Abstract
Clinical chorioamnionitis, the most common infection-related diagnosis in labor and delivery units, is an antecedent of puerperal infection and neonatal sepsis. The condition is suspected when intrapartum fever is associated with two other maternal and fetal signs of local or systemic inflammation (eg, maternal tachycardia, uterine tenderness, maternal leukocytosis, malodorous vaginal discharge or amniotic fluid, and fetal tachycardia). Clinical chorioamnionitis is a syndrome caused by intraamniotic infection, sterile intraamniotic inflammation (inflammation without bacteria), or systemic maternal inflammation induced by epidural analgesia. In cases of uncertainty, a definitive diagnosis can be made by analyzing amniotic fluid with methods to detect bacteria (Gram stain, culture, or microbial nucleic acid) and inflammation (white blood cell count, glucose concentration, interleukin-6, interleukin-8, matrix metalloproteinase-8). The most common microorganisms are Ureaplasma species, and polymicrobial infections occur in 70% of cases. The fetal attack rate is low, and the rate of positive neonatal blood cultures ranges between 0.2% and 4%. Intrapartum antibiotic administration is the standard treatment to reduce neonatal sepsis. Treatment with ampicillin and gentamicin have been recommended by professional societies, although other antibiotic regimens, eg, cephalosporins, have been used. Given the importance of Ureaplasma species as a cause of intraamniotic infection, consideration needs to be given to the administration of antimicrobial agents effective against these microorganisms such as azithromycin or clarithromycin. We have used the combination of ceftriaxone, clarithromycin, and metronidazole, which has been shown to eradicate intraamniotic infection with microbiologic studies. Routine testing of neonates born to affected mothers for genital mycoplasmas can improve the detection of neonatal sepsis. Clinical chorioamnionitis is associated with decreased uterine activity, failure to progress in labor, and postpartum hemorrhage; however, clinical chorioamnionitis by itself is not an indication for cesarean delivery. Oxytocin is often administered for labor augmentation, and it is prudent to have uterotonic agents at hand to manage postpartum hemorrhage. Infants born to mothers with clinical chorioamnionitis near term are at risk for early-onset neonatal sepsis and for long-term disability such as cerebral palsy. A frontier is the noninvasive assessment of amniotic fluid to diagnose intraamniotic inflammation with a transcervical amniotic fluid collector and a rapid bedside test for IL-8 for patients with ruptured membranes. This approach promises to improve diagnostic accuracy and to provide a basis for antimicrobial administration.
Keywords: abnormal fetal heart rate pattern, adverse maternal outcome, adverse neonatal outcome, amniocentesis, amniotic fluid, ampicillin, antibiotics, antipyretics, azithromycin, Batson’s plexus, biomarker, carbapenem, cerebral palsy, chemokine, clarithromycin, clavulanic acid, cytokine, epidural, fetal heart rate tracing, fetal inflammatory response syndrome, fetal tachycardia, fever, funisitis, Gardnerella vaginalis, genital mycoplasma, gentamycin, histologic chorioamnionitis, infection, inflammasome, interleukin-6 (IL-6);, interleukin-8 (IL-8);, intraamniotic infection, intraamniotic inflammation, intrapartum fever, maternal infection, maternal leukocytosis, maternal N-acetylcysteine (NAC), matrix metalloproteinase-8 (MMP-8), NLRP3 inflammasome, neonatal bacteremia, neonatal sepsis, overshoot, piperacillin, postpartum hemorrhage, pyrogenic, sterile intraamniotic inflammation, tazobactam, ticarcillin, Ureaplasma species
Introduction
Clinical chorioamnionitis, a syndrome defined by the presence of maternal and fetal signs of local and systemic inflammation, is mainly caused by intraamniotic infection or by sterile intraamniotic inflammation.1, 2 This condition is the most common infection-related diagnosis in labor and delivery units, affecting 1% to 6% of term gestations.3–10 Moreover, the disorder is an antecedent of puerperal infections such as endometritis,11–13 pelvic peritonitis,14 wound infections,4, 13, 15 pelvic abscess,4 septic pelvic thrombophlebitis,4,16 and necrotizing fasciitis.17,18 Clinical chorioamnionitis is the leading cause of maternal 19–22 and neonatal sepsis.4,23–26 Patients with this diagnosis are at risk for uterine atony, postpartum hemorrhage, and other complications listed in Table 1.4,9,10
Table 1.
Adverse maternal outcomes in relationship to the diagnosis of clinical chorioamnionitis at term
Adverse maternal outcomes | No clinical chorioamnionitis(N = 14,685) | Clinical chorioamnionitis(N = 1,965) | RR(95% CI) |
---|---|---|---|
Uterine atony | 5.7% | 14.2% | 2.50 (2.20–2.84) |
Blood transfusion | 2.0% | 4.5% | 2.25 (1.78–2.83) |
Pelvic abscess | 0.1% | 0.3% | 3.74 (1.28–10.92) |
Septic pelvic thrombophlebitis | 0.2% | 0.6% | 2.74 (1.38–5.46) |
Thromboembolic disease | 0.1% | 0.1% | 1.49 (0.33–6.82) |
Wound complication | 1.1% | 1.5% | 1.39 (0.95–2.05) |
Necrotizing fasciitis | 0.0% | 0.1% | 2.49 (0.26–23.93) |
ICU admission | 0.5% | 0.6% | 1.03 (0.55–1.93) |
Exploratory laparotomy | 0.2% | 0.3% | 1.07 (0.42–2.72) |
Hysterectomy | 0.3% | 0.4% | 1.57 (0.74–3.37) |
Death | 0.1% | 0.0% | – |
CI: confidence interval; ICU: intensive care unit; RR: relative risk.
Adapted from Rouse DJ, Landon M, Leveno KJ, Leindecker S, Varner MW, Caritis SN, et al. The Maternal-Fetal Medicine Units cesarean registry: chorioamnionitis at term and its duration-relationship to outcomes. Am J Obstet Gynecol. 2004;191(1):211–6.4
The term “clinical” distinguishes this entity from “histologic” chorioamnionitis, which is diagnosed by pathologic examination of the placenta (Figure 1).27 Often, the term “intraamniotic infection” is used interchangeably with “clinical chorioamnionitis”—this is unfortunate because they are not synonymous. The designation of intraamniotic infection should be reserved for cases in which there is laboratory evidence of microorganisms in the amniotic fluid coupled with intraamniotic inflammation. This article reviews advances in the understanding of clinical chorioamnionitis at term and its association with intraamniotic infection, focusing on the definition, pathogenesis, microbiology, inflammatory response, diagnosis, and treatment.
Figure 1. Definitions of chorioamnionitis: clinical, histologic, and microbiologic.
Chorioamnionitis can be defined as a clinical or a histopathologic identity or from a microbiologic point of view: (1) the clinical diagnosis of chorioamnionitis is based on clinical signs, eg, maternal fever, uterine tenderness, malodorous discharge, and maternal and fetal tachycardia, and on laboratory abnormalities, ie, leukocytosis; (2) histologic chorioamnionitis is diagnosed by histopathologic examination of the placenta; the characteristic morphologic feature of acute histologic chorioamnionitis is diffuse infiltration of neutrophils into the chorioamniotic membranes; and (3) microbiologic chorioamnionitis, or intraamniotic infection, refers to the presence of microorganisms in the amniotic fluid, retrieved by amniocentesis. These terms are not synonymous, and confusion is caused when they are used interchangeably.
The amniotic cavity does not contain bacteria in normal pregnancy
Under normal circumstances, the fetus lives in an environment devoid of bacteria28, 29 Therefore, birth represents a critical stage for the acquisition of the first microbiota (the collection of living microorganisms present in a defined site). During the passage through the birth canal, the fetus is exposed to the vaginal microbiota,30–32 and these bacteria become the pioneer microorganisms that invade the formerly sterile body of the infant,33 thus establishing the first neonatal microbiota.34,35 This microbiota differs according to the mode of delivery (vaginal vs cesarean delivery)36–38 although mother-to-infant microbial transmission routes develop across several body sites after birth.39,40
Microbial invasion of the amniotic cavity (MIAC) by bacteria, fungi, and viruses can predispose to fetal infection. Microorganisms can enter the fetus through different sites, ie, the mucous membranes of the airway, gastrointestinal tract, tympanic membrane, conjunctiva, or skin. Once bacteria are in contact with the mucous membranes, microorganisms can gain access to the fetal circulation and cause bacteremia, which can lead to sepsis, septic shock, and fetal death.41 The fetal immune system is capable of mounting innate and adaptive immune responses, which are key in host defense against infection.42–47 Indeed, recent studies show that the amniotic fluid harbors cells of the innate immune system, largely of fetal origin, that participate in host defense against microbes invading the amniotic cavity.48–56
Nomenclature of conditions related to the presence or absence of microorganisms and intraamniotic inflammation
Microbial invasion of the amniotic cavity is defined as the presence of microorganisms in amniotic fluid retrieved by transabdominal amniocentesis or at the time of cesarean delivery and detected by cultivation or molecular microbiologic techniques (Figure 2). The term MIAC indicates the presence of microorganisms regardless of the presence or absence of an inflammatory response. Intraamniotic inflammation refers to the presence of an inflammatory response in the amniotic cavity, which can be detected by the presence of inflammatory cells such as neutrophils57–61 or by an elevated concentration of an inflammatory mediator, eg, interleukin (IL)-658−60, 62–69 or matrix metalloproteinase (MMP)-8.70 A wide range of inflammatory products can be detected in biological fluids, and their concentrations can be used for diagnostic purposes.. The most frequently employed analytes in the diagnosis of intraamniotic inflammation are cytokines (IL-658−60,62–69,71 and IL-872−74), acute-phase reactants (C-reactive protein),61,75–79 or other compounds eg, MMP-8 (also known as neutrophil collagenase).70,80–82
Figure 2. Nomenclature of states related to intraamniotic inflammation and intraamniotic infection.
Microorganisms in amniotic fluid can be detected by cultivation methods and/or by molecular microbiologic techniques. Intraamniotic inflammation is defined by the presence of inflammatory cells (white blood cell count ≥ 50 cells/mm3) or by an elevated concentration of a biomarker of inflammation, eg, an interleukin-6 concentration ≥2.6 ng/mL or a matrix metalloproteinase-8 concentration >23 ng/mL. On the basis of the results of the presence of microorganisms in amniotic fluid and intraamniotic inflammation, patients can be classified into four subgroups; (1) no intraamniotic infection (negative amniotic fluid by culture and PCR and the absence of intra-amniotic inflammation); (2) microorganisms in amniotic fluid without intraamniotic inflammation (positive amniotic fluid by either culture or PCR but the absence of intraamniotic inflammation); (3) sterile intraamniotic inflammation (negative amniotic fluid by culture and PCR but the presence of intraamniotic inflammation); and (4) intraamniotic infection (positive amniotic fluid by either culture or PCR and the presence of intraamniotic inflammation).
PCR: polymerase chain reaction.
The term intraamniotic infection refers to the combination of MIAC and intraamniotic inflammation (Figure 2). Most cases in which microorganisms are present in amniotic fluid without evidence of inflammation are caused by specimen contamination with skin flora or with bacteria in the laboratory or in reagents.83 When intraamniotic inflammation is present in the absence of microorganisms, the condition is referred to as sterile intraamniotic inflammation.1, 2,84–87 This term should ideally be reserved for samples that are negative for microorganisms, ascertained by cultivation and molecular microbiologic techniques.1,2,84–86
Clinical chorioamnionitis refers to a syndrome characterized by the presence of a maternal fever (temperature ≥37.8°C or ≥38.0°C) and 2 or more of these 5 clinical signs: (1) maternal tachycardia (>100 beats/min); (2) fetal tachycardia (>160 beats/min); (3) uterine tenderness; (4) purulent or malodorous amniotic fluid or vaginal discharge; and (5) maternal leukocytosis (white blood cell count >15,000/mm3).20,88–95
Acute histologic chorioamnionitis indicates the presence of neutrophils in the chorioamniotic membranes or in the chorionic plate and represents a maternal host response,27,96–106 whereas funisitis and chorionic vasculitis are evidence of a fetal response (Figure 3). Funisitis is defined as inflammation of the umbilical cord (umbilical vein, artery, and Wharton’s jelly).107,108 Chorionic vasculitis consists of inflammation of the fetal vessels on the surface of the chorionic plate. Acute histologic chorioamnionitis is the pathologic expression of intraamniotic inflammation (Figure 3).97,99,109−115Acute histologic chorioamnionitis is not synonymous with intraamniotic infection. Only a fraction of patients diagnosed with histologic chorioamnionitis has proven intraamniotic infection. The rest have sterile intraamniotic inflammation, which may be accompanied by funisitis. The term acute chorioamnionitis refers to a specific inflammatory lesion in which neutrophils are the predominant cell type. By contrast, chronic chorioamnionitis refers to inflammatory lesions characterized by the infiltration of lymphocytes. Chronic inflammatory lesions occur when specific infectious agents, eg, viruses, are present, yet the main cause of chronic inflammatory lesions is maternal anti-fetal rejection.116–123 Interested readers are referred to a recent review by the authors on this subject.123
Figure 3. Anatomy of the placental disc and chorioamniotic membranes and acute inflammatory lesions of the placenta: chorioamnionitis, chorionic vasculitis, and funisitis.
A. The placenta is composed of 3 major structures: the chorioamniotic membranes, the placental disc, and the umbilical cord. Acute inflammatory lesions of the placenta are characterized by the infiltration of neutrophils in any of these structures. Specifically, when the inflammatory process affects the chorion and amnion, the term is acute chorioamnionitis. If the inflammatory process involves the umbilical cord (umbilical vein, umbilical artery, and the Wharton’s jelly), this is referred to as acute funisitis, the histologic counterpart of the fetal inflammatory response syndrome. B. Acute chorioamnionitis (stage 2 acute inflammation of the chorioamniotic membranes): neutrophilic migration into the amniotic connective tissue is shown (asterisk). C. Chorionic vasculitis is inflammation on the surface of the fetal vessels within the chorionic plate. Acute chorionic vasculitis (asterisk) is a stage 1 fetal inflammatory response. D. Funisitis: its characteristic feature is concentric, perivascular distribution of degenerated neutrophils (asterisk). Modified from Kim CJ et al. Acute chorioamnionitis and funisitis: definition, pathologic features, and clinical significance. Am J Obstet Gynecol. 2015;213(4 Suppl):S29–52.27
A panel proposed to replace the term “clinical chorioamnionitis” with the term “intrauterine inflammation or infection or both,” abbreviated as “Triple I.”124 However, this proposal has not been adopted because it implies that the inflammatory status of the amniotic cavity and the presence of microorganisms can be determined only by clinical signs or symptoms. There is solid evidence that this is not the case.125–127 Indeed, a subset of patients that meets the clinical criteria of chorioamnionitis does not show evidence of intraamniotic infection or intraamniotic inflammation.1, 2 Therefore, we favor the use of the term “clinical chorioamnionitis” to refer to the clinical syndrome before the analysis of amniotic fluid. Examination of amniotic fluid allows ascertainment of whether intraamniotic inflammation is present in patients with a fever and likely etiology, ie, microbial or sterile.
The definition of clinical chorioamnionitis according to the reVITALize Initiative (a USA multidisciplinary collaboration formed to standardize obstetrical data definitions) includes unexplained fever (>38°C or 100.4°F) with one or more of the following symptoms or signs: (1) uterine tenderness and irritability, (2) leukocytosis, (3) fetal tachycardia, (4) maternal tachycardia, and (5) malodorous vaginal discharge.128 Amniotic fluid analysis in patients with intact membranes requires an amniocentesis. If the membranes are ruptured, a transcervical amniotic fluid collector and an assessment of inflammatory products, eg, IL-8, makes possible the diagnosis of intraamniotic inflammation (for details, see the section on diagnosis).129, 130 Amniotic fluid could also be retrieved for microbiologic studies at the time of cesarean delivery.
Clinical chorioamnionitis as a syndrome
Patients diagnosed with clinical chorioamnionitis at term, can be categorized into 3 groups according to the results of amniotic fluid analysis of bacteria and inflammation: (1) intraamniotic infection, ie, the presence of microorganisms and intraamniotic inflammation, (2) sterile intraamniotic inflammation, and (3) maternal signs or symptoms of systemic inflammation without evidence of intraamniotic inflammation or intraamniotic infection.1, 2 The prevalence of the three conditions relates to the status of membrane rupture and the administration of an epidural.1, 2 The relative frequency of each of the three conditions is intraamniotic infection, 65%; sterile intraamniotic inflammation, 15%; and absence of intraamniotic inflammation and/or intraamniotic infection, 20% (Figure 4).1, 2 The etiology of maternal systemic inflammation in the latter is unclear and has been attributed to a neuroinflammatory process after the administration of epidural analgesia.
Figure 4. The syndrome of chorioamnionitis at term: intraamniotic infection and sterile intraamniotic inflammation.
Patients with clinical chorioamnionitis at term, when studied with molecular microbiologic techniques and assessed for intraamniotic inflammation, are grouped into 3 categories: (1) intraamniotic infection (presence of microorganisms and intraamniotic inflammation); (2) sterile intraamniotic inflammation; and (3) maternal signs or symptoms of systemic inflammation without evidence of intraamniotic infection. The relative frequencies of these 3 conditions were calculated as 65% (58/89) in intraamniotic infection; 15% (13/89) in intraamniotic inflammation; and 20% (18/89) for no evidence of intraamniotic infection.
