250–67.
Hart SN, Cui Y, Klaassen CD, Zhong X. Three patterns of cytochrome P450 gene expression during liver maturation in mice. Drug Metab Dispos. 2009;37:116–21.
Xu S-F, Hu A-L, Xie L, Liu J-J, Wu Q, Liu J. Age-associated changes of cytochrome P450 and related phase-2 gene/proteins in livers of rats. PeerJ. 2019;7:e7429.
Green MD, Shires TK, Fischer LJ. Hepatotoxicity of acetaminophen in neonatal and young rats. I. Age-related changes in susceptibility. Toxicol Appl Pharm. 1984;74:116–24.
Hart JG, Timbrell JA. The effect of age on paracetamol hepatotoxicity in mice. Biochem Pharm. 1979;28:3015–7.
Oesch F, Fabian E, Landsiedel R. Xenobiotica-metabolizing enzymes in the lung of experimental animals, man and in human lung models. Arch Toxicol. 2019;93:3419–89.
Hukkanen J, Pelkonen O, Hakkola J, Raunio H. Expression and regulation of xenobiotic-metabolizing cytochrome P450 (CYP) enzymes in human lung. Crit Rev Toxicol. 2002;32:391–411.
Hart SG, Cartun RW, Wyand DS, Khairallah EA, Cohen SD. Immunohistochemical localization of acetaminophen in target tissues of the CD-1 mouse: correspondence of covalent binding with toxicity. Fundam Appl Toxicol J Soc Toxicol. 1995;24:260–74.
Placke ME, Wyand DS, Cohen SD. Extrahepatic lesions induced by acetaminophen in the mouse. Toxicol Pathol. 1987;15:381–7.
Jeffery EH, Haschek WM. Protection by dimethylsulfoxide against acetaminophen-induced hepatic, but not respiratory toxicity in the mouse. Toxicol Appl Pharm. 1988;93:452–61.
Gu J, Cui H, Behr M, Zhang L, Zhang Q-Y, Yang W, et al. In vivo mechanisms of tissue-selective drug toxicity: effects of liver-specific knockout of the NADPH-cytochrome P450 reductase gene on acetaminophen toxicity in kidney, lung, and nasal mucosa. Mol Pharm. 2005;67:623–30.
Neff SB, Neff TA, Kunkel SL, Hogaboam CM. Alterations in cytokine/chemokine expression during organ-to-organ communication established via acetaminophen-induced toxicity. Exp Mol Pathol. 2003;75:187–93.
Bartolone JB, Beierschmitt WP, Birge RB, Hart SG, Wyand S, Cohen SD, et al. Selective acetaminophen metabolite binding to hepatic and extrahepatic proteins: an in vivo and in vitro analysis. Toxicol Appl Pharm. 1989;99:240–9.
Chen TS, Richie JP, Lang CA. Life span profiles of glutathione and acetaminophen detoxification. Drug Metab Dispos Biol Fate Chem. 1990;18:882–7.
Micheli L, Cerretani D, Fiaschi AI, Giorgi G, Romeo MR, Runci FM. Effect of acetaminophen on glutathione levels in rat testis and lung. Environ Health Perspect. 1994;102:63–64.
Bulera SJ, Cohenb SD, Khairallah EA. Acetaminophen-arylated proteins are detected in hepatic subcellular fractions and numerous extra-hepatic tissues in CD-1 and C57B1/6J mice. Toxicology. 1996;109:85–99.
Dimova S, Hoet PHM, Dinsdale D, Nemery B. Acetaminophen decreases intracellular glutathione levels and modulates cytokine production in human alveolar macrophages and type II pneumocytes in vitro. Int J Biochem Cell Biol. 2005;37:1727–37.
Dimova S, Hoet PH, Nemery B. Paracetamol (acetaminophen) cytotoxicity in rat type II pneumocytes and alveolar macrophages in vitro. Biochem Pharm. 2000;59:1467–75.
Nassini R, Materazzi S, Andrè E, Sartiani L, Aldini G, Trevisani M, et al. Acetaminophen, via its reactive metabolite N -acetyl- p -benzo-quinoneimine and transient receptor potential ankyrin-1 stimulation, causes neurogenic inflammation in the airways and other tissues in rodents. FASEB J. 2010;24:4904–16.
Dobrinskikh E, Al-Juboori SI, Zarate MA, Zheng L, De Dios R, Balasubramaniyan D, et al. Pulmonary implications of acetaminophen exposures independent of hepatic toxicity. Am J Physiol Lung Cell Mol Physiol. 2021;321:L941–L953.