Pathophysiology of intraamniotic infection
Microorganisms may gain access to the amniotic cavity through three main pathways131–133: (1) ascending from the vagina and cervix109, 134–137; (2) hematogenous dissemination through the placenta (transplacental infection)138–145; and (3) accidental introduction at the time of invasive procedures such as amniocentesis, percutaneous fetal blood sampling, chorionic villus sampling, or shunting.146–152
The most common pathway of intraamniotic infection is the ascending route131, 132, 137 based on the following evidence: (1) the nucleotide sequences of microorganisms grown in culture from amniotic fluid samples of patients with intraamniotic infection are similar to those from the vagina in the same patients137; (2) in twin gestations, acute histologic chorioamnionitis, more common in the first-born twin, is rarely observed in the second twin. Moreover, when intraamniotic infection is detected, the presenting sac is nearly always involved153; and (3) in virtually all cases of congenital pneumonia, inflammation of the chorioamniotic membranes is present.138, 154
Risk factors for clinical chorioamnionitis
Ruptured fetal membranes
The chorioamniotic membranes represent a physical and a biochemical barrier to microorganisms, as the fetal membranes produce antimicrobial peptides155–157 and contain cells of the innate immune system capable of protecting the host against bacteria.158–161 The frequency of MIAC in patients with ruptured membranes at term is 34%,162 which is higher than in those at term without labor (1%)163 or in those with spontaneous labor with intact membranes (19%) (Figure 5).163 In patients with clinical chorioamnionitis at term, the frequency of intraamniotic infection is higher when the membranes are ruptured than when they are intact [ruptured membranes, 78% vs intact membranes, 38%; p=0.01 (Figure 6)].2 Rupture of the membranes increases the risk of a postpartum uterine infection, eg, endometritis, endomyometritis, and endoparametritis.14, 164 The increased risk for postpartum endometritis conferred by rupture of the membranes has been documented in studies performed before the routine use of antimicrobial prophylaxis for cesarean delivery.165 In patients whose membranes had been ruptured for longer than six hours, bacteria were detected in all cases for which amniotic fluid had been obtained at the time of cesarean delivery; 95% of patients developed postpartum myometritis.165
Figure 5. Frequency of microorganisms in the amniotic cavity of patients at term without labor, patients in labor with intact membranes, and those with prelabor rupture of membranes at term.
In the presence of ruptured membranes, 34% of patients at term have microorganisms in amniotic fluid, which is higher compared to those without labor (1%) and to those with spontaneous labor with intact membranes (19%). Patients at term, not in labor, and with intact membranes rarely have bacteria detected in the amniotic cavity (left side of the figure, with a frequency of 1%). Patients in labor at term with intact membranes have a higher frequency of bacteria in the amniotic cavity even if the membranes are intact (middle part of the figure, with a frequency of 19%). Rupture of the chorioamniotic membranes at term prior to the onset of labor has a higher frequency of bacteria in the amniotic cavity (right side of the figure and table, with a frequency of approximately 34%). This frequency is similar to that observed in patients with preterm PROM.
PROM: premature rupture of the membranes.
Figure 6. Frequency of intraamniotic infection and sterile intraamniotic inflammation in patients with clinical chorioamnionitis at term according to membrane status.
Prevalence of intraamniotic infection and sterile intraamniotic inflammation in patients with clinical chorioamnionitis at term according to the status of the membranes at the time of amniocentesis. Modified from Romero R et al. Clinical chorioamnionitis at term X: microbiology, clinical signs, placental pathology, and neonatal bacteremia—implications for clinical care. J Perinat Med. 2021;49(3):275–98.2
Prolonged labor
Uterine contractions increase the risk of microbial invasion of the amniotic cavity. An experimental study indicates that uterine contractions exert a “suction-like effect” whereby vaginal fluid ascends into the uterine cavity (demonstrated by hysterosalpingoscintigraphy and radio-labeled carbon particles).166 Similar observations have been reported in animals after the placement of fluorescent bacteria in the vagina (unpublished observations). The frequency of microorganisms in the amniotic fluid of patients with spontaneous labor at term with intact membranes is 19%, which is higher than 1% observed in patients at term without labor167 (Figure 5). Seong et al 163 reported that the frequency of microorganisms in amniotic fluid at term increased over time during labor—3.5% during early labor (cervical dilatation <4 cm)—whereas the frequency increased 4-fold (13%) when patients were in active labor (cervical dilatation ≥ 4cm),163 suggesting that the longer the labor and the greater the cervical dilatation, the higher the risk of microorganisms in the amniotic cavity.
Digital examination
Digital examination of the uterine cervix before or during labor has been implicated in the genesis of intraamniotic infection as microorganisms may be carried from the vaginal ecosystem into the lower pole of the chorioamniotic sac. This has led to the view that digital examination should not be performed in patients with preterm prelabor rupture of the membranes (PROM) or even PROM at term because of the adage that “once an examination has been performed, the clock of infection starts to tick”.168 Several studies have addressed the clinical consequences of digital examination of the cervix during labor.169–176 Figure 7 illustrates that the risk of clinical chorioamnionitis at term increases with each additional cervical examination.176 Moreover, the risk of clinical chorioamnionitis is greater when the number of digital examinations increases in the setting of ruptured membranes during the intrapartum period.170, 176
Figure 7. Probability of clinical chorioamnionitis by the number of digital cervical examinations and by the number of hours after rupture of membranes.
Each additional cervical examination confers an incremental risk of clinical chorioamnionitis. A greater length of time from rupture to delivery is associated with an increased risk of clinical chorioamnionitis. Modified from Gomez Slagle HB et al. Incremental risk of clinical chorioamnionitis associated with cervical examination. Am J Obstet Gynecol MFM. 2022;4(1):100524.176
History of clinical chorioamnionitis in a previous pregnancy
A history of clinical chorioamnionitis increases the odds of recurrence in a subsequent pregnancy approximately by 2- to 3-fold compared to patients who did not have this diagnosis in a previous pregnancy.177–179 The frequency of recurrent clinical chorioamnionitis is the highest in patients with clinical chorioamnionitis in the first pregnancy who delivered at 20 to 24 weeks of gestation. The most likely explanation of this is that the rate of intraamniotic infection is extremely high the lower the gestational age at birth.
Microbiology of intraamniotic infection in clinical chorioamnionitis at term
Microorganisms responsible for intraamniotic infection are shown in Table 2. Polymicrobial infections are detected in 70% of patients with intraamniotic infection and clinical chorioamnionitis.1,2 The microorganisms most commonly found in the amniotic fluid are Ureaplasma species (U. parvum or U. urealyticum), Gardnerella vaginalis, Mycoplasma hominis, Streptococcus agalactiae, Staphylococcus aureus, and Bacteroides spp.1,2,20,180–183 Sneathia spp. can be readily identified by the use of molecular microbiologic techniques. Some microorganisms are typical inhabitants of the mouth, eg, Porphyromonas spp., Fusobacterium spp., and Streptococcus oralis.1,2 Fungi (Candida spp.) can cause intraamniotic infection, particularly in patients who become pregnant while using an intrauterine device.184 Viruses such as herpes simplex virus-1 and roseolovirus have been identified in the amniotic fluid, but it is unclear whether they have an etiologic role. To date, Chlamydia spp. has not been detected in the amniotic cavity of patients diagnosed with clinical chorioamnionitis at term; this is consistent with the observation that Chlamydia trachomatis is rarely detected in patients diagnosed with preterm PROM.185
Table 2.
Microorganisms identified in the amniotic fluid of patients with clinical chorioamnionitis at term
Ureaplasma species (Ureaplasma urealyticum, Ureaplasma parvum) |
Gardnerella vaginalis |
Mycoplasma hominis |
Streptococcus agalactiae |
Lactobacillus species |
Bacteroides species |
Fusobacterium species (Fusobacterium nucleatum, Fusobacterium varium) |
Sneathia species |
Streptococcus viridans |
Porphyromonas species |
Veillonella species |
Peptostreptococcus |
Escherichia coli |
Pseudomonas aeruginosa |
Staphylococcus aureus |
Candida species |
Enterococcus faecalis |
Acinetobacter species |
Others: Candida albicans, Staphylococcus epidermidis, Propionibacterium acnes |
Detected with the use of cultivation and molecular microbiologic techniques in the amniotic fluid of patients with clinical chorioamnionitis at term.
Microbial burden has been studied with molecular microbiologic techniques, and the range of microbial load is broad (6–978 genome equivalents per PCR well).2 A consistent finding in our studies is that the greater the microbial burden, the stronger the intensity of the intraamniotic inflammatory response, as reflected by the concentration of IL-6 in amniotic fluid.1,2
Genital mycoplasmas as a cause of intraamniotic infection, maternal infection, and neonatal sepsis
Genital mycoplasmas (U. urealyticum, U. parvum, M. hominis) are the most frequently detected microorganisms in the amniotic cavity in a wide range of obstetrical complications, including clinical chorioamnionitis at term.1, 2, 68, 162, 186–197 These organisms have been implicated in postpartum infections, such as endometritis, and in wound infections in nearly 62% of cases.198 For example, Andrews et al199 reported that patients who had U. urealyticum in the chorioamniotic membranes had a 3-fold increased risk of endometritis. In another study, genital mycoplasmas were detected in 30% of endometrial samples obtained from patients with puerperal endometritis.200 Although some investigators have proposed that genital mycoplasmas are of low virulence and may not be pathogenic, our studies have clearly shown that the presence of genital mycoplasmas in amniotic fluid is associated with intraamniotic, maternal, and fetal inflammatory responses,196,201,202 and, in some cases, that the magnitude of the inflammatory process is more severe than that associated with other bacteria.196 Experimental evidence indicates that the incubation of genital mycoplasmas with chorioamniotic membranes elicits the release of inflammatory mediators, eg, tumor necrosis factor (TNF)-α, into the culture medium.203Importantly, intraamniotic administration of Ureaplasma spp. leads to the onset of labor and fetal infection in nonhuman primates and mice.204–208
The role of genital mycoplasmas in neonatal disease has also been subject to intensive investigation. There is good evidence that genital mycoplasmas can be isolated in cases congenital pneumonia,154, 209–211 neonatal meningitis,212–220 and sepsis.154, 210, 211 Such findings are not unexpected, given that 23% of preterm neonates born between 23 and 33 weeks of gestation have demonstrable bacteremia with genital mycoplasmas at the time of birth when appropriate cultures for these organisms are obtained from umbilical cord blood.221, 222 Therefore, the role of genital mycoplasmas in neonatal infections is well supported despite initial skepticism. Regrettably, these organisms are not routinely cultured, in obstetrics or in neonatal medicine, nor are they effectively treated by standard antimicrobial regimens (ampicillin and gentamicin) in neonatal intensive care units (NICU) or obstetrical units.223–225 Key issues that need to be addressed include the rapid and sensitive diagnosis of infections caused by Ureaplasma spp. in neonates and the modification of the current antimicrobial therapy in the neonatal period. Antibiotics effective against genital mycoplasmas should be considered.
The inflammatory response in the amniotic, maternal, and fetal compartments in clinical chorioamnionitis at term
Microbial or non-microbial stimuli can induce an inflammatory response in 1) the amniotic cavity; 2) the maternal compartment; or 3) the fetal compartment. Typically, in cases of ascending infection from the lower genital tract, the inflammatory process begins in the amniotic cavity and can extend into the fetus and/or the mother. In cases of hematogenous infection, such as maternal listeriosis, the mother is affected first and the fetus and the amniotic compartment secondarily.139, 140, 142–145 The frequency of relative involvement of the amniotic, maternal, and fetal compartments has not been subject to rigorous investigation. Clinical signs of infection are unreliable indicators of involvement; most infections in neonates are subclinical and the same is the case for mothers. The advent of new molecular microbiologic techniques and high-dimensional methods to assess inflammation can be leveraged to ascertain the rate of maternal and fetal involvement.
Inflammation has cellular and soluble components. The typical changes in the soluble mediators are those that occur in the inflammatory cytokine network, which may extend to other inflammatory products such as prostaglandins.226–235 The cellular components involve mainly those of the innate immune system such as neutrophils and macrophages.236
The intraamniotic inflammatory response
Microbial products or alarmins engage the innate immune system to induce an intraamniotic inflammatory response. Neutrophils are the most common subset of leukocytes in the amniotic fluid (Figure 8), followed by monocytes and macrophages. Other immune cells such as T and B lymphocytes, as well as natural killer cells, are less frequent in the amniotic fluid.48, 183 The absolute count of neutrophils, monocytes, and macrophages in amniotic fluid is significantly higher in patients with intraamniotic infection than in patients with sterile intraamniotic inflammation.48, 52 These cells in amniotic fluid can be of maternal, fetal, or mixed origin, as demonstrated by DNA fingerprinting.49, 52 The earlier the gestational age, the more likely they are predominantly of fetal origin (capable of inducing a fever). The closer to term, the more likely that neutrophils, monocytes, and macrophages would be of maternal origin.49, 52 Neutrophils produce TNF-α and macrophage inflammatory protein (MIP)-1β predominantly, whereas monocytes and macrophages express mainly IL-1β.48 These cytokines are implicated both in the process of parturition and in the host response against intraamniotic infection.73, 237–251 The amniotic fluid concentrations of pro-inflammatory cytokines, eg, IL-1β, interferon (IFN)-γ, TNF-α, TNF-β, IL-2, and IL-6, are significantly higher in patients with clinical chorioamnionitis at term than in those with spontaneous labor at term without clinical chorioamnionitis (Figure 9A).251 Among patients with intraamniotic inflammation, the changes in the amniotic fluid cytokine concentrations are similar in magnitude between those with intraamniotic infection and those with sterile intraamniotic inflammation (Figure 9A).251
Figure 8. Amniotic fluid neutrophils and monocytes in patients with clinical chorioamnionitis at term.
Hematoxylin and eosin staining shows the typical morphology of neutrophils (red arrow) and monocytes (green arrow) in the amniotic fluid of patients with clinical chorioamnionitis. Magnification 400X. Scale bars: 50 μm. Modified from Martinez-Varea A. et al. Clinical chorioamnionitis at term VII: the amniotic fluid cellular immune response. J Perinat Med. 2017;45(5):523–38.48
Figure 9. The amniotic, maternal, and fetal inflammatory responses in patients with clinical chorioamnionitis at term.
(A) Amniotic fluid concentrations of inflammatory cytokines (IL-1β, IFN-γ, TNF-α, and TNF-β) are higher in patients with clinical chorioamnionitis at term than in those with spontaneous labor at term. Modified from Romero R et al. Clinical chorioamnionitis at term II: the intra-amniotic inflammatory response. J Perinat Med. 2016;44(1):5–22.251
(B) Maternal plasma concentrations of pyrogenic cytokines (IL-2, IL-6, IL-1β, and IL-17α) are higher in patients with clinical chorioamnionitis at term than in those with spontaneous labor at term. Modified from Romero R et al. Clinical chorioamnionitis at term IV: the maternal plasma cytokine profile. J Perinat Med. 2016;44(1):77–98.252
(C) Umbilical cord plasma concentrations of inflammatory cytokines and chemokines (IL-12p70, IL-6, IL-16, and IL-8) are higher in fetuses with clinical chorioamnionitis at term than in those delivered by women with spontaneous labor at term. Modified from Romero R et al. Clinical chorioamnionitis at term V: umbilical cord plasma cytokine profile in the context of a systemic maternal inflammatory response. J Perinat Med. 2016;44(1):53–76.276
* p < 0.05
IFN: interferon; IL: interleukin; TNF: tumor necrosis factor
Maternal inflammatory response
Microbial products, cytokines, or alarmins can gain access to the maternal compartment and then induce a local inflammatory response, such as deciduitis, followed by a systemic response that can be detected by changes in the concentrations of inflammatory mediators in maternal blood or by clinical signs of inflammation. Indeed, most of the signs of clinical chorioamnionitis (except fetal tachycardia) are thought to reflect a maternal inflammatory response.92 Maternal plasma concentrations of pyrogenic cytokines such as IL-2, IL-6, IL-1β, and IL-17A are higher in patients with clinical chorioamnionitis at term than in those with spontaneous labor at term without clinical chorioamnionitis (Figure 9B).252 In addition, patients with clinical chorioamnionitis at term without intraamniotic infection have a greater maternal plasma concentration of pyrogenic cytokines than those with spontaneous labor at term without this clinical condition (Figure 9B).252 However, maternal plasma concentrations of cytokines are not significantly different among the 3 major subgroups of patients with clinical chorioamnionitis at term (Figure 9B).252 These findings indicate that the absolute concentration of cytokines in the maternal plasma cannot be used to identify patients who have intraamniotic infection.252
The fetal inflammatory response
When microbial and non-microbial stimuli reach the human fetus, a local and then a systemic inflammatory response are elicited. The pattern of engagement of the fetal immune system depends upon the pathway of infection. For example, in cases of ascending infection, microorganisms can reach the fetal skin and tympanic membranes or can be aspirated into the respiratory tract (explaining the occurrence of fetal dermatitis, otitis media, pneumonia, etc., in cases of intraamniotic infection). If the infection is hematogenous, microorganisms can elicit a fetal systemic inflammatory response by engaging the fetal endothelium during the course of bacteremia. This systemic inflammatory response is a precursor of fetal and neonatal sepsis. Indeed, there is a dose–response relationship between the concentrations of cytokines, such as TNF-α, and early neonatal sepsis.253
Neonates born to mothers diagnosed with clinical chorioamnionitis are at risk for sepsis,4, 23–26, 254–259 meconium aspiration syndrome,254, 260–264 neonatal encephalopathy,265–268 and long-term neurodevelopmental disabilities, including cognitive impairment,269–273 cerebral palsy,272 and neonatal death.6, 10, 112, 259, 274, 275 The concentration of inflammatory cytokines and chemokines (IL-12p70, IL-6, IL-16, and IL-8) in the umbilical cord is higher in fetuses born to mothers diagnosed with clinical chorioamnionitis at term than in those whose mothers underwent spontaneous labor at term without this clinical condition (Figure 9C).276 The fetal inflammatory response syndrome (FIRS; defined as an elevated fetal plasma IL-6 concentration ≥11pg/ml) is present in fetuses with clinical chorioamnionitis (Figure 10).277 Of note, the fetuses affected with FIRS generally are born to mothers with proven intraamniotic infection,276 a finding reflecting the severity of the fetal inflammatory response tends to be greater in such cases.276 Interestingly, the fetuses of mothers with clinical chorioamnionitis at term without intraamniotic infection or sterile intraamniotic inflammation also have a higher concentration of inflammatory cytokines in umbilical cord blood than those without clinical chorioamnionitis, suggesting that maternal fever in the absence of intraamniotic infection leads to a change in the fetal cytokine network (Figure 9C).276 Furthermore, there are significant positive correlations between maternal and umbilical cord plasma IL-6 and IL-8 concentrations; these observations are consistent with the possibility of placental transfer of cytokines.276, 278
Figure 10. Fetal inflammatory response syndrome in clinical chorioamnionitis.
A. Fetal inflammatory response syndrome can be diagnosed by an increased concentration of umbilical cord plasma IL-6 ( ≥ 11pg/mL).
B. The median concentration of umbilical cord plasma IL-6 is higher in fetuses with clinical chorioamnionitis than in those delivered by patients with near-term labor without clinical chorioamnionitis [27.46 pg/mL vs. 2.13 pg/mL; p<0.001]. Sixty-two percent of fetuses (16/26) with clinical chorioamnionitis have a fetal plasma concentration of IL-6 >11 pg/mL. Modified from Chaiworapongsa T et al. Evidence for fetal involvement in the pathologic process of clinical chorioamnionitis. Am J Obstet Gynecol. 2002;186(6):1178–82.277
IL: interleukin.
Criteria for the diagnosis of clinical chorioamnionitis
The current criteria for the diagnosis of clinical chorioamnionitis20, 89, 279 are based on clinical signs, including fever (±37.8°C or ±100.4°F) and two or more of the following: A) maternal tachycardia (>100 beats per minute); B) maternal leukocytosis (white blood cell count >15,000 cells/mm3; C) uterine tenderness; D) fetal tachycardia (>160 beats per minute); or E) purulent or malodorous cervical discharge.1, 19, 20, 89–92, 94, 279–284 The rationale for the precise cut-off values to define fever, maternal tachycardia, and fetal tachycardia was reviewed in detail by Newton,92 who proposed the 90th percentile threshold for both maternal tachycardia and fetal tachycardia and the 80th percentile for a white blood cell count (Table 3).
Table 3.
The origin of the criteria of clinical chorioamnionitis
Parameter | Mean ± standard deviation | 95th percentile | Diagnosis of intraamniotic infection |
---|---|---|---|
Temperature, °C | 36.9 ± 0.5 | 37.85 | 37.8 |
Maternal heart rate, beats/min | 85 ± 13 | 110 | 100 |
Fetal heart rate, beats/min | 139 ± 10 | 159 | 160 |
White blood cell count, cells/mm3 | 12,500 ± 3.9 | 20,100 | 15,000 |
Adapted from Newton ER. Chorioamnionitis and intraamniotic infection. Clin Obstet Gynecol. 1993;36(4):795–808.92
The origin and evolution of the criteria for the current definition of clinical chorioamnionitis is interesting and has been influenced by the work of Gibbs et al.20 They reported that patients who met the clinical criteria, proposed by their group, were more likely to have a higher concentration of microorganisms in amniotic fluid, using quantitative microbiology, and had isolates with higher virulence. This observation was based on a study of 52 patients diagnosed with clinical chorioamnionitis at term and matched with 52 controls in whom amniotic fluid was retrieved by a transcervical catheter.20 The key observation was that 81% (42/52) of patients with clinical chorioamnionitis had >102 colony-forming units (CFU)/mL compared to 31% (16/52) in the control group (p<0.001). Organisms considered to be of high virulence were present in 69% (36/52) of patients with clinical chorioamnionitis and in 8% (4/52) of controls (p<0.01). Moreover, maternal bacteremia was detected in 10% (4/39) of patients diagnosed with clinical chorioamnionitis who had a blood culture.20 Of note, a limitation recognized by the authors at the time of publication was that amniotic fluid for microbiologic studies had been obtained through a transcervical catheter and that despite discarding the first 7 mL of each sample, 31% of patients in the control group presented more than 102 CFU/mL, suggesting contamination by microorganisms in the lower genital tract.20 Indeed, in studies of patients in term labor when amniotic fluid collection was performed by transabdominal amniocentesis at the time of cesarean delivery, the frequency of positive amniotic fluid cultures ranged between 13% and 19%.163,167 Therefore, an accurate assessment of microbial status requires that the amniotic fluid sample be obtained transabdominally. The clinical criteria developed by Gibbs et al.20 and Newton92 have continued to be applied because they have been perceived as useful in the differential diagnosis of intrapartum fever.
How well do the criteria for clinical chorioamnionitis perform in the identification of intraamniotic infection?
Studies of patients with the diagnosis of clinical chorioamnionitis at term in which cultivation and molecular microbiologic methods were used to detect intraamniotic infection show that the traditional criteria do not accurately distinguish between patients with and without intraamniotic infection.2, 95 The sensitivity of maternal tachycardia, fetal tachycardia, and maternal leukocytosis ranged from 75% to 90%; however, the specificity was poor for these criteria, ranging from 0% to 30%. By contrast, malodorous amniotic fluid and uterine tenderness had high specificity (95% and 95%, respectively) but a low sensitivity (8% and 12%, respectively) for the identification of intraamniotic infection.2, 95 Collectively, the diagnostic accuracy for each clinical sign ranges between 40% and 58% for the diagnosis of intraamniotic infection (Table 4).95 The disappointing performance of clinical criteria has substantial implications: a large number of mothers and neonates are treated with antibiotics and undergo evaluation for suspected sepsis, given the fear that infection will be missed. Further studies are required to identify biomarkers that reliably and rapidly differentiate the patients with a fever due to intraamniotic infection or sterile intraamniotic inflammation, or those without intraamniotic infection or intraamniotic inflammation.
Table 4.
The diagnostic accuracy of clinical criteria for the identification of intraamniotic infection in patients with clinical chorioamnionitis at term
Clinical signs | Sensitivity | Specificity | Positive likelihood ratio(95% CI) | Negative likelihood ratio(95% CI) | Diagnostic accuracy% (n/N) | ||
---|---|---|---|---|---|---|---|
% (n/N) | (95% CI) | % (n/N) | (95% CI) | ||||
Maternal tachycardia | 88.0 (22/25) | (68.75–97.31) | 5.0 (1/20) | (0.83–24.95) | 0.93 (0.78–1.10) |
2.40 (0.27–21.35) |
51.1 (23/45) |
Fetal tachycardia | 80.0 (20/25) | (59.29–93.09) | 30 (6/20) | (11.97–54.27) | 1.14 (0.81–1.62) |
0.67 (0.24–1.87) |
57.8 (26/45) |
Maternal leukocytosis | 76 (19/25) | (54.87–90.58) | 30 (6/20) | (11.97–54.27) | 1.09 (0.76–1.56) |
0.80 (0.30–2.10) |
55.6 (25/45) |
Malodorous vaginal discharge | 8.00 (2/25) | (1.22–26.07) | 95 (19/20) | (75.05–99.17) | 1.60 (0.16–16.40) |
0.97 (0.83–1.13) |
46.7 (21/45) |
Uterine tenderness | 12.0 (3/25) | (2.69–31.25) | 95 (19/20) | (75.05–99.17) | 2.40 (0.27–21.35) |
0.93 (0.78–1.10) |
48.9 (22/45) |
≥ 3 criteria | 56.00 (14/25) | (34.94–75.57) | 55 (11/20) | (31.55–76.90) | 1.24 (0.69–2.26) |
0.80 (0.44–1.45) |
55.6 (25/45) |
> 4 criteria | 8.00 (2/25) | (1.22–26.07) | 100 (20/20) | (83.01–100) | 0.92 (0.82–1.03) |
– | 48.9 (22/45) |
Maternal tachycardia (> 100 beats/min); fetal tachycardia (> 160 beats/min); and maternal leukocytosis ( > 15,000 cells/mm3).
CI: confidence interval.
Adapted from Romero R, Chaemsaithong P, Korzeniewski SJ, Kusanovic JP, Docheva N, Martinez-Varea A, et al. Clinical chorioamnionitis at term III: how well do clinical criteria perform in the identification of proven intra-amniotic infection? J Perinat Med. 2016;44(1):23–32.95
Laboratory diagnosis of intraamniotic inflammation and intraamniotic infection
The diagnosis of clinical chorioamnionitis is based on the presence of maternal fever and two signs of maternal or fetal inflammation. Table 4 shows the sensitivity and specificity of clinical criteria to detect the patient with intraamniotic infection.
Amniotic fluid analysis o is indicated in cases of diagnostic certainty. Indeed, analysis of amniotic fluid is the gold standard for the diagnosis of intraamniotic inflammation and intraamniotic infection (Figure 11). Clinical criteria only allow a presumptive diagnosis. A conclusive, or definitive, diagnosis of infection requires identification of microorganisms and inflammation. Fluid can be retrieved at the time of cesarean delivery or by transabdominal amniocentesis. Tests could include the Gram stain, white blood cell count, and glucose concentration. These tests are employed to analyze cerebrospinal fluid in cases of suspected meningitis and are widely available in clinical laboratories all over the world.
Figure 11. Laboratory diagnosis of intraamniotic inflammation and intraamniotic infection.
Definitive diagnoses of intraamniotic inflammation and intraamniotic infection require a transabdominal amniocentesis to collect amniotic fluid. Amniotic fluid should be assessed by (1) a WBC count and differential; (2) a glucose concentration; (3) a Gram stain and a bacterial culture that include aerobic/anaerobic bacteria and genital mycoplasmas; (4) MMP-8; (5) cytokines, eg, IL-6; and (6) other tests (eg, rapid tests, PCR). Intraamniotic inflammation is the presence of an inflammatory response in the amniotic cavity, which can be diagnosed by a WBC count ≥ 50 cells/mm3, an amniotic fluid glucose concentration <14 mg/dL, an IL-6 concentration ≥ 2.6 ng/mL, or an MMP-8 concentration >23 ng/mL. Rapid tests for amniotic fluid MMP-8 (Yoon’s MMP-8 Check®; OBMed Co., Ltd., Seoul, Republic of Korea) and IL-6 (Milenia QuickLine®; Milenia Biotec, Bad Nauheim, Germany) are available to be performed at the bedside. Microorganisms in amniotic fluid can be identified by Gram stain, cultivation methods, or molecular microbiologic techniques.
IL: interleukin; MMP: matrix metalloproteinase; PCR: polymerase chain reaction; rRNA: ribosomal RNA; WBC: white blood cell.
Under normal circumstances, bacteria are not visible in amniotic fluid with a Gram stain, and a white blood cell count should be <50 cells/mm3 (criteria listed in Figure 11).57–60 The amniotic fluid glucose concentration is used to assess the likelihood of intraamniotic inflammation in preterm gestation; however, the concentration of glucose decreases with gestational age, and a clear threshold has not been established at term, although amniotic fluid glucose is extremely low [median (interquartile range), 1 (1–4) mg/dL] in patients with intraamniotic infection at term.1, 2 Amniotic fluid concentrations of MMP-8 or IL-6 are excellent tests for the detection of inflammation. A rapid assay for MMP-8 is available, which can be performed at the bedside (Yoon’s MMP-8 Check®; OBMed Co., Ltd. Seoul, Republic of Korea); its diagnostic accuracy in identifying intraamniotic inflammation in patients with clinical chorioamnionitis at term is 84% (sensitivity, 82%; specificity, 90%; positive likelihood ratio, 8.2; negative likelihood ratio, 0.2).285 A rapid test to determine amniotic fluid IL-6 is commercially available in Europe (Milenia QuickLine®; Milenia Biotec, Bad Nauheim, Germany) (Figure 11).
A transcervical amniotic fluid collector is a non-invasive method that assesses the inflammatory status of the amniotic cavity in patients with rupture of the membranes (Figure 12A).129, 130 Amniotic fluid analysis can be performed by utilizing point-of-care assays such as an IL-8 test (Figure 12B).130 Future studies to determine whether a transcervical amniotic fluid collector can be useful in the differential diagnosis of patients with clinical chorioamnionitis with intraamniotic inflammation vs neural inflammation related to an epidural are needed.
Figure 12. Amniotic fluid collector and rapid test to assess the presence of intraamniotic inflammation by determining interleukin-8 at the bedside.
A. A transcervical collector allows sampling of amniotic fluid after rupture of the membranes.
B. Rapid analysis of an interleukin-8 concentration at the bedside can be used to diagnose intraamniotic inflammation.
Cultures for aerobic and anaerobic bacteria as well as for genital mycoplasmas are performed to diagnose intraamniotic infection; however, results for these tests can take days to become available. Molecular microbiologic techniques allow the detection of the 16S ribosomal RNA gene to identify bacteria within a few hours.286 Indeed, successful use of nanopore sequencing, a long-read, real-time DNA sequencing technique, to diagnose intraamniotic infection within 5 to 9 hours has been recently reported.287 We envision that this technology will replace culture, allow the identification of intraamniotic infection, and guide the selection of appropriate antimicrobial agents for mothers and neonates in the future. It is possible to identify the microorganisms and to determine whether they have antimicrobial resistant genes in less than 24 hours.
Effect of clinical chorioamnionitis on uterine contractility
Patients with clinical chorioamnionitis are more likely to experience decreased or ineffective uterine contractility,3, 9, 288–290 prolonged labor,3, 9, 288–290 and cesarean delivery for failure to progress or for non-reassuring fetal heart rate tracing,3, 4, 8, 9, 280, 288, 290, 291 and to receive oxytocin for augmentation of labor3, 280, 288, 290 than those without clinical chorioamnionitis. Duff et al288 reported that 75% of patients with clinical chorioamnionitis have decreased uterine contractility and that, despite oxytocin augmentation, 34% require cesarean delivery because of the diagnosis of failure to progress in labor. A careful study by the group of Goetzl et al reported a significant decrease in uterine activity two hours after the onset of a fever (Figure 13). This occurred in nulliparous and multiparous patients.292
Figure 13. Relationship between maternal fever and uterine contractility.
Uterine contractility significantly and steadily declined 2 hours after the onset of maternal fever. Modified from Zackler A et al. Suspected Chorioamnionitis and Myometrial Contractility: Mechanisms for Increased Risk of Cesarean Delivery and Postpartum Hemorrhage. Reprod Sci. 2019;26(2):178–83.292
The mechanisms whereby maternal inflammation predisposes to myometrial dysfunction have not been determined. Proinflammatory cytokines, eg, IL-1β or TNF-α, can stimulate uterine contractility and have been implicated in the mechanisms responsible for the onset of preterm parturition in the context of intraamniotic infection293, 294 as well as spontaneous labor at term.295, 296 This concept would appear to conflict with the clinical findings that maternal fever and clinical chorioamnionitis are associated with impaired uterine contractility at term296, 297 and uterine atony in the postpartum period.280, 292, 298 How can these findings be reconciled? One possibility is that labor is stimulated during the early stages of intraamniotic infection. However, when the infection and the inflammatory process extend to the myometrium, pathologic inflammation impairs uterine contractility. Cierny et al 296 reported that the relationship between the concentration of maternal plasma cytokines and the progression of term labor is complex and likely to be bimodal. A low maternal plasma concentration of IL-6 has been associated with a prolonged latent phase of labor, suggesting that an inadequate inflammatory process slows labor (normal labor is considered a sterile inflammatory process), as demonstrated by transcriptomic analysis of the myometrium,299, 300 cervix,301 and chorioamniotic membranes.302, 303 However, once active labor is established, patients with elevated maternal plasma cytokines (IL-1β and TNF-α) may experience prolonged labor.296 This suggests that pathologic inflammation can lead to an impairment of myometrial contractility. Indeed, an arrest of dilatation304 and an arrest of descent305 have features of exaggerated inflammation compared to normal labor.
Fetal heart rate in clinical chorioamnionitis
Abnormal fetal heart rate patterns observed in patients with clinical chorioamnionitis include tachycardia, absence of accelerations, variable and late decelerations, a sinusoidal pattern, persistently reduced variability, and absence of cycling.288, 306–309 Of these abnormalities, fetal tachycardia is the most common finding (~81%),1, 2, 95 and it is thought to reflect an increase in maternal temperature310, 311 and/or in fetal systemic inflammation (Figure 14).277, 312–315
Figure 14. Fetal tachycardia in a case of clinical chorioamnionitis at 40 weeks of gestation.
A. Fetal heart rate of 190 beats per minute (bpm). The associated loss of variability is also noteworthy.
B. Tachycardia is associated with decelerations. The amniotic fluid culture was positive for Ureaplasma urealyticum and Staphylococcus aureus. The concentration of interleukin-6 in amniotic fluid was 37.49 ng/mL (cut-off 2.6 ng/mL), which is consistent with intraamniotic inflammation. The fetus presented fetal systemic inflammation, as demonstrated by the presence of funisitis. The newborn was septic, and the neonatal blood culture was positive for Staphylococcus aureus, the same bacterium identified in the amniotic fluid.
The frequency of fetal acidemia assessed by umbilical cord pH and blood gases is similar in patients with and without clinical chorioamnionitis.306, 316, 317 A study of 197 patients diagnosed with clinical chorioamnionitis showed no significant association between fetal acidemia (umbilical artery pH <7.20) and abnormal fetal heart rate patterns, including loss of variability, absence of accelerations, and tachycardia.306 Another study of 139 patients with intrauterine bacterial infection (defined as clinical chorioamnionitis plus a positive bacterial amniotic fluid culture or a neonatal infection) reported that fetal heart rate deceleration patterns (eg, decreased variability and absence of accelerations) were not significantly associated with the risk of cerebral palsy at two years of age.307
Placental pathology: acute histologic chorioamnionitis and funisitis
Histologic examination of the placenta is more helpful in excluding intraamniotic infection/inflammation than in making a positive diagnosis. In the absence of funisitis and chorionic vasculitis, fetal inflammation is unlikely to be an etiologic factor in neonatal complications and long-term handicap. Acute placental inflammatory lesions are observed in 79% of the placentas of patients with clinical chorioamnionitis at term (acute histologic chorioamnionitis: 79%; funisitis: 67%).2 Most patients with proven intraamniotic infection have acute chorioamnionitis (93%) or funisitis (67%), and 52% of such cases have severe lesions (defined as stage 3 and grade 2 acute histologic chorioamnionitis and/or funisitis).2 Patients with clinical chorioamnionitis but without intraamniotic infection and intraamniotic inflammation generally do not show evidence of severe acute histologic chorioamnionitis.2
Differential diagnosis of intraamniotic infection
Sterile intraamniotic inflammation: a new entity
A subset of patients with clinical chorioamnionitis in term and preterm gestation shows evidence of intraamniotic inflammation without the presence of bacteria in amniotic fluid, ie, sterile intraamniotic inflammation.1, 2 The presence of microorganisms has been excluded by a combination of methods (culture, presence of nucleic acids, and metagenomics).318 The magnitude of inflammatory response in the mother, fetus, and amniotic fluid in patients with sterile intraamniotic inflammation is similar to or of a lesser magnitude than that found in patients with intraamniotic infection (Figure 9).251
Sterile intraamniotic inflammation is thought to result from danger signals, or alarmins,283, 319–322 or from infections that escaped detection with current cultivation and molecular microbiologic techniques. Sterile inflammatory processes are well-recognized in other fields of medicine and occur when the injurious agent is of non-microbial origin. For example, a burn induces a sterile inflammatory process,323 urate crystals in the case of gout,324, 325 and calcium pyrophosphate dihydrate crystals in pseudogout.326 Urate crystals or calcium pyrophosphate engage the innate immune system by activating inflammasomes.
Damage-associated molecular patterns (DAMPs, also referred to as danger signals or alarmins) are molecules released by cells undergoing stress, necrosis, or senescence.327–330 Cell death of the chorioamniotic membranes (ie, pyroptosis) or in fetal tissues could lead to the accumulation of alarmins and generate sterile intraamniotic inflammation in a wide range of obstetrical complications.322 For example, in gastroschisis, exposure of the fetal bowel to amniotic fluid results in a sterile intraamniotic inflammatory process characterized by an elevated MMP-8 concentration in amniotic fluid.331 Fetal surgery can also result in a transient sterile intraamniotic inflammatory process as surgical trauma elicits an inflammatory response. Alarmins are detectable in the amniotic fluid of patients with an episode of preterm labor with intraamniotic inflammation but without detectable bacteria,294,332 and the intraamniotic administration of alarmins can induce preterm labor in vivo.321 Sterile intraamniotic inflammation generates a chemotactic gradient, thus acute histologic chorioamnionitis similar to that generated by intraamniotic infection. The same can be said about funisitis and chorionic vasculitis.
Several interventions, including the blockade of the NLRP3 inflammasome,333 macrolides,334 and betamethasone,335 can be used to treat sterile intraamniotic inflammation. In animal models, these interventions reduced spontaneous preterm delivery and neonatal morbidity. Blockade of the NLRP3 inflammasome can be accomplished with a specific inhibitor which is currently being tested in non-pregnant subjects. N-acetylcysteine (NAC), a small molecule used to treat acetaminophen intoxication during pregnancy, has anti-inflammatory effects that have been attributed to attenuation of oxidative stress (ie, as a scavenger of reactive oxygen species). NAC also inhibits the NLRP3 inflammasome.368
Clinical chorioamnionitis without intraamniotic inflammation: a clinical enigma
Twenty percent of patients with the diagnosis of clinical chorioamnionitis at term do not show evidence of intraamniotic infection or intraamniotic inflammation after thorough examination of amniotic fluid samples obtained by transabdominal amniocentesis.1,2 Interestingly, these patients have maternal plasma concentrations of cytokines similar to those with proven intraamniotic infection or sterile intraamniotic inflammation,251 and this raises the question of the source of the systemic inflammatory response. One explanation for the fever is neuroinflammation caused by epidural analgesia. This condition is important, given that 73% of patients in the United States receive epidural analgesia during labor336–338 and that 14% to 33% of patients who receive this anesthetic develop a fever.339–345
Epidural analgesia and maternal fever
The association between epidural analgesia and maternal fever is supported by the following observations: 1) maternal body temperature increases within 4 to 6 hours after an epidural placement (Figure 15A),346–350 and 2) an epidural increases the odds ratio (OR) of fever (≥37.5°C or ≥99.5°F) substantially [OR, 5.26; 95% confidence interval (CI), 4.98–5.56].351 Not widely known is that an epidural-related fever is a particular phenomenon observed in pregnant women in labor. Indeed, an epidural administered to pregnant women not in labor is rarely associated with a fever (eg, an epidural for cesarean delivery).352–354
Figure 15. Epidural analgesia, maternal temperature, and serum interleukin-6 concentrations.
A. Mean vaginal temperature (°C) of patients during labor according to intrapartum pain control. Pregnant women who received epidural analgesia (red) had an increased mean temperature after the administration of anesthetic agents. By contrast, the temperature of women receiving pethidine (non-epidural, blue) remained constant. Modified from Fusi L et al. Maternal pyrexia associated with the use of epidural analgesia in labour. Lancet. 1989 Jun 3;1(8649):1250–2.346
B. Maternal serum interleukin-6 (IL-6) concentrations significantly increase with the duration of epidural analgesia. Modified Goetzl et al. Elevated maternal and fetal serum interleukin-6 levels are associated with epidural fever. Am J Obstet Gynecol. 2002 Oct;187(4):834–8.358
In the case of epidural-induced maternal fever, the process is thought to represent a case of sterile systemic inflammation.355, 356 Riley et al 355 reported that 95% of epidural-related fever cases had a negative placental culture, and a systematic review and meta-analysis showed no association between epidural analgesia and neonatal sepsis (risk ratio, 0.56; 95% CI, 0.08–3.9),356 strengthening the case for a non-infection-related etiology. Systematic studies have shown that women who receive epidural analgesia have a higher mean maternal serum IL-6 concentration than those without epidural analgesia357 and that the longer the duration of epidural analgesia, the higher the maternal serum IL-6 concentration (Figure 15B).358
The precise etiology of the inflammatory process may be related to local anesthetics (eg, ropivacaine or bupivacaine) that can induce a maternal systemic inflammation after absorption from the epidural space into the vasculature.354, 359 During pregnancy, the density of the vascular network of epidural veins, ie, Batson’s plexus, is higher than that of non-pregnant subjects as shown by endoscopic studies of the epidural space, Figure 16A).360 Compression of the inferior vena cava by the pregnant uterus is thought to cause congestion of the epidural veins and a decrease in the volume of the epidural space.361 Thus, a similar volume of local anesthetic agents would spread more extensively in pregnant women than in non-pregnant patients. Recently, Wohlrab et al 362 reported that in vitro ropivacaine causes endothelial cell apoptosis and the release of alarmins, or danger signals, that, in turn, could induce the production of pyrogenic cytokines, eg, IL-1β and IL-6 (Figure 16B).345, 354, 362–364 For further details about epidural analgesia and maternal fever, the reader is referred to the review articles by Goetzl365 and Patel et al 366 in this Supplement.
Figure 16. A needle in the epidural space, optical imaging, and the mechanisms of fever.
A. Fiberscope of the epidural space. During pregnancy, the density of the epidural vessels is higher than that of non-pregnant subjects. It has been proposed that during pregnancy the inferior vena cava becomes compressed by the pregnant uterus and that this causes the epidural veins to engorge and the volume of the epidural space (*, blue) to decrease. Thus, a similar volume of local anesthetic agents can spread more extensively in the pregnant state than in the non-pregnant state. Modified from Eltzschig HK et al. Regional anesthesia and analgesia for labor and delivery. N Engl J Med. 2003 Jan 23;348(4):319–32359 and from Igarashi T et al. The fiberscopic findings of the epidural space in pregnant women. Anesthesiology. 2000 Jun;92(6):1631–6.360
B. Local anesthetics such as ropivacaine used during epidural analgesia can induce maternal systemic inflammation. Ropivacaine can directly cause endothelial cell apoptosis, and this induces the release of damage-associated molecular patterns (DAMP), also known as alarmins, that trigger the production of pyrogenic cytokines (eg, interleukin (IL)-1β, IL-6, IL-8), which may be responsible for fever in pregnant women following epidural analgesia.
Management
Antibiotic administration
The standard treatment of clinical chorioamnionitis at term is the administration of antibiotics and augmentation of labor.91, 279, 282, 367, 368 There is consensus that patients with clinical chorioamnionitis should receive intrapartum antibiotic therapy to prevent adverse perinatal outcomes.124, 369–372 This recommendation is based on the findings of a randomized clinical trial that compared intrapartum antibiotic treatment to immediate postpartum treatment.281 This influential trial changed clinical practice. Therefore, the context of the study, design, conduct, and outcome are worth reviewing.
This study was conceived at a time when intrapartum antibiotic administration for clinical chorioamnionitis was not the standard of practice. At that time, neonatologists often requested that obstetricians delay antibiotic treatment until the postpartum period so that a blood culture of the neonate could be obtained to assess the likelihood of sepsis and to identify the organisms.
The trial included patients with clinical chorioamnionitis after 34 weeks of gestation281 who were randomized to receive the combination of ampicillin (2 grams IV every 6 hours) and gentamicin (1.5 mg/kg IV every 8 hours) intrapartum versus postpartum (the postpartum treatment began after clamping of the umbilical cord). Patients who underwent cesarean delivery also received clindamycin (900 mg intravenously every 8 hours) after cord clamping. Intravenous antibiotics were continued until patients were afebrile for more than 48 hours. Timing and route of delivery were based on obstetrical criteria.
The primary endpoint of the trial was neonatal sepsis, defined as either bacteremia or death with the clinical diagnosis of sepsis and a positive culture. Evaluation of neonates after birth was based on one set of blood cultures and a chest x-ray, read by a radiologist unaware of the clinical course. Neonates received IV antibiotic treatment with ampicillin (75 mg/kg) and gentamicin (2.5 mg/kg) every 12 hours. If the neonatal blood culture and chest x-rays were negative, and the clinical course uncomplicated, antibiotics were discontinued after 72 hours. If sepsis or pneumonia was diagnosed, treatment was continued for a minimum of 10 days.
The trial was stopped by the Data and Safety Monitoring Committee after the enrollment of 48 patients (26 in the intrapartum treatment and 22 in the postpartum treatment), given that antibiotic administration was associated with a significantly lower rate of neonatal sepsis (21% in the postpartum treatment group and 0% in the intrapartum group).281 In addition, intrapartum treatment was associated with a significant decrease in the risk of neonatal pneumonia or sepsis (32% vs 0.0%; p = 0.03), length of neonatal stay (mean: 5.7 days vs 3.8 days; p = 0.02), and maternal postpartum hospital stay (mean: 5.0 vs 4.0 days; p = 0.05)281 compared to immediate postpartum treatment.
Some aspects of the trial deserve commentary. First, the rate of neonatal sepsis in the control group was 21% (4/19),281 which is high compared to the rate of neonatal sepsis in clinical chorioamnionitis at the time, or now.373, 374 Second, the observed beneficial effects of antibiotics, namely a decrease in the rate of neonatal sepsis, could be due to suppression of bacterial growth in culture—rather than a true decrease in the rate of neonatal sepsis. This possibility must be considered in light of the evidence that antimicrobial administration can reduce the rate of positive blood cultures.375
This trial was not replicated after publication. Subsequently, other studies examined the effects of different antimicrobial agents on postpartum febrile morbidity and endometritis.376–379 However, neonatal endpoints have not been revisited. In essence, the overall evidence for efficacy of antibiotics in medicine is so persuasive and the risks of neonatal sepsis so high that withholding antibiotics is difficult to justify.373Three-and-a-half decades after this important trial, a number of questions has emerged about the optimal diagnosis and treatment of clinical chorioamnionitis and suspected neonatal sepsis. Observational studies have shown that microorganisms isolated from infection sites such as wound infections or pelvic abscesses are the same as those found in amniotic fluid at the time of cesarean delivery.165 These observations suggest that antibiotic choice needs to be modified to consider the microbiology of intraamniotic infection in clinical chorioamnionitis at term, which is now well-known, using state-of-the-art culture techniques and molecular microbiologic methods.1,2 Several antibiotic regimens have been proposed for the treatment of clinical chorioamnionitis, and they include different generations of cephalosporins and other antimicrobials listed in Table 5.
Table 5.
Empiric antibiotic regimens proposed for the treatment of clinical chorioamnionitis
Antibiotic regimen proven to eradicate intraamniotic infection 119–122 |
Ceftriaxone 1 g IV every 24 hours plus clarithromycin 500 mg IV every 12 hours plus metronidazole 500 mg IV every 8 hours (clarithromycin is sometimes not available for intravenous administration but rather for oral administration) |
Antibiotic regimen by recommended by ACOG (alternatives for patients allergic to penicillin are described in the text) 371 |
Ampicillin 2 g IV every 6 hours plus gentamicin 2 mg/kg load followed by 1.5 mg/kg every 8 hours (or gentamicin 5 mg/kg IV every 24 hours)a,b (clindamycin is added if patients have a cesarean delivery after clamping of the umbilical cord) |
Alternative regimens used for the treatment of clinical chorioamnionitis 368 |
Cefotetan 2 g IV every 12 hours |
Cefoxitin 2 g IV every 6 to 8 hours |
Ceftizoxime 2 g IV every 12 hours |
Cefotaxime 2 g IV every 8 to 12 hours |
Cefuroxime 1.5 g IV every 8 hours |
Cefazolin 1 g IV every 8 hours plus gentamicin 5 mg/kg IV every 24 hours or 1.5 mg/kg IV every 8 hours |
Cefuroxime 750 mg IV every 8 hours plus metronidazole 500 mg IV every 8 hours |
Mezlocillin 3–4 g IV every 6 hours |
Piperacillin-Tazobactam 3.375 g IV every 6 hours |
Piperacillin-Tazobactam 4 g IV every 6 hours plus clarithromycin 500 mg orally every 12 hours |
Ticarcillin-clavulanic acid 3.1 g IV every 6 hours |
Ertapenem 1 g IV every 24 hours |
Meropenem 1 g IV every 12 hours |
Imipenem-cilastatin 500 mg IV every 6 hours |
Modified from Conde-Agudelo et al.368 (specific citations for each combination are in the original Table 2 in the article)
ACOG, American College of Obstetricians and Gynecologists; IV, intravenous.
For mild penicillin allergy: cefazolin (2 g IV every 8 hours) as an alternative to ampicillin; For severe penicillin allergy: clindamycin (900mg IV every 8 hours) or vancomycin (1g IV every 12 hours as an alternative to ampicillin. Vancomycin should be used if the woman is colonized with group B streptococci resistant to either clindamycin or erythromycin (unless clindamycin-inducible resistance testing is available and is negative) or if the woman is colonized with group B streptococci and antibiotic sensitivities are not available.
Post-cesarean delivery antibiotic regimen: One additional dose of the chosen regimen is indicated. Add clindamycin 900 mg IV or metronidazole 500 mg IV for at least one additional dose. Post-vaginal delivery: No additional doses required; but if given, clindamycin is not indicated.
Vancomycin should be used if the woman is colonized with Group B Streptococci resistant to either clindamycin or erythromycin (unless clindamycin-inducible resistance testing is available and is negative) or if the woman is colonized with Group B Streptococci and antibiotic sensitivities are not available.
Empiric antibiotic treatment of clinical chorioamnionitis
Antimicrobial treatment is initiated as soon as the diagnosis is made. Patients can be treated with ampicillin 2 g IV every 6 hours combined with gentamicin 5 mg/kg every 24 hours or 1.5 mg/kg every 8 hours or ampicillin plus sulbactam 3 g IV every 6 hours. In case of a cesarean delivery, patients receive clindamycin 900 mg IV at the time of umbilical cord clamping. Based on expert opinion, metronidazole 500 mg IV has been proposed as an alternative to clindamycin in the event of cesarean delivery. In penicillin-allergic patients, clindamycin 900 mg IV every 8 hours, or vancomycin 1 g IV every 12 hours, or erythromycin 500 mg to 1 g IV every 6 hours can be used instead of ampicillin.
Other alternatives to ampicillin plus gentamicin have emerged. A major advance in antimicrobial treatment has been the development of newer beta-lactam antibiotics, a term describing agents containing a beta-lactam ring that binds with enzymes in the cell wall of the bacteria, such as penicillin-binding proteins, thereby disrupting the crosslinking of peptidoglycan, a key component of the bacterial cell wall. A weak bacterial cell wall causes osmotic stress; therefore, these types of antibiotics are bactericidal instead of bacteriostatic.
Since bacteria can produce beta-lactamases, which inhibit the beta-lactam ring and lead to antibiotic resistance, beta-lactamase inhibitors have been developed for use in combination with beta-lactam antimicrobial agents. Examples include clavulanic acid, sulbactam, tazobactam, and cilastatin. Popular combinations are ampicillin plus sulbactam, which appears to be equivalent to the combination of ampicillin and gentamicin (for details, see reviews of antimicrobial agents in 368 and 407), piperacillin and tazobactam with and without clarithromycin as well as ticarcillin plus clavulanic acid. Piperacillin, a penicillin with a polar side chain that enhances penetration into Gram-negative bacteria, is frequently used for the treatment of Pseudomonas aeruginosa infection (often called antipseudomonal penicillin) but has little activity against Staphylococcus aureus. Ticarcillin is also effective against Gram-negative bacteria such as Pseudomonas aeruginosa and Proteus vulgaris. Ticarcillin is one of the few antibiotics effective against Stenotrophomonas maltophilia, which has been implicated in infections in immunocompromised hosts, including preterm neonates.
Cephalosporins are a suitable alternative to ampicillin and gentamicin. Several generations of cephalosporins have been used. Initially, cefazolin, a first-generation cephalosporin, was an alternative to ampicillin in combination with gentamicin. Subsequent-generation cephalosporins have been used without gentamicin (eg, cefotetan, cefoxitin, ceftizoxime, etc.). Newer cephalosporins are of interest because of their broader antimicrobial coverage. Another reason to explore alternative antimicrobial agents is that gentamicin can be nephrotoxic and ototoxic.
Carbapenems, a class of antibiotics usually reserved for known or suspected multi-drug-resistant bacterial infections, include ertapenem, meropenem, and imipenem. These antibiotics are often used for the treatment of multi-resistant Gram-negative bacteria such as Pseudomonas spp. and Acinetobacter spp. Ertapenem is administered intravenously every 24 hours, which makes it an attractive agent. The antibiotics mentioned here have been used for the treatment of patients with clinical chorioamnionitis, and the dose, route, and schedule of administration are shown in Table 5. Some of the antibiotics can also be administered intramuscularly; however, the intravenous route is preferable.
An important consideration is to administer antimicrobials effective against genital mycoplasmas, such as macrolides (azithromycin or clarithromycin). We have used the combination of ceftriaxone, clarithromycin, and metronidazole. The rationale is described in the next section of this article.
There is no evidence that one antimicrobial regimen is superior to others. There is evidence demonstrating that antibiotic use in pregnancy is associated with the risk of antibiotic-resistant strains on the infant gut microbiota.380 The current obstetrical practice has evolved so that mothers with intrapartum fever are given antibiotics and their neonates observe or undergo septic workups. Since a fraction of patients with intrapartum fever has no evidence of infection, a sizable group of term neonates is exposed to antibiotics unnecessarily. This practice is costly, uncomfortable (because of blood culture and lumbar punctures in neonates), creates risk for the emergence of resistant strains of bacteria, and exposes infants to the long-term risks of early antimicrobial exposure (eg, obesity).381
New concepts about the microbiology of intraamniotic infection and implications for antimicrobial agent selection
The use of ampicillin and gentamicin (ampicillin 2 g IV every 6 hours combined with gentamicin 5 mg/kg every 24 hours) is recommended by the American College of Obstetricians and Gynecologists whenever an intraamniotic infection is suspected or confirmed.371 However, these antibiotics are not effective against Ureaplasma spp. or Mycoplasma hominis. These bacteria lack a cell wall; therefore, β-lactams (penicillins and cephalosporins) and glycopeptides (vancomycin) are not effective antimicrobial agents.382, 383 Similarly, gentamicin is also not effective against U. parvum and U. urealyticum.384
Macrolides, such as erythromycin, clarithromycin, and azithromycin, provide good antibacterial activity against Ureaplasma spp.385–387; experimental evidence in non-human primates has shown that azithromycin can eradicate Ureaplasma spp. from the amniotic cavity and can reduce fetal lung injury.388, 389 Macrolides have been used to treat intraamniotic infection as a single agent or in combination with other antibiotics.207, 334, 368, 388, 390–398 In preterm gestations, eradication of intraamniotic infection by macrolides has been shown by serial amniocenteses.392–395 In addition, adjunctive azithromycin has been shown to reduce the rate of surgical site infection in patients undergoing cesarean delivery. Specfically, in a large randomized clinical trial reported by Tita et al, patients at ≥24 weeks of gestation who were to undergo cesarean delivery during labor or those with rupture of the membranes for ≥4 hours were randomly assigned to adjunctive treatment with azithromycin or to a placebo.399 Patients with clinical chorioamnionitis were excluded from this study. All patients received cefazolin, a standard antibiotic prophylaxis. Allergic patients received clindamycin alone or clindamycin plus gentamicin. Patients allocated to receive adjunctive azithromycin had a reduction in the rate of postpartum endometritis [6.1% (61/994) vs 3.8% (39/1,019); p=0.02], wound infection [6.6% (66/994) vs 2.4% (24/1,019); p<0.001], and serious maternal adverse events [2.9% (29/994) vs 1.5% (15/1,019); p=0.03].399 Although there was no difference in the neonatal composite outcome, the findings of this study were considered sufficiently persuasive by professional organizations in the United States to recommend adjunctive azithromycin treatment.400
Clarithromycin has emerged as an effective treatment against Ureaplasma spp. Indeed, it has a higher rate of transplacental passage than azithromycin.401 U. urealyticum isolated from pregnant patients is less resistant (0.9%–25%) to clarithromycin than to other antibiotics,385, 386, 402 and a recent report suggests that clarithromycin reduces the burden of Ureaplasma spp. DNA in the amniotic fluid of patients with preterm PROM.397 Randomized clinical trials comparing ampicillin to gentamycin vs other regimens, which include antibiotics effective against genital mycoplasmas (eg, clarithromycin and azithromycin) in patients with clinical chorioamnionitis, should be considered. Clarithromycin can be given intravenously or orally.
Given that most intraamniotic infections in clinical chorioamnionitis are polymicrobial, we have explored a combination of antimicrobial agents consisting of ceftriaxone, clarithromycin, and metronidazole in preterm gestations. This combination has successfully eradicated intraamniotic infection caused by genital mycoplasmas as well as mixed anaerobic and/or aerobic bacteria.392–395 The rationale for the administration of this antibiotic regimen is the following: 1) ceftriaxone has enhanced coverage of aerobic bacteria and a high rate of transplacental passage403, 404; 2) clarithromycin is effective against Ureaplasma spp.385, 386, 402; and 3) metronidazole has shown optimal coverage of anaerobic microorganisms.405, 406 This combination has been effective in eradicating intraamniotic infection and sterile intraamniotic inflammation in patients with preterm PROM,392, 393 preterm labor with intact membranes,394 cervical insufficiency,395, 396 and threatened midtrimester miscarriage.398 Macrolides not only inhibit bacterial proliferation but also have anti-inflammatory properties, hence their value in treating infections with Ureaplasma spp. and also sterile intraamniotic inflammation.344 In the absence of strong evidence for a specific antibiotic regimen to treat patients with chorioamnionitis at term,368, 407 this new antibiotic regimen can be considered and should be the subject of investigation.
For how long should patients with clinical chorioamnionitis be treated with an antibiotic regimen?
The duration of antibiotic treatment has been the subject of discussion and of clinical trials.408–412 Most professional societies do not recommend continuing antibiotic administration in the postpartum period in cases of vaginal delivery.371, 407, 410 Presumably, this suggestion is based on the notion that intraamniotic infection is confined to the amniotic cavity and the chorioamniotic membranes and that the infectious process is considered resolved after the expulsion of the placenta. Others recommend continuing antibiotics in patients who undergo a cesarean delivery for at least one dose or for 24 to 48 hours.371
Studies about the administration of antibiotics in the postpartum period have generally used a febrile index or febrile morbidity as an endpoint for the cure.413, 414 However, clinicians need to consider that clinical chorioamnionitis is a risk factor for secondary infertility,415 and inadequate treatment of infection may result in this complication. We do not believe that fever, an indication of a systemic host response, is an adequate endpoint to assess the cure of a soft-tissue infection, such as endomyometritis, or the resolution of pelvic peritonitis caused by the spillage of bacteria into the perineal cavity at the time of cesarean delivery. The World Health Organization recommends that patients with puerperal endometritis be treated for at least 24 to 48 hours after the complete resolution of signs and symptoms, ie, fever, uterine tenderness, purulent lochia, and leukocytosis.407
Antipyretics
Maternal hyperthermia, even in the absence of infection, seems to have deleterious effects on the fetus. Experimental studies have shown that maternal hyperthermia leads to changes in fetal hemodynamic and metabolic parameters.416, 417 For example, in non-human primates, hyperthermia, induced with heat from a lamp (in the absence of infection), is associated with the development of fetal tachycardia,417 hypoxia,416 metabolic acidosis,416 and hypotension.416 Adverse neonatal outcomes reported in patients who developed intrapartum fever include low Apgar scores,418–421 acidosis,3 hypotonia,418, 419 requirement of resuscitation immediately after delivery,418 oxygen therapy or mechanical ventilation,3, 418, 419, 422 NICU admission,420–422 neonatal sepsis,3, 420, 421 neurological morbidities,3, 268, 418, 419, 421–426 and neonatal mortality.427 The extent to which these complications are a result of maternal fever is uncertain. A systematic review and meta-analysis show that intrapartum maternal fever increases the odds of adverse neurological outcomes, including neonatal seizures, encephalopathy, or cerebral palsy (OR, 2.48; 95% CI, 2.28–2.70).351 Of note, intrapartum fever remains a significant risk factor for neonatal brain injury (OR, 2.79; 95% CI, 2.54–3.06), after excluding those with a diagnosis of clinical chorioamnionitis.351
Antipyretics have been recommended in patients with clinical chorioamnionitis.94, 369, 371 Acetaminophen is the most frequently used antipyretic and can be administered orally, rectally, or intravenously. Serum peak levels (~12 μg/mL) and half-life (~1.5 hours) of acetaminophen in pregnant women are similar to those observed in non-pregnant adults.428 The conventional oral dose of acetaminophen is 325–650 mg every 4 to 6 hours; total daily doses should not exceed 4 g. Studies on the effect of acetaminophen on maternal and fetal temperatures during labor, as well as on adverse obstetrical and neonatal outcomes, are sparse and show conflicting results. In 1989, a case series described the effects of acetaminophen administration (650 mg rectally) in 8 febrile patients diagnosed with clinical chorioamnionitis.429 When the temperature remained >38.3°C, the dose was repeated for one to two hours. Acetaminophen administration resulted in a mean decrease in temperature of 1.2°C.429 In addition, the fetal heart rate tracings improved once the maternal fever was reduced.429Significant improvements in acid-base status were noted at birth as compared to that of the fetal scalp blood at the peak of the maternal fever.429
A retrospective observational study reported that acetaminophen administration in patients with intrapartum fever was associated with a significant decrease in the frequency of failure to progress in labor in comparison to no administration of acetaminophen [18% (22/122) vs 32% (12/38); p = 0.04].430 On the other hand, there was no evidence that acetaminophen reduced the frequency of meconium-stained amniotic fluid, fetal distress, or NICU admission.
Oral administration of 1000 mg of acetaminophen to patients with intrapartum fever ≥38.0°C (n=18) has been reported to decrease neither maternal axillary nor fetal scalp temperatures even though acetaminophen appeared to halt ongoing increases in fetal temperatures.431 Re-analysis of the study data demonstrated that maternal and fetal temperatures decreased after acetaminophen administration.432 A more recent study of 54 patients who presented with an intrapartum fever ≥38°C, of whom only 3 were diagnosed with clinical chorioamnionitis, reported the effects of oral administration of 650 mg of acetaminophen.433 Overall, no significant differences were observed between patients who received acetaminophen (N=41) and those who did not (N=13) in the frequency of cesarean delivery, presence of meconium, requirement for neonatal bag/mask ventilation or continuous positive pressure ventilation, and NICU admission.433
Intravenous administration of acetaminophen may be an alternative when patients are unable to tolerate oral administration or when an earlier onset of action is desirable. Indeed, intravenous acetaminophen has increased bioavailability and more rapid onset of action.434 A recent report showed that two patients with intrapartum fever and fetal tachycardia had a reduction of maternal temperature and resolution of fetal tachycardia 20 minutes after intravenous administration of 1 g of acetaminophen.435 Each patient had an uncomplicated vaginal delivery of a healthy neonate. The authors argued that the resolution of maternal fever and fetal tachycardia may have prevented cesarean delivery secondary to a non-reassuring fetal heart rate status. A randomized clinical trial is in progress to compare two regimens: acetaminophen 1g IV every 6 hours (up to 2 doses) vs acetaminophen 1 g orally every 6 hours (up to 2 doses) in women in active labor who present with a fever >38.0°C (NCT02625454).436
In summary, although there is no clear evidence that the treatment of intrapartum fever reduces the risk of adverse obstetrical and neonatal outcomes, antipyretics, mainly acetaminophen, can be used to treat hyperthermia as well as to reduce fetal tachycardia. Randomized controlled trials evaluating the effects of acetaminophen on intrapartum fever and on obstetrical and neonatal outcomes in patients with clinical chorioamnionitis are necessary.
Conclusions
Clinical chorioamnionitis is a syndrome defined by intrapartum fever combined with the presence of maternal and fetal signs of systemic inflammation, mainly caused by intraamniotic infection or sterile intraamniotic inflammation. A subset of patients with clinical chorioamnionitis does not have intraamniotic infection or sterile intraamniotic inflammation, and some of these cases are attributed to a systemic maternal inflammatory process related to epidural anesthesia and analgesia. The differential diagnosis of this syndrome and optimal treatment are important issues in clinical obstetrics, and biomarkers for the detection of intraamniotic infection are needed. Genital mycoplasmas are the most frequent microorganisms found in amniotic fluid. The standard treatment is intrapartum administration of antibiotics to reduce the rate of neonatal sepsis. The fetal attack rate by microorganisms in the amniotic cavity is low; however, neonatal sepsis in patients with clinical chorioamnionitis remains an important problem. Antipyretics, mainly acetaminophen, can be used to treat hyperthermia as well as to reduce fetal tachycardia; however, there is no clear evidence that the treatment of intrapartum fever reduces the risk of adverse obstetrical and neonatal outcomes. Important questions for future investigations are the following: 1) can amniotic fluid retrieved by a transcervical collector be used to diagnose intraamniotic inflammation?; 2) can this information be used to select patients who could benefit from antibiotic treatment and those who do not?; 3) is treatment with antimicrobial agents effective against genital mycoplasmas (azithromycin or clarithromycin) warranted to improve maternal and neonatal outcomes?; 4) should neonatologists include tests to identify genital mycoplasmas in septic workups of neonates born to patients with clinical chorioamnionitis?; and 5) what is the etiology of clinical chorioamnionitis in the absence of intraamniotic infection and intraamniotic inflammation, particularly if epidural analgesia or anesthesia has not been used?
Acknowledgments
Dr. B. H. Yoon holds patents for the amniotic fluid transcervical collector, an MMP-8 rapid test, and a point-of-care assay of IL-8 for the diagnosis of intraamniotic inflammation. These patents were awarded when Dr. Yoon was a faculty member at the Seoul National University.
FUNDING
This work was supported, in part, by the Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services (NICHD/NIH/DHHS); and, in part, with Federal funds from NICHD/NIH/DHHS under Contract No. HHSN275201300006C. Dr Gomez-Lopez was also supported by the Wayne State University School of Medicine Perinatal Initiative for Maternal, Perinatal and Child Health. Dr Romero has contributed to this work as part of his official duties as an employee of the United States Federal Government.
This work was conducted at the Perinatology Research Branch, NICHD/NIH/DHHS, in Detroit, Michigan; the Branch has since been renamed as the Pregnancy Research Branch, NICHD/NIH/DHHS.
Footnotes
Disclosure: The authors report no conflicts of interest.
References
- 1.Romero R , Miranda J , Kusanovic JP, et al. Clinical chorioamnionitis at term I: microbiology of the amniotic cavity using cultivation and molecular techniques. J Perinat Med 2015;43:19–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Romero R , Pacora P , Kusanovic JP, et al. Clinical chorioamnionitis at term X: microbiology, clinical signs, placental pathology, and neonatal bacteremia - implications for clinical care. J Perinat Med 2021;49:275–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Alexander JM , McIntire DM , Leveno KJ. Chorioamnionitis and the prognosis for term infants. Obstet Gynecol 1999;94:274–8. [DOI] [PubMed] [Google Scholar]
- 4.Rouse DJ , Landon M , Leveno KJ, et al. The Maternal-Fetal Medicine Units cesarean registry: chorioamnionitis at term and its duration-relationship to outcomes. Am J Obstet Gynecol 2004;191:211–6. [DOI] [PubMed] [Google Scholar]
- 5.Fassett MJ , Wing DA , Getahun D. Temporal trends in chorioamnionitis by maternal race/ethnicity and gestational age (1995–2010). International journal of reproductive medicine 2013;2013:906467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Malloy MH. Chorioamnionitis: epidemiology of newborn management and outcome United States 2008. J Perinatol 2014;34:611–5. [DOI] [PubMed] [Google Scholar]
- 7.Braun D , Bromberger P , Ho NJ , Getahun D. Low Rate of Perinatal Sepsis in Term Infants of Mothers with Chorioamnionitis. Am J Perinatol 2016;33:143–50. [DOI] [PubMed] [Google Scholar]
- 8.Bommarito KM , Gross GA , Willers DM , Fraser VJ , Olsen MA. The Effect of Clinical Chorioamnionitis on Cesarean Delivery in the United States. Health Serv Res 2016;51:1879–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Perry AK , Rossi RM , DeFranco EA. Severe adverse maternal outcomes associated with chorioamnionitis. Am J Obstet Gynecol MFM 2019;1:100027. [DOI] [PubMed] [Google Scholar]
- 10.Racusin DA , Chen HY , Bhalwal A , Wiley R , Chauhan SP. Chorioamnionitis and adverse outcomes in low-risk pregnancies: a population-based study. J Matern Fetal Neonatal Med 2021:1–9. [DOI] [PubMed] [Google Scholar]
- 11.Casey BM , Cox SM. Chorioamnionitis and endometritis. Infect Dis Clin North Am 1997;11:203–22. [DOI] [PubMed] [Google Scholar]
- 12.DeNoble AE , Heine RP , Dotters-Katz SK. Chorioamnionitis and Infectious Complications after Vaginal Delivery. Am J Perinatol 2019;36:1437–41. [DOI] [PubMed] [Google Scholar]
- 13.Venkatesh KK , Glover AV , Vladutiu CJ , Stamilio DM. Association of chorioamnionitis and its duration with adverse maternal outcomes by mode of delivery: a cohort study. BJOG 2019;126:719–27. [DOI] [PubMed] [Google Scholar]
- 14.Eschenbach DA. Acute postpartum infections. Emerg Med Clin North Am 1985;3:87–115. [PubMed] [Google Scholar]
- 15.Dotters-Katz SK , Feldman C , Puechl A , Grotegut CA , Heine RP. Risk factors for post-operative wound infection in the setting of chorioamnionitis and cesarean delivery. J Matern Fetal Neonatal Med 2016;29:1541–5. [DOI] [PubMed] [Google Scholar]
- 16.Witlin AG , Mercer BM , Sibai BM. Septic pelvic thrombophlebitis or refractory postpartum fever of undetermined etiology. J Matern Fetal Med 1996;5:355–8. [DOI] [PubMed] [Google Scholar]
- 17.Goepfert AR , Guinn DA , Andrews WW , Hauth JC. Necrotizing fasciitis after cesarean delivery. Obstet Gynecol 1997;89:409–12. [DOI] [PubMed] [Google Scholar]
- 18.Kang-Auger G , Chassé M , Quach C , Ayoub A , Auger N. Necrotizing Fasciitis: Association with Pregnancy-related Risk Factors Early in Life. Yale J Biol Med 2021;94:573–84. [PMC free article] [PubMed] [Google Scholar]
- 19.Koh KS , Chan FH , Monfared AH , Ledger WJ , Paul RH. The changing perinatal and maternal outcome in chorioamnionitis. Obstet Gynecol 1979;53:730–4. [PubMed] [Google Scholar]
- 20.Gibbs RS , Blanco JD , St Clair PJ , Castaneda YS. Quantitative bacteriology of amniotic fluid from women with clinical intraamniotic infection at term. J Infect Dis 1982;145:1–8. [DOI] [PubMed] [Google Scholar]
- 21.Romero R , Kadar N , Vaisbuch E , Hassan SS. Maternal death following cardiopulmonary collapse after delivery: amniotic fluid embolism or septic shock due to intrauterine infection? Am J Reprod Immunol 2010;64:113–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chebbo A , Tan S , Kassis C , Tamura L , Carlson RW. Maternal Sepsis and Septic Shock. Crit Care Clin 2016;32:119–35. [DOI] [PubMed] [Google Scholar]
- 23.Ericson JE , Laughon MM. Chorioamnionitis: implications for the neonate. Clin Perinatol 2015;42:155–65, ix. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Randis TM , Rice MM , Myatt L, et al. Incidence of early-onset sepsis in infants born to women with clinical chorioamnionitis. J Perinat Med 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Venkatesh KK , Jackson W , Hughes BL , Laughon MM , Thorp JM , Stamilio DM. Association of chorioamnionitis and its duration with neonatal morbidity and mortality. J Perinatol 2019;39:673–82. [DOI] [PubMed] [Google Scholar]
- 26.Stoll BJ , Puopolo KM , Hansen NI, et al. Early-Onset Neonatal Sepsis 2015 to 2017, the Rise of Escherichia coli, and the Need for Novel Prevention Strategies. JAMA pediatrics 2020;174:e200593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kim CJ , Romero R , Chaemsaithong P , Chaiyasit N , Yoon BH , Kim YM. Acute chorioamnionitis and funisitis: definition, pathologic features, and clinical significance. Am J Obstet Gynecol 2015;213:S29–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Romero R , Espinoza J , Goncalves LF , Kusanovic JP , Friel L , Hassan S. The role of inflammation and infection in preterm birth. Semin Reprod Med 2007;25:21–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Liu Y , Li X , Zhu B, et al. Midtrimester amniotic fluid from healthy pregnancies has no microorganisms using multiple methods of microbiologic inquiry. Am J Obstet Gynecol 2020;223:248.e1–48.e21. [DOI] [PubMed] [Google Scholar]
- 30.Zhou X , Brotman RM , Gajer P, et al. Recent advances in understanding the microbiology of the female reproductive tract and the causes of premature birth. Infect Dis Obstet Gynecol 2010;2010:737425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Lamont RF , Sobel JD , Akins RA, et al. The vaginal microbiome: new information about genital tract flora using molecular based techniques. BJOG 2011;118:533–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Macklaim JM , Cohen CR , Donders G, et al. Exploring a road map to counter misconceptions about the cervicovaginal microbiome and disease. Reprod Sci 2012;19:1154–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Romero R , Korzeniewski SJ. Are infants born by elective cesarean delivery without labor at risk for developing immune disorders later in life? Am J Obstet Gynecol 2013;208:243–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Palmer C , Bik EM , DiGiulio DB , Relman DA , Brown PO. Development of the human infant intestinal microbiota. PLoS Biol 2007;5:e177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Martin R , Makino H , Cetinyurek Yavuz A, et al. Early-Life Events, Including Mode of Delivery and Type of Feeding, Siblings and Gender, Shape the Developing Gut Microbiota. PLoS One 2016;11:e0158498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Dominguez-Bello MG , Costello EK , Contreras M, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proceedings of the National Academy of Sciences 2010;107:11971–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bäckhed F , Roswall J , Peng Y, et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell host & microbe 2015;17:690–703. [DOI] [PubMed] [Google Scholar]
- 38.Stokholm J , Thorsen J , Chawes BL, et al. Cesarean section changes neonatal gut colonization. J Allergy Clin Immunol 2016;138:881–89.e2. [DOI] [PubMed] [Google Scholar]
- 39.Chu DM , Ma J , Prince AL , Antony KM , Seferovic MD , Aagaard KM. Maturation of the infant microbiome community structure and function across multiple body sites and in relation to mode of delivery. Nat Med 2017;23:314–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Mitchell CM , Mazzoni C , Hogstrom L, et al. Delivery Mode Affects Stability of Early Infant Gut Microbiota. Cell reports Medicine 2020;1:100156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Jaiman S , Romero R , Gotsch F, et al. Fetal sepsis: a cause of stillbirth. J Matern Fetal Neonatal Med 2022:1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Adkins B , Leclerc C , Marshall-Clarke S. Neonatal adaptive immunity comes of age. Nat Rev Immunol 2004;4:553–64. [DOI] [PubMed] [Google Scholar]
- 43.Levy O Innate immunity of the newborn: basic mechanisms and clinical correlates. Nat Rev Immunol 2007;7:379–90. [DOI] [PubMed] [Google Scholar]
- 44.Ygberg S , Nilsson A. The developing immune system - from foetus to toddler. Acta Paediatr 2012;101:120–7. [DOI] [PubMed] [Google Scholar]
- 45.Cuenca AG , Wynn JL , Moldawer LL , Levy O. Role of innate immunity in neonatal infection. Am J Perinatol 2013;30:105–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Basha S , Surendran N , Pichichero M. Immune responses in neonates. Expert Rev Clin Immunol 2014;10:1171–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jung E , Romero R , Yeo L, et al. The fetal inflammatory response syndrome: the origins of a concept, pathophysiology, diagnosis, and obstetrical implications. Semin Fetal Neonatal Med 2020;25:101146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Martinez-Varea A , Romero R , Xu Y, et al. Clinical chorioamnionitis at term VII: the amniotic fluid cellular immune response. J Perinat Med 2017;45:523–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gomez-Lopez N , Romero R , Xu Y, et al. Are amniotic fluid neutrophils in women with intraamniotic infection and/or inflammation of fetal or maternal origin? Am J Obstet Gynecol 2017;217:693.e1–93.e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Gomez-Lopez N , Romero R , Xu Y, et al. The immunophenotype of amniotic fluid leukocytes in normal and complicated pregnancies. Am J Reprod Immunol 2018;79:e12827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gomez-Lopez N , Romero R , Galaz J, et al. Cellular immune responses in amniotic fluid of women with preterm labor and intra-amniotic infection or intra-amniotic inflammation. Am J Reprod Immunol 2019;82:e13171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Gomez-Lopez N , Romero R , Leng Y, et al. The origin of amniotic fluid monocytes/macrophages in women with intra-amniotic inflammation or infection. J Perinat Med 2019;47:822–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gomez-Lopez N , Romero R , Xu Y, et al. Fetal T Cell Activation in the Amniotic Cavity during Preterm Labor: A Potential Mechanism for a Subset of Idiopathic Preterm Birth. J Immunol 2019;203:1793–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Galaz J , Romero R , Slutsky R, et al. Cellular immune responses in amniotic fluid of women with preterm prelabor rupture of membranes. J Perinat Med 2020;48:222–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Galaz J , Romero R , Xu Y, et al. Cellular immune responses in amniotic fluid of women with a sonographic short cervix. J Perinat Med 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Galaz J , Romero R , Xu Y, et al. Cellular immune responses in amniotic fluid of women with preterm clinical chorioamnionitis. Inflamm Res 2020;69:203–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Romero R , Quintero R , Nores J, et al. Amniotic fluid white blood cell count: a rapid and simple test to diagnose microbial invasion of the amniotic cavity and predict preterm delivery. Am J Obstet Gynecol 1991;165:821–30. [DOI] [PubMed] [Google Scholar]
- 58.Romero R , Yoon BH , Mazor M, et al. The diagnostic and prognostic value of amniotic fluid white blood cell count, glucose, interleukin-6, and gram stain in patients with preterm labor and intact membranes. Am J Obstet Gynecol 1993;169:805–16. [DOI] [PubMed] [Google Scholar]
- 59.Romero R , Yoon BH , Mazor M, et al. A comparative study of the diagnostic performance of amniotic fluid glucose, white blood cell count, interleukin-6, and gram stain in the detection of microbial invasion in patients with preterm premature rupture of membranes. Am J Obstet Gynecol 1993;169:839–51. [DOI] [PubMed] [Google Scholar]
- 60.Gomez R , Romero R , Galasso M , Behnke E , Insunza A , Cotton DB. The value of amniotic fluid interleukin-6, white blood cell count, and gram stain in the diagnosis of microbial invasion of the amniotic cavity in patients at term. Am J Reprod Immunol 1994;32:200–10. [DOI] [PubMed] [Google Scholar]
- 61.Yoon BH , Jun JK , Park KH , Syn HC , Gomez R , Romero R. Serum C-reactive protein, white blood cell count, and amniotic fluid white blood cell count in women with preterm premature rupture of membranes. Obstet Gynecol 1996;88:1034–40. [DOI] [PubMed] [Google Scholar]
- 62.Romero R , Avila C , Santhanam U , Sehgal PB. Amniotic fluid interleukin 6 in preterm labor. Association with infection. J Clin Invest 1990;85:1392–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Santhanam U , Avila C , Romero R, et al. Cytokines in normal and abnormal parturition: elevated amniotic fluid interleukin-6 levels in women with premature rupture of membranes associated with intrauterine infection. Cytokine 1991;3:155–63. [DOI] [PubMed] [Google Scholar]
- 64.Romero R , Sepulveda W , Kenney JS , Archer LE , Allison AC , Sehgal PB. Interleukin 6 determination in the detection of microbial invasion of the amniotic cavity. Ciba Found Symp 1992;167:205–20; discussion 20–3. [DOI] [PubMed] [Google Scholar]
- 65.Romero R , Yoon BH , Kenney JS , Gomez R , Allison AC , Sehgal PB. Amniotic fluid interleukin-6 determinations are of diagnostic and prognostic value in preterm labor. Am J Reprod Immunol 1993;30:167–83. [DOI] [PubMed] [Google Scholar]
- 66.Andrews WW , Hauth JC , Goldenberg RL , Gomez R , Romero R , Cassell GH. Amniotic fluid interleukin-6: correlation with upper genital tract microbial colonization and gestational age in women delivered after spontaneous labor versus indicated delivery. Am J Obstet Gynecol 1995;173:606–12. [DOI] [PubMed] [Google Scholar]
- 67.Yoon BH , Romero R , Kim CJ, et al. Amniotic fluid interleukin-6: a sensitive test for antenatal diagnosis of acute inflammatory lesions of preterm placenta and prediction of perinatal morbidity. Am J Obstet Gynecol 1995;172:960–70. [DOI] [PubMed] [Google Scholar]
- 68.Yoon BH , Romero R , Moon JB, et al. Clinical significance of intra-amniotic inflammation in patients with preterm labor and intact membranes. Am J Obstet Gynecol 2001;185:1130–36. [DOI] [PubMed] [Google Scholar]
- 69.Gervasi MT , Romero R , Bracalente G, et al. Midtrimester amniotic fluid concentrations of interleukin-6 and interferon-gamma-inducible protein-10: evidence for heterogeneity of intra-amniotic inflammation and associations with spontaneous early (<32 weeks) and late (>32 weeks) preterm delivery. J Perinat Med 2012;40:329–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Moon JB , Kim JC , Yoon BH, et al. Amniotic fluid matrix metalloproteinase-8 and the development of cerebral palsy. J Perinat Med 2002;30:301–6. [DOI] [PubMed] [Google Scholar]
- 71.Liechty KW , Koenig JM , Mitchell MD , Romero R , Christensen RD. Production of interleukin-6 by fetal and maternal cells in vivo during intraamniotic infection and in vitro after stimulation with interleukin-1. Pediatr Res 1991;29:1–4. [DOI] [PubMed] [Google Scholar]
- 72.Cherouny PH , Pankuch GA , Romero R, et al. Neutrophil attractant/activating peptide-1/interleukin-8: association with histologic chorioamnionitis, preterm delivery, and bioactive amniotic fluid leukoattractants. Am J Obstet Gynecol 1993;169:1299–303. [DOI] [PubMed] [Google Scholar]
- 73.Yoon BH , Romero R , Jun JK, et al. Amniotic fluid cytokines (interleukin-6, tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-8) and the risk for the development of bronchopulmonary dysplasia. Am J Obstet Gynecol 1997;177:825–30. [DOI] [PubMed] [Google Scholar]
- 74.Ghezzi F , Gomez R , Romero R, et al. Elevated interleukin-8 concentrations in amniotic fluid of mothers whose neonates subsequently develop bronchopulmonary dysplasia. Eur J Obstet Gynecol Reprod Biol 1998;78:5–10. [DOI] [PubMed] [Google Scholar]
- 75.Yoon BH , Yang SH , Jun JK , Park KH , Kim CJ , Romero R. Maternal blood C-reactive protein, white blood cell count, and temperature in preterm labor: a comparison with amniotic fluid white blood cell count. Obstet Gynecol 1996;87:231–7. [DOI] [PubMed] [Google Scholar]
- 76.Yoon BH , Romero R , Shim JY , Shim SS , Kim CJ , Jun JK. C-reactive protein in umbilical cord blood: a simple and widely available clinical method to assess the risk of amniotic fluid infection and funisitis. J Matern Fetal Neonatal Med 2003;14:85–90. [DOI] [PubMed] [Google Scholar]
- 77.Shim SS , Romero R , Jun JK , Moon KC , Kim G , Yoon BH. C-reactive protein concentration in vaginal fluid as a marker for intra-amniotic inflammation/infection in preterm premature rupture of membranes. J Matern Fetal Neonatal Med 2005;18:417–22. [DOI] [PubMed] [Google Scholar]
- 78.Sorokin Y , Romero R , Mele L, et al. Maternal serum interleukin-6, C-reactive protein, and matrix metalloproteinase-9 concentrations as risk factors for preterm birth <32 weeks and adverse neonatal outcomes. Am J Perinatol 2010;27:631–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Sorokin Y , Romero R , Mele L, et al. Umbilical cord serum interleukin-6, C-reactive protein, and myeloperoxidase concentrations at birth and association with neonatal morbidities and long-term neurodevelopmental outcomes. Am J Perinatol 2014;31:717–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Maymon E , Romero R , Chaiworapongsa T, et al. Amniotic fluid matrix metalloproteinase-8 in preterm labor with intact membranes. Am J Obstet Gynecol 2001;185:1149–55. [DOI] [PubMed] [Google Scholar]
- 81.Park JS , Romero R , Yoon BH, et al. The relationship between amniotic fluid matrix metalloproteinase-8 and funisitis. Am J Obstet Gynecol 2001;185:1156–61. [DOI] [PubMed] [Google Scholar]
- 82.Yoon BH , Oh SY , Romero R, et al. An elevated amniotic fluid matrix metalloproteinase-8 level at the time of mid-trimester genetic amniocentesis is a risk factor for spontaneous preterm delivery. Am J Obstet Gynecol 2001;185:1162–67. [DOI] [PubMed] [Google Scholar]
- 83.Jung E , Romero R , Yoon BH, et al. Bacteria in the amniotic fluid without inflammation: early colonization vs. contamination. J Perinat Med 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Romero R , Miranda J , Chaiworapongsa T, et al. A novel molecular microbiologic technique for the rapid diagnosis of microbial invasion of the amniotic cavity and intra-amniotic infection in preterm labor with intact membranes. Am J Reprod Immunol 2014;71:330–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Romero R , Miranda J , Chaiworapongsa T, et al. Prevalence and clinical significance of sterile intra-amniotic inflammation in patients with preterm labor and intact membranes. Am J Reprod Immunol 2014;72:458–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Romero R , Miranda J , Chaemsaithong P, et al. Sterile and microbial-associated intra-amniotic inflammation in preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med 2015;28:1394–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Romero R , Miranda J , Chaiworapongsa T, et al. Sterile intra-amniotic inflammation in asymptomatic patients with a sonographic short cervix: prevalence and clinical significance. J Matern Fetal Neonatal Med 2015;28:1343–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Chorioamnionitis MacVicar J.. Clin Obstet Gynecol 1970;13:272–90. [DOI] [PubMed] [Google Scholar]
- 89.Gibbs RS. Diagnosis of intra-amniotic infection. Semin Perinatol 1977;1:71–7. [PubMed] [Google Scholar]
- 90.Hollander D Diagnosis of chorioamnionitis. Clin Obstet Gynecol 1986;29:816–25. [DOI] [PubMed] [Google Scholar]
- 91.Gilstrap LC , 3rd, Cox SM. Acute chorioamnionitis. Obstet Gynecol Clin North Am 1989;16:373–9. [PubMed] [Google Scholar]
- 92.Newton ER. Chorioamnionitis and intraamniotic infection. Clin Obstet Gynecol 1993;36:795–808. [DOI] [PubMed] [Google Scholar]
- 93.Newton ER. Preterm labor, preterm premature rupture of membranes, and chorioamnionitis. Clin Perinatol 2005;32:571–600. [DOI] [PubMed] [Google Scholar]
- 94.Tita AT , Andrews WW. Diagnosis and management of clinical chorioamnionitis. Clin Perinatol 2010;37:339–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Romero R , Chaemsaithong P , Korzeniewski SJ, et al. Clinical chorioamnionitis at term III: how well do clinical criteria perform in the identification of proven intra-amniotic infection? J Perinat Med 2016;44:23–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Driscoll S The placenta and membranes. Philadelphia: Lea & Febiger, 1973. [Google Scholar]
- 97.Russell P Inflammatory lesions of the human placenta: Clinical significance of acute chorioamnionitis. Am J Diagn Gynecol Obstet 1979;2:127–37. [Google Scholar]
- 98.Driscoll SG. Chorioamnionitis: perinatal morbidity and mortality. Pediatr Infect Dis 1986;5:S273–5. [PubMed] [Google Scholar]
- 99.Romero R , Salafia CM , Athanassiadis AP, et al. The relationship between acute inflammatory lesions of the preterm placenta and amniotic fluid microbiology. Am J Obstet Gynecol 1992;166:1382–8. [DOI] [PubMed] [Google Scholar]
- 100.Redline RW , Faye-Petersen O , Heller D, et al. Amniotic infection syndrome: Nosology and reproducibility of placental reaction patterns. Pediatr Dev Pathol 2003;6:435–48. [DOI] [PubMed] [Google Scholar]
- 101.Redline RW. Placental inflammation. Semin Neonatol 2004;9:265–74. [DOI] [PubMed] [Google Scholar]
- 102.Fox H , Sebire N. Infections and inflammatory lesions of the placenta. Pathology of the placenta. Pathology of the placenta Philadelphia: Saunders, 2007. [Google Scholar]
- 103.Menon R , Taylor RN , Fortunato SJ. Chorioamnionitis--a complex pathophysiologic syndrome. Placenta 2010;31:113–20. [DOI] [PubMed] [Google Scholar]
- 104.Redline RW. Inflammatory response in acute chorioamnionitis. Semin Fetal Neonatal Med 2012;17:20–5. [DOI] [PubMed] [Google Scholar]
- 105.Benirschke K , Burton GJ , Baergen RN. Infectious disease. Pathology of the human placenta. Berlin: Springer, 2012. [Google Scholar]
- 106.Romero R , Chaemsaithong P , Docheva N, et al. Clinical chorioamnionitis at term VI: acute chorioamnionitis and funisitis according to the presence or absence of microorganisms and inflammation in the amniotic cavity. J Perinat Med 2016;44:33–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Benirschke K , Clifford SH. Intrauterine bacterial infection of the newborn infant: frozen sections of the cord as an aid to early detection. J Pediatr 1959;54:11–18. [DOI] [PubMed] [Google Scholar]
- 108.Pacora P , Chaiworapongsa T , Maymon E, et al. Funisitis and chorionic vasculitis: the histological counterpart of the fetal inflammatory response syndrome. J Matern Fetal Neonatal Med 2002;11:18–25. [DOI] [PubMed] [Google Scholar]
- 109.Blanc WA. Amniotic infection syndrome; pathogenesis, morphology, and significance in circumnatal mortality. Clin Obstet Gynecol 1959;2:705–34. [PubMed] [Google Scholar]
- 110.Olding L Value of placentitis as a sign of intrauterine infection in human subjects. Acta Pathol Microbiol Scand A 1970;78:256–64. [DOI] [PubMed] [Google Scholar]
- 111.Hillier SL , Martius J , Krohn M , Kiviat N , Holmes KK , Eschenbach DA. A case-control study of chorioamnionic infection and histologic chorioamnionitis in prematurity. N Engl J Med 1988;319:972–8. [DOI] [PubMed] [Google Scholar]
- 112.Hillier SL , Krohn MA , Kiviat NB , Watts DH , Eschenbach DA. Microbiologic causes and neonatal outcomes associated with chorioamnion infection. Am J Obstet Gynecol 1991;165:955–61. [DOI] [PubMed] [Google Scholar]
- 113.Greig PC , Ernest JM , Teot L , Erikson M , Talley R. Amniotic fluid interleukin-6 levels correlate with histologic chorioamnionitis and amniotic fluid cultures in patients in premature labor with intact membranes. Am J Obstet Gynecol 1993;169:1035–44. [DOI] [PubMed] [Google Scholar]
- 114.Altshuler G , Hyde SR. Clinicopathologic implications of placental pathology. Clin Obstet Gynecol 1996;39:549–70. [DOI] [PubMed] [Google Scholar]
- 115.Holzman C , Lin X , Senagore P , Chung H. Histologic chorioamnionitis and preterm delivery. Am J Epidemiol 2007;166:786–94. [DOI] [PubMed] [Google Scholar]
- 116.Kim MJ , Romero R , Kim CJ, et al. Villitis of unknown etiology is associated with a distinct pattern of chemokine up-regulation in the feto-maternal and placental compartments: implications for conjoint maternal allograft rejection and maternal anti-fetal graft-versus-host disease. J Immunol 2009;182:3919–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Kim CJ , Romero R , Kusanovic JP, et al. The frequency, clinical significance, and pathological features of chronic chorioamnionitis: a lesion associated with spontaneous preterm birth. Mod Pathol 2010;23:1000–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Lee J , Romero R , Xu Y, et al. A signature of maternal anti-fetal rejection in spontaneous preterm birth: chronic chorioamnionitis, anti-human leukocyte antigen antibodies, and C4d. PLoS One 2011;6:e16806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Lee J , Romero R , Xu Y, et al. Maternal HLA panel-reactive antibodies in early gestation positively correlate with chronic chorioamnionitis: evidence in support of the chronic nature of maternal anti-fetal rejection. Am J Reprod Immunol 2011;66:510–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Lee J , Romero R , Chaiworapongsa T, et al. Characterization of the fetal blood transcriptome and proteome in maternal anti-fetal rejection: evidence of a distinct and novel type of human fetal systemic inflammatory response. Am J Reprod Immunol 2013;70:265–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Lee J , Romero R , Xu Y, et al. Detection of anti-HLA antibodies in maternal blood in the second trimester to identify patients at risk of antibody-mediated maternal anti-fetal rejection and spontaneous preterm delivery. Am J Reprod Immunol 2013;70:162–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Gomez-Lopez N , StLouis D , Lehr MA , Sanchez-Rodriguez EN , Arenas-Hernandez M. Immune cells in term and preterm labor. Cell Mol Immunol 2014;11:571–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Kim CJ , Romero R , Chaemsaithong P , Kim JS. Chronic inflammation of the placenta: definition, classification, pathogenesis, and clinical significance. Am J Obstet Gynecol 2015;213:S53–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Higgins RD , Saade G , Polin RA, et al. Evaluation and Management of Women and Newborns With a Maternal Diagnosis of Chorioamnionitis: Summary of a Workshop. Obstet Gynecol 2016;127:426–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Apuzzio JJ , Williams SF. Evaluation and Management of Women and Newborns With a Maternal Diagnosis of Chorioamnionitis: Summary of a Workshop. Obstet Gynecol 2016;128:205–6. [DOI] [PubMed] [Google Scholar]
- 126.Barth WH Jr Lost in Translation: The Changing Language of Our Specialty. Obstet Gynecol 2016;127:423–25. [DOI] [PubMed] [Google Scholar]
- 127.Menard MK , Main EK , Currigan SM. Executive summary of the reVITALize initiative: standardizing obstetric data definitions. Obstet Gynecol 2014;124:150–53. [DOI] [PubMed] [Google Scholar]
- 128.reVITALize. Obstetric data definitions. Available at: https://www.acog.org/practice-management/health-it-and-clinical-informatics/revitalize-obstetrics-data-definitions. [Google Scholar]
- 129.Lee SM , Romero R , Park JS , Chaemsaithong P , Jun JK , Yoon BH. A transcervical amniotic fluid collector: a new medical device for the assessment of amniotic fluid in patients with ruptured membranes. J Perinat Med 2015;43:381–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Oh KJ , Lee J , Romero R , Park HS , Hong JS , Yoon BH. A new rapid bedside test to diagnose and monitor intraamniotic inflammation in preterm PROM using transcervically collected fluid. Am J Obstet Gynecol 2020;223:423.e1–23.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.BENIRSCHKE K. Routes and types of infection in the fetus and the newborn. AMA J Dis Child 1960;99:714–21. [DOI] [PubMed] [Google Scholar]
- 132.Romero R , Mazor M. Infection and preterm labor. Clin Obstet Gynecol 1988;31:553–84. [DOI] [PubMed] [Google Scholar]
- 133.Goldenberg RL , Hauth JC , Andrews WW. Intrauterine infection and preterm delivery. N Engl J Med 2000;342:1500–7. [DOI] [PubMed] [Google Scholar]
- 134.Naeye RL , Dellinger WS , Blanc WA. Fetal and maternal features of antenatal bacterial infections. J Pediatr 1971;79:733–9. [DOI] [PubMed] [Google Scholar]
- 135.Naeye RL , Peters EC. Causes and consequences of premature rupture of fetal membranes. Lancet 1980;1:192–4. [DOI] [PubMed] [Google Scholar]
- 136.Evaldson GR , Malmborg AS , Nord CE. Premature rupture of the membranes and ascending infection. Br J Obstet Gynaecol 1982;89:793–801. [DOI] [PubMed] [Google Scholar]
- 137.Romero R , Gomez-Lopez N , Winters AD, et al. Evidence that intra-amniotic infections are often the result of an ascending invasion - a molecular microbiological study. J Perinat Med 2019;47:915–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Blanc WA. Pathways of fetal and early neonatal infection. Viral placentitis, bacterial and fungal chorioamnionitis. J Pediatr 1961;59:473–96. [DOI] [PubMed] [Google Scholar]
- 139.Romero R , Winn HN , Wan M , Hobbins JC. Listeria monocytogenes chorioamnionitis and preterm labor. Am J Perinatol 1988;5:286–8. [DOI] [PubMed] [Google Scholar]
- 140.Robischon K , Amstey MS. Evidence for In utero hematogenous transmission of group B beta-hemolytic streptococcus. Infect Dis Obstet Gynecol 1994;2:184–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Kaul AK , Khan S , Martens MG , Crosson JT , Lupo VR , Kaul R. Experimental gestational pyelonephritis induces preterm births and low birth weights in C3H/HeJ mice. Infect Immun 1999;67:5958–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Jadhav AR , Belfort MA , Dildy GA 3rd. Eikenella corrodens chorioamnionitis: modes of infection? Am J Obstet Gynecol 2009;200:e4–5. [DOI] [PubMed] [Google Scholar]
- 143.Sorano S , Goto M , Matsuoka S, et al. Chorioamnionitis caused by Staphylococcus aureus with intact membranes in a term pregnancy: A case of maternal and fetal septic shock. J Infect Chemother 2016;22:261–4. [DOI] [PubMed] [Google Scholar]
- 144.Radochova V , Kacerovska Musilova I , Stepan M, et al. Periodontal disease and intra-amniotic complications in women with preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med 2018;31:2852–61. [DOI] [PubMed] [Google Scholar]
- 145.Dunphy L , Polkampali M , Simmons W , Fowler G. Maternal sepsis caused by Listeria monocytogenes with a fatal fetal outcome. BMJ Case Rep 2022;15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Scott JM , Henderson A. Acute villous inflammation in the placenta following intrauterine transfusion. J Clin Pathol 1972;25:872–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Thadepalli H , Appleman MD , Chan WH , Maidman JC , Davidson EC , Jr. Amniotic fluid contamination during internal fetal monitoring. J Reprod Med 1978;20:93–6. [PubMed] [Google Scholar]
- 148.Fray RE , Davis TP , Brown EA. Clostridium welchii infection after amniocentesis. Br Med J (Clin Res Ed) 1984;288:901–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.McColgin SW , Hess LW , Martin RW , Martin JN Jr. , MORRISON JC. Group B streptococcal sepsis and death in utero following funipuncture. Obstet Gynecol 1989;74:464–5. [PubMed] [Google Scholar]
- 150.Tongsong T , Wanapirak C , Kunavikatikul C , Sirirchotiyakul S , Piyamongkol W , Chanprapaph P. Cordocentesis at 16–24 weeks of gestation: experience of 1,320 cases. Prenat Diagn 2000;20:224–8. [PubMed] [Google Scholar]
- 151.Hamoda H , Chamberlain PF. Clostridium welchii infection following amniocentesis: a case report and review of the literature. Prenat Diagn 2002;22:783–5. [DOI] [PubMed] [Google Scholar]
- 152.Li Kim Mui SV , Chitrit Y , Boulanger MC , Maisonneuve L , Choudat L , de Bièvre P. Sepsis due to Clostridium perfringens after pregnancy termination with feticide by cordocentesis: a case report. Fetal Diagn Ther 2002;17:124–6. [DOI] [PubMed] [Google Scholar]
- 153.Romero R , Shamma F , Avila C, et al. Infection and labor. VI. Prevalence, microbiology, and clinical significance of intraamniotic infection in twin gestations with preterm labor. Am J Obstet Gynecol 1990;163:757–61. [DOI] [PubMed] [Google Scholar]
- 154.Morioka I , Fujibayashi H , Enoki E , Yokoyama N , Yokozaki H , Matsuo M. Congenital pneumonia with sepsis caused by intrauterine infection of Ureaplasma parvum in a term newborn: a first case report. J Perinatol 2010;30:359–62. [DOI] [PubMed] [Google Scholar]
- 155.Erez O , Romero R , Tarca AL, et al. Differential expression pattern of genes encoding for anti-microbial peptides in the fetal membranes of patients with spontaneous preterm labor and intact membranes and those with preterm prelabor rupture of the membranes. J Matern Fetal Neonatal Med 2009;22:1103–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Boldenow E , Jones S , Lieberman RW, et al. Antimicrobial peptide response to group B Streptococcus in human extraplacental membranes in culture. Placenta 2013;34:480–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Zare-Bidaki M , Sadrinia S , Erfani S , Afkar E , Ghanbarzade N. Antimicrobial Properties of Amniotic and Chorionic Membranes: A Comparative Study of Two Human Fetal Sacs. Journal of reproduction & infertility 2017;18:218–24. [PMC free article] [PubMed] [Google Scholar]
- 158.Boldenow E , Gendrin C , Ngo L, et al. Group B Streptococcus circumvents neutrophils and neutrophil extracellular traps during amniotic cavity invasion and preterm labor. Science immunology 2016;1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Gomez-Lopez N , Romero R , Leng Y, et al. Neutrophil extracellular traps in acute chorioamnionitis: A mechanism of host defense. Am J Reprod Immunol 2017;77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Doster RS , Kirk LA , Tetz LM , Rogers LM , Aronoff DM , Gaddy JA. Staphylococcus aureus Infection of Human Gestational Membranes Induces Bacterial Biofilm Formation and Host Production of Cytokines. J Infect Dis 2017;215:653–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Kothary V , Doster RS , Rogers LM, et al. Group B Streptococcus Induces Neutrophil Recruitment to Gestational Tissues and Elaboration of Extracellular Traps and Nutritional Immunity. Frontiers in cellular and infection microbiology 2017;7:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Romero R , Mazor M , Morrotti R, et al. Infection and labor. VII. Microbial invasion of the amniotic cavity in spontaneous rupture of membranes at term. Am J Obstet Gynecol 1992;166:129–33. [DOI] [PubMed] [Google Scholar]