Breen K, Wandscheer JC, Peignoux M, Pessayre D. In situ formation of the acetaminophen metabolite covalently bound in kidney and lung. Supportive evidence provided by total hepatectomy. Biochem Pharm. 1982;31:115–6.
Dobrinskikh E, Sherlock LG, Orlicky DJ, Zheng L, De Dios R, Balasubramaniyan D, et al. The developing murine lung is susceptible to acetaminophen toxicity. Am J Physiol Lung Cell Mol Physiol. 2021;320:L969–L978.
LungMAP - Home. https://www.lungmap.net/. Accessed 4 Mar 2023.
Xie T, Wang Y, Deng N, Huang G, Taghavifar F, Geng Y, et al. Single-cell deconvolution of fibroblast heterogeneity in mouse pulmonary fibrosis. Cell Rep. 2018;22:3625–40.
Negretti NM, Plosa EJ, Benjamin JT, Schuler BA, Habermann AC, Jetter CS, et al. A single-cell atlas of mouse lung development. Dev Camb Engl. 2021;148:dev199512.
Boström H, Willetts K, Pekny M, Levéen P, Lindahl P, Hedstrand H, et al. PDGF-A signaling is a critical event in lung alveolar myofibroblast development and alveogenesis. Cell. 1996;85:863–73.
Li R, Li X, Hagood J, Zhu M-S, Sun X. Myofibroblast contraction is essential for generating and regenerating the gas-exchange surface. J Clin Invest. 2020;130:2859–71.
Li C, Li M, Li S, Xing Y, Yang C-Y, Li A, et al. Progenitors of secondary crest myofibroblasts are developmentally committed in early lung mesoderm. Stem Cells. 2015;33:999–1012.
Lindahl P, Karlsson L, Hellström M, Gebre-Medhin S, Willetts K, Heath JK, et al. Alveogenesis failure in PDGF-A-deficient mice is coupled to lack of distal spreading of alveolar smooth muscle cell progenitors during lung development. Dev Camb Engl. 1997;124:3943–53.
Gouveia L, Betsholtz C, Andrae J. Expression analysis of platelet-derived growth factor receptor alpha and its ligands in the developing mouse lung. Physiol Rep. 2017;5:e13092.
Soriano P. The PDGF alpha receptor is required for neural crest cell development and for normal patterning of the somites. Dev Camb Engl. 1997;124:2691–2700.
Li R, Bernau K, Sandbo N, Gu J, Preissl S, Sun X. Pdgfra marks a cellular lineage with distinct contributions to myofibroblasts in lung maturation and injury response. eLife. 2018;7:e36865.
Du Y, Guo M, Whitsett JA, Xu Y. ‘LungGENS’: a web-based tool for mapping single-cell gene expression in the developing lung. Thorax. 2015;70:1092–4.
Du Y, Kitzmiller JA, Sridharan A, Perl AK, Bridges JP, Misra RS, et al. Lung Gene Expression Analysis (LGEA): an integrative web portal for comprehensive gene expression data analysis in lung development. Thorax. 2017;72:481–4.
Du Y, Ouyang W, Kitzmiller JA, Guo M, Zhao S, Whitsett JA, et al. Lung gene expression analysis web portal version 3: lung-at-a-glance. Am J Respir Cell Mol Biol. 2021;64:146–9.
Bandoli G, Palmsten K, Chambers C. Acetaminophen use in pregnancy: examining prevalence, timing, and indication of use in a prospective birth cohort. Paediatr Perinat Epidemiol. 2020;34:237–46.
International Agency for Research on Cancer. Pharmaceutical drugs: views and experts opinions of an IARC Working Group on the Evaluation of Carcinogenic Risks to Humans, which met in Lyon 17–24 Oct. 1989. Lyon: IARC; 1990.
Collins E. Maternal and fetal effects of acetaminophen and salicylates in pregnancy. Obstet Gynecol. 1981;58:57S–62S.
Niederhoff H, Zahradnik HP. Analgesics during pregnancy. Am J Med. 1983;75:117–20.
Lubawy WC, Garrett RJ. Effects of aspirin and acetaminophen on fetal and placental growth in rats. J Pharm Sci. 1977;66:111–3.
Lambert GH, Thorgeirsson SS. Glutathione in the developing mouse liver—I. Biochem Pharm. 1976;25:1777–81.
Bauer AZ, Swan SH, Kriebel D, Liew Z, Taylor HS, Bornehag C-G, et al. Paracetamol use during pregnancy — a call for precautionary action. Nat Rev Endocrinol. 2021;17:757–66.
Statement on the use of acetaminophen for analgesia and fever in pregnancy. https://sogc.org/en/content/featured-news/Statement_on_the_use_of_acetaminophen.aspx?WebsiteKey=4d1aa07b-5fc4-4673-9721-b91ff3c0be30. Accessed 4 Mar 2023.
ACOG Response to Consensus Statement on Paracetamol Use During Pregnancy | ACOG. https://www.acog.org/news/news-articles/2021/09/response-to-consensus-statement-on-paracetamol-use-during-pregnancy. Accessed 5/9/2023.
Official ENTIS Position Statement: Paracetamol (acetaminophen, APAP) use in pregnancy – ENTIS. https://www.entis-org.eu/entis-news/official-entis-position-statement-paracetamol-acetaminophen-apap-use-in-pregnancy. Accessed 4 Mar 2023.
Shaheen SO. Paracetamol use in pregnancy and wheezing in early childhood. Thorax. 2002;57:958–63.
Shaheen SO, Sterne JA, Songhurst CE, Burney PG. Frequent paracetamol use and asthma in adults. Thorax. 2000;55:266–70.
Newson RB, Shaheen SO, Chinn S, Burney PG. Paracetamol sales and atopic disease in children and adults: an ecological analysis. Eur Respir J. 2000;16:817–23.
Fan G, Wang B, Liu C, Li D. Prenatal paracetamol use and asthma in childhood: a systematic review and meta-analysis. Allergol Immunopathol. 2017;45:528–33.
Sakulchit T, Goldman RD. Acetaminophen use and asthma in children. Can Fam Physician Med Fam Can. 2017;63:211–3.
Singh M, Varukolu S, Chauhan A, Jaiswal N, Pradhan P, Mathew JL, et al. Paracetamol exposure and asthma: what does the evidence say? An overview of systematic reviews. Pediatr Pulmonol. 2021;56:3189–99.
Kluckow M, Carlisle H, Broom M, Woods P, Jeffery M, Desai D, et al. A pilot randomised blinded placebo-controlled trial of paracetamol for later treatment of a patent ductus arteriosus. J Perinatol. 2019;39:102–7.
Davidson JM, Ferguson J, Ivey E, Philip R, Weems MF, Talati AJ. A randomized trial of intravenous acetaminophen versus indomethacin for treatment of hemodynamically significant PDAs in VLBW infants. J Perinatol. 2021;41:93–99.
Schindler T, Smyth J, Bolisetty S, Michalowski J, Mallitt K-A, Singla A, et al. Early PARacetamol (EPAR) trial: a randomized controlled trial of early paracetamol to promote closure of the ductus arteriosus in preterm infants. Neonatology. 2021;118:274–81.
Hochwald O, Mainzer G, Borenstein-Levin L, Jubran H, Dinur G, Zucker M, et al. Adding paracetamol to ibuprofen for the treatment of patent ductus arteriosus in preterm infants: a double-blind, randomized, placebo-controlled pilot study. Am J Perinatol. 2018;35:1319–25.
Oncel MY, Yurttutan S, Erdeve O, Uras N, Altug N, Oguz SS, et al. Oral paracetamol versus oral ibuprofen in the management of patent ductus arteriosus in preterm infants: a randomized controlled trial. J Pediatr. 2014;164:510–514.e1.
Patel R, Sushko K, van den Anker J, Samiee-Zafarghandy S. Long-term safety of prenatal and neonatal exposure to paracetamol: a systematic review. Int J Environ Res Public Health. 2022;19:2128.
Luecke CM, Liviskie CJ, Zeller BN, Vesoulis ZA, McPherson C. Acetaminophen for patent ductus arteriosus in extremely low-birth-weight neonates. J Pediatr Pharm Ther. 2017;22:461–6.
Sehgal A, Nitzan I, Krishnamurthy MB, Pharande P, Tan K. Toward rational management of patent ductus arteriosus: ductal disease staging and first line paracetamol. J Matern Fetal Neonatal Med. 2021;34:3940–5.
Mashally S, Nield LE, McNamara PJ, Martins FF, El-Khuffash A, Jain A, et al. Late oral acetaminophen versus immediate surgical ligation in preterm infants with persistent large patent ductus arteriosus. J Thorac Cardiovasc Surg. 2018;156:1937–44.