- 163.Seong HS , Lee SE , Kang JH , Romero R , Yoon BH. The frequency of microbial invasion of the amniotic cavity and histologic chorioamnionitis in women at term with intact membranes in the presence or absence of labor. Am J Obstet Gynecol 2008;199:375 e1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Roberts DJ. Chapter 18. Perinatal Infections. In: Kradin, Richard L. Diagnostic Pathology of Infectious Disease, Second Edition, 2018. pp. 501. [Google Scholar]
- 165.Gilstrap LC 3rd , CUNNINGHAM FG. The bacterial pathogenesis of infection following cesarean section. Obstet Gynecol 1979;53:545–9. [PubMed] [Google Scholar]
- 166.Zervomanolakis I , Ott HW , Hadziomerovic D, et al. Physiology of upward transport in the human female genital tract. Ann N Y Acad Sci 2007;1101:1–20. [DOI] [PubMed] [Google Scholar]
- 167.Romero R , Nores J , Mazor M, et al. Microbial invasion of the amniotic cavity during term labor. Prevalence and clinical significance. J Reprod Med 1993;38:543–8. [PubMed] [Google Scholar]
- 168.Santolaya-Forgas J , Romero R , Espinoza J, et al. Prelabor rupture of the membranes. In: Albert Reece MDPDMBA E, Hobbins JC, eds. Clinical Obstetrics: The Fetus and Mother: Blackwell Publishing Ltd, 2008. [Google Scholar]
- 169.Schutte MF , Treffers PE , Kloosterman GJ , Soepatmi S. Management of premature rupture of membranes: the risk of vaginal examination to the infant. Am J Obstet Gynecol 1983;146:395–400. [DOI] [PubMed] [Google Scholar]
- 170.Soper DE , Mayhall CG , Dalton HP. Risk factors for intraamniotic infection: a prospective epidemiologic study. Am J Obstet Gynecol 1989;161:562–6; discussion 66–8. [DOI] [PubMed] [Google Scholar]
- 171.Adoni A , Ben Chetrit A , Zacut D , Palti Z , Hurwitz A. Prolongation of the latent period in patients with premature rupture of the membranes by avoiding digital examination. Int J Gynaecol Obstet 1990;32:19–21. [DOI] [PubMed] [Google Scholar]
- 172.Lewis DF , Major CA , Towers CV , Asrat T , Harding JA , Garite TJ. Effects of digital vaginal examinations on latency period in preterm premature rupture of membranes. Obstet Gynecol 1992;80:630–4. [PubMed] [Google Scholar]
- 173.Sukcharoen N , Vasuratna A. Effects of digital cervical examinations on duration of latency period, maternal and neonatal outcome in preterm premature rupture of membranes. J Med Assoc Thai 1993;76:203–9. [PubMed] [Google Scholar]
- 174.Seaward PG , Hannah ME , Myhr TL, et al. International Multicentre Term Prelabor Rupture of Membranes Study: evaluation of predictors of clinical chorioamnionitis and postpartum fever in patients with prelabor rupture of membranes at term. Am J Obstet Gynecol 1997;177:1024–9. [DOI] [PubMed] [Google Scholar]
- 175.Gluck O , Mizrachi Y , Ganer Herman H , Bar J , Kovo M , Weiner E. The correlation between the number of vaginal examinations during active labor and febrile morbidity, a retrospective cohort study. BMC Pregnancy Childbirth 2020;20:246. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Gomez Slagle HB , Hoffman MK , Fonge YN , Caplan R , Sciscione AC. Incremental risk of clinical chorioamnionitis associated with cervical examination. Am J Obstet Gynecol MFM 2022;4:100524. [DOI] [PubMed] [Google Scholar]
- 177.Laibl VR , Sheffield JS , Roberts S , McIntire DD , Wendel GD Jr. Recurrence of clinical chorioamnionitis in subsequent pregnancies. Obstet Gynecol 2006;108:1493–7. [DOI] [PubMed] [Google Scholar]
- 178.Cohen-Cline HN , Kahn TR , Hutter CM. A population-based study of the risk of repeat clinical chorioamnionitis in Washington State, 1989–2008. Am J Obstet Gynecol 2012;207:473.e1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Sperling MM , Sie L , Leonard SA , Girsen AI , Lee HC , Gibbs RS. Effect of gestational age at first delivery and interpregnancy interval on the recurrence of clinical chorioamnionitis. AJOG global reports 2022;2:100116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Blanco JD , Gibbs RS , Malherbe H , Strickland-Cholmley M , St Clair PJ , Castaneda YS. A controlled study of genital mycoplasmas in amniotic fluid from patients with intra-amniotic infection. J Infect Dis 1983;147:650–3. [DOI] [PubMed] [Google Scholar]
- 181.Gibbs RS , Weiner MH , Walmer K , St Clair PJ. Microbiologic and serologic studies of Gardnerella vaginalis in intra-amniotic infection. Obstet Gynecol 1987;70:187–90. [PubMed] [Google Scholar]
- 182.Sweeney EL , Dando SJ , Kallapur SG , Knox CL. The Human Ureaplasma Species as Causative Agents of Chorioamnionitis. Clin Microbiol Rev 2017;30:349–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Romero R , Gomez-Lopez N , Kusanovic JP, et al. Clinical Chorioamnionitis at Term: New Insights into the Etiology, Microbiology, and the Fetal, Maternal and Amniotic Cavity Inflammatory Responses. Nogyogyaszati es szuleszeti tovabbkepzo szemle 2018;20:103–12. [PMC free article] [PubMed] [Google Scholar]
- 184.Kim SK , Romero R , Kusanovic JP, et al. The prognosis of pregnancy conceived despite the presence of an intrauterine device (IUD). J Perinat Med 2010;38:45–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Kacerovsky M , Romero R , Pliskova L, et al. Presence of Chlamydia trachomatis DNA in the amniotic fluid in women with preterm prelabor rupture of membranes. J Matern Fetal Neonatal Med 2019:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Romero R , Quintero R , Oyarzun E, et al. Intraamniotic infection and the onset of labor in preterm premature rupture of the membranes. Am J Obstet Gynecol 1988;159:661–6. [DOI] [PubMed] [Google Scholar]
- 187.Romero R , Sirtori M , Oyarzun E, et al. Infection and labor. V. Prevalence, microbiology, and clinical significance of intraamniotic infection in women with preterm labor and intact membranes. Am J Obstet Gynecol 1989;161:817–24. [DOI] [PubMed] [Google Scholar]
- 188.Romero R , Ghidini A , Mazor M , Behnke E. Microbial invasion of the amniotic cavity in premature rupture of membranes. Clin Obstet Gynecol 1991;34:769–78. [DOI] [PubMed] [Google Scholar]
- 189.Shim S , Yoon BH , Romero R, et al. The clinical significance of detecting Ureaplasma Urealyticum by PCR in the amniotic fluid of patients with preterm labor and intact membranes. Am J Obstet Gynecol 2003;187:S129. [DOI] [PubMed] [Google Scholar]
- 190.Witt A , Berger A , Gruber CJ, et al. Increased intrauterine frequency of Ureaplasma urealyticum in women with preterm labor and preterm premature rupture of the membranes and subsequent cesarean delivery. Am J Obstet Gynecol 2005;193:1663–9. [DOI] [PubMed] [Google Scholar]
- 191.Gomez R , Romero R , Nien JK, et al. A short cervix in women with preterm labor and intact membranes: a risk factor for microbial invasion of the amniotic cavity. Am J Obstet Gynecol 2005;192:678–89. [DOI] [PubMed] [Google Scholar]
- 192.Bujold E , Morency AM , Rallu F, et al. Bacteriology of amniotic fluid in women with suspected cervical insufficiency. J Obstet Gynaecol Can 2008;30:882–87. [DOI] [PubMed] [Google Scholar]
- 193.Madan I , Romero R , Kusanovic JP, et al. The frequency and clinical significance of intra-amniotic infection and/or inflammation in women with placenta previa and vaginal bleeding: an unexpected observation. J Perinat Med 2010;38:275–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Oh KJ , Lee SE , Jung H , Kim G , Romero R , Yoon BH. Detection of ureaplasmas by the polymerase chain reaction in the amniotic fluid of patients with cervical insufficiency. J Perinat Med 2010;38:261–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.DiGiulio DB , Romero R , Kusanovic JP, et al. Prevalence and diversity of microbes in the amniotic fluid, the fetal inflammatory response, and pregnancy outcome in women with preterm pre-labor rupture of membranes. Am J Reprod Immunol 2010;64:38–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Oh KJ , Lee KA , Sohn YK, et al. Intraamniotic infection with genital mycoplasmas exhibits a more intense inflammatory response than intraamniotic infection with other microorganisms in patients with preterm premature rupture of membranes. Am J Obstet Gynecol 2010;203:211.e1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Combs CA , Gravett M , Garite TJ, et al. Amniotic fluid infection, inflammation, and colonization in preterm labor with intact membranes. Am J Obstet Gynecol 2014;210:125.e1–25.e15. [DOI] [PubMed] [Google Scholar]
- 198.Roberts S , Maccato M , Faro S , Pinell P. The microbiology of post-cesarean wound morbidity. Obstet Gynecol 1993;81:383–6. [PubMed] [Google Scholar]
- 199.Andrews WW , Shah SR , Goldenberg RL , Cliver SP , Hauth JC , Cassell GH. Association of post-cesarean delivery endometritis with colonization of the chorioamnion by Ureaplasma urealyticum. Obstet Gynecol 1995;85:509–14. [DOI] [PubMed] [Google Scholar]
- 200.Watts DH , Eschenbach DA , Kenny GE. Early postpartum endometritis: the role of bacteria, genital mycoplasmas, and Chlamydia trachomatis. Obstet Gynecol 1989;73:52–60. [PubMed] [Google Scholar]
- 201.Kim M , Kim G , Romero R , Shim SS , Kim EC , Yoon BH. Biovar diversity of Ureaplasma urealyticum in amniotic fluid: distribution, intrauterine inflammatory response and pregnancy outcomes. J Perinat Med 2003;31:146–52. [DOI] [PubMed] [Google Scholar]
- 202.Oh KJ , Romero R , Park JY , Hong JS , Yoon BH. The earlier the gestational age, the greater the intensity of the intra-amniotic inflammatory response in women with preterm premature rupture of membranes and amniotic fluid infection by Ureaplasma species. J Perinat Med 2019;47:516–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Aaltonen R , Heikkinen J , Vahlberg T , Jensen JS , Alanen A. Local inflammatory response in choriodecidua induced by Ureaplasma urealyticum. BJOG 2007;114:1432–5. [DOI] [PubMed] [Google Scholar]
- 204.Kelleher MA , Liu Z , Wang X, et al. Beyond the uterine environment: a nonhuman primate model to investigate maternal-fetal and neonatal outcomes following chronic intrauterine infection. Pediatr Res 2017;82:244–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Novy MJ , Duffy L , Axthelm MK, et al. Ureaplasma parvum or Mycoplasma hominis as sole pathogens cause chorioamnionitis, preterm delivery, and fetal pneumonia in rhesus macaques. Reprod Sci 2009;16:56–70. [DOI] [PubMed] [Google Scholar]
- 206.Yoder BA , Coalson JJ , Winter VT , Siler-Khodr T , Duffy LB , Cassell GH. Effects of antenatal colonization with ureaplasma urealyticum on pulmonary disease in the immature baboon. Pediatr Res 2003;54:797–807. [DOI] [PubMed] [Google Scholar]
- 207.Motomura K , Romero R , Xu Y, et al. Intra-Amniotic Infection with Ureaplasma parvum Causes Preterm Birth and Neonatal Mortality That Are Prevented by Treatment with Clarithromycin. mBio 2020;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Kelleher MA , Lee JY , Roberts VHJ, et al. Maternal azithromycin therapy for Ureaplasma parvum intraamniotic infection improves fetal hemodynamics in a nonhuman primate model. Am J Obstet Gynecol 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Quinn PA , Gillan JE , Markestad T, et al. Intrauterine infection with Ureaplasma urealyticum as a cause of fatal neonatal pneumonia. Pediatr Infect Dis 1985;4:538–43. [DOI] [PubMed] [Google Scholar]
- 210.Waites KB , Crouse DT , Philips JB 3rd, CANUPP KC, CASSELL GH. Ureaplasmal pneumonia and sepsis associated with persistent pulmonary hypertension of the newborn. Pediatrics 1989;83:79–85. [PubMed] [Google Scholar]
- 211.Brus F , van Waarde WM , Schoots C , Oetomo SB. Fatal ureaplasmal pneumonia and sepsis in a newborn infant. Eur J Pediatr 1991;150:782–3. [DOI] [PubMed] [Google Scholar]
- 212.Gewitz M , Dinwiddie R , Rees L, et al. Mycoplasma hominis. A cause of neonatal meningitis. Arch Dis Child 1979;54:231–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Waites KB , Duffy LB , Crouse DT, et al. Mycoplasmal infections of cerebrospinal fluid in newborn infants from a community hospital population. Pediatr Infect Dis J 1990;9:241–5. [DOI] [PubMed] [Google Scholar]
- 214.Stahelin-Massik J , Levy F , Friderich P , Schaad UB. Meningitis caused by Ureaplasma urealyticum in a full term neonate. Pediatr Infect Dis J 1994;13:419–21. [PubMed] [Google Scholar]
- 215.Alonso-Vega C , Wauters N , Vermeylen D , Muller MF , Serruys E. A fatal case of Mycoplasma hominis meningoencephalitis in a full-term newborn. J Clin Microbiol 1997;35:286–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Knausz M , Niederland T , Dosa E , Rozgonyi F. Meningo-encephalitis in a neonate caused by maternal Mycoplasma hominis treated successfully with chloramphenicol. J Med Microbiol 2002;51:187–8. [DOI] [PubMed] [Google Scholar]
- 217.Rao RP , Ghanayem NS , Kaufman BA , Kehl KS , Gregg DC , Chusid MJ. Mycoplasma hominis and Ureaplasma species brain abscess in a neonate. Pediatr Infect Dis J 2002;21:1083–5. [DOI] [PubMed] [Google Scholar]
- 218.Wolthers KC , Kornelisse RF , Platenkamp G, et al. A case of Mycoplasma hominis meningo-encephalitis in a full-term infant: rapid recovery after start of treatment with ciprofloxacin. Eur J Pediatr 2003;162:514–16. [DOI] [PubMed] [Google Scholar]
- 219.Hata A , Honda Y , Asada K , Sasaki Y , Kenri T , Hata D. Mycoplasma hominis meningitis in a neonate: case report and review. J Infect 2008;57:338–43. [DOI] [PubMed] [Google Scholar]
- 220.Biran V , Dumitrescu AM , Doit C, et al. Ureaplasma parvum meningitis in a full-term newborn. Pediatr Infect Dis J 2010;29:1154. [DOI] [PubMed] [Google Scholar]
- 221.Goldenberg RL , Andrews WW , Goepfert AR, et al. The Alabama Preterm Birth Study: umbilical cord blood Ureaplasma urealyticum and Mycoplasma hominis cultures in very preterm newborn infants. Am J Obstet Gynecol 2008;198:43 e1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Romero R , Garite TJ. Twenty percent of very preterm neonates (23–32 weeks of gestation) are born with bacteremia caused by genital Mycoplasmas. Am J Obstet Gynecol 2008;198:1–3. [DOI] [PubMed] [Google Scholar]
- 223.Viscardi RM. Ureaplasma species: role in neonatal morbidities and outcomes. Arch Dis Child Fetal Neonatal Ed 2014;99:F87–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Viscardi RM , Kallapur SG. Role of Ureaplasma Respiratory Tract Colonization in Bronchopulmonary Dysplasia Pathogenesis: Current Concepts and Update. Clin Perinatol 2015;42:719–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Viscardi RM , Terrin ML , Magder LS, et al. Randomised trial of azithromycin to eradicate Ureaplasma in preterm infants. Arch Dis Child Fetal Neonatal Ed 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Romero R , Emamian M , Quintero R , Wan M , Hobbins JC , Mitchell MD. Amniotic fluid prostaglandin levels and intra-amniotic infections. Lancet 1986;1:1380. [DOI] [PubMed] [Google Scholar]
- 227.Romero R , Emamian M , Wan M , Quintero R , Hobbins JC , Mitchell MD. Prostaglandin concentrations in amniotic fluid of women with intra-amniotic infection and preterm labor. Am J Obstet Gynecol 1987;157:1461–7. [DOI] [PubMed] [Google Scholar]
- 228.Romero R , Wu YK , Mazor M , Hobbins JC , Mitchell MD. Amniotic fluid prostaglandin E2 in preterm labor. Prostaglandins Leukot Essent Fatty Acids 1988;34:141–5. [DOI] [PubMed] [Google Scholar]
- 229.Romero R , Wu YK , Sirtori M, et al. Amniotic fluid concentrations of prostaglandin F2 alpha, 13,14-dihydro-15-keto-prostaglandin F2 alpha (PGFM) and 11-deoxy-13,14-dihydro-15-keto-11, 16-cyclo-prostaglandin E2 (PGEM-LL) in preterm labor. Prostaglandins 1989;37:149–61. [DOI] [PubMed] [Google Scholar]
- 230.Romero R , Wu YK , Mazor M , Oyarzun E , Hobbins JC , Mitchell MD. Amniotic fluid arachidonate lipoxygenase metabolites in preterm labor. Prostaglandins Leukot Essent Fatty Acids 1989;36:69–75. [DOI] [PubMed] [Google Scholar]
- 231.Bry K , Hallman M. Prostaglandins, inflammation, and preterm labor. Journal of perinatology : official journal of the California Perinatal Association 1989;9:60–5. [PubMed] [Google Scholar]
- 232.Mazor M , Wiznitzer A , Maymon E , Leiberman JR , Cohen A. Changes in amniotic fluid concentrations of prostaglandins E2 and F2 alpha in women with preterm labor. Isr J Med Sci 1990;26:425–8. [PubMed] [Google Scholar]
- 233.Hsu CD , Meaddough E , Aversa K, et al. Dual roles of amniotic fluid nitric oxide and prostaglandin E2 in preterm labor with intra-amniotic infection. Am J Perinatol 1998;15:683–7. [DOI] [PubMed] [Google Scholar]
- 234.Lee SE , Park IS , Romero R , Yoon BH . Amniotic fluid prostaglandin F2 increases even in sterile amniotic fluid and is an independent predictor of impending delivery in preterm premature rupture of membranes. J Matern Fetal Neonatal Med 2009;22:880–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Park JY , Romero R , Lee J , Chaemsaithong P , Chaiyasit N , Yoon BH. An elevated amniotic fluid prostaglandin F concentration is associated with intra-amniotic inflammation/infection, and clinical and histologic chorioamnionitis, as well as impending preterm delivery in patients with preterm labor and intact membranes. J Matern Fetal Neonatal Med 2015:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Kim MJ , Romero R , Gervasi MT, et al. Widespread microbial invasion of the chorioamniotic membranes is a consequence and not a cause of intra-amniotic infection. Lab Invest 2009;89:924–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Romero R , Brody DT , Oyarzun E, et al. Infection and labor. III. Interleukin-1: a signal for the onset of parturition. Am J Obstet Gynecol 1989;160:1117–23. [DOI] [PubMed] [Google Scholar]
- 238.Romero R , Manogue KR , Mitchell MD, et al. Infection and labor. IV. Cachectin-tumor necrosis factor in the amniotic fluid of women with intraamniotic infection and preterm labor. Am J Obstet Gynecol 1989;161:336–41. [DOI] [PubMed] [Google Scholar]
- 239.Romero R , Parvizi ST , Oyarzun E, et al. Amniotic fluid interleukin