Karabulut B, Paytoncu S. Efficacy and safety of oral paracetamol vs. oral ibuprofen in the treatment of symptomatic patent ductus arteriosus in premature infants. Pediatr Drugs. 2019;21:113–21.
Vaidya R, Wilson D, Paris Y, Madore L, Singh R. Use of acetaminophen for patent ductus arteriosus treatment: a single center experience. J Matern Fetal Neonatal Med. 2020;33:2723–9.
Kimani S, Surak A, Miller M, Bhattacharya S. Use of combination therapy with acetaminophen and ibuprofen for closure of the patent ductus arteriosus in preterm neonates. Paediatr Child Health. 2021;26:e177–e183.
Okulu E, Erdeve O, Arslan Z, Demirel N, Kaya H, Gokce IK, et al. An observational, prospective, multicenter, registry-based cohort study comparing conservative and medical management for patent ductus arteriosus. Front Pediatr. 2020;8:434.
Cakir U, Tayman C, Karacaglar NB, Beser E, Ceran B, Unsal H. Comparison of the effect of continuous and standard intermittent bolus paracetamol infusion on patent ductus arteriosus. Eur J Pediatr. 2021;180:433–40.
Balasubramanian H, Jain V, Bhalgat P, Parikh S, Kabra N, Mohan D, et al. Low dose paracetamol for management of patent ductus arteriosus in very preterm infants: a randomised non-inferiority trial. Arch Dis Child Fetal Neonatal Ed. 2023;108:130–5.
Gover A, Levy PT, Rotschild A, Golzman M, Molad M, Lavie-Nevo K, et al. Oral versus intravenous paracetamol for patent ductus arteriosus closure in preterm infants. Pediatr Res. 2022;92:1146–52.
Murphy C, Bussmann N, Staunton D, McCallion N, Franklin O, EL-Khuffash A. The effect of patent ductus arteriosus treatment with paracetamol on pulmonary vascular resistance. J Perinatol. 2022;42:1697–8.
Torres-Cuevas I, Parra-Llorca A, Sánchez-Illana A, Nuñez-Ramiro A, Kuligowski J, Cháfer-Pericás C, et al. Oxygen and oxidative stress in the perinatal period. Redox Biol. 2017;12:674–81.
Berkelhamer SK, Farrow KN. Developmental regulation of antioxidant enzymes and their impact on neonatal lung disease. Antioxid Redox Signal. 2014;21:1837–48.
Flint RB, Roofthooft DW, van Rongen A, van Lingen RA, van den Anker JN, van Dijk M, et al. Exposure to acetaminophen and all its metabolites upon 10, 15, and 20 mg/kg intravenous acetaminophen in very-preterm infants. Pediatr Res. 2017;82:678–84.
Abadier M, Wong A, Stathakis P, Singsit J, Pillay M, Graudins A. A case of accidental neonatal paracetamol overdose with prolonged half-life and measured metabolites. Clin Toxicol. 2019;57:1154–6.
Chiewa A, Buckleyb N, Paoc LT, Robertsd D. Pharmacokinetics of acetaminophen and metabolites after accidental acute overdose in a neonate. North American Congress of Clinical Toxicology (NACCT) Abstracts 2018. Clin Toxicol. 2018;56:951–2.
Juujärvi S, Saarela T, Pokka T, Hallman M, Aikio O. Intravenous paracetamol for neonates: long-term diseases not escalated during 5 years of follow-up. Arch Dis Child Fetal Neonatal Ed. 2021;106:178–83.
Prophylactic Treatment of the Ductus Arteriosus in Preterm Infants by Acetaminophen - Full Text View - ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT04459117. Accessed 4 Mar 2023.
Canadian National PDA Treatment Study - Full Text View - ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT04347720. Accessed 4 Mar 2023.
Funding
This work was supported by NIH grants R01HL132941 and R01HD107700 to CJW, NIH grant R01HL146859 to DJM.
Author information
Authors and Affiliations
Contributions
CJW and EAJ responsible for the initial draft. CJW, EAJ, DJM and SM edited the draft. All authors agree with submission of final version.
Corresponding author
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Wright, C.J., McCulley, D.J., Mitra, S. et al. Acetaminophen for the patent ductus arteriosus: has safety been adequately demonstrated?. J Perinatol 43, 1230–1237 (2023). https://doi.org/10.1038/s41372-023-01697-2
Received:
Revised:
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1038/s41372-023-01697-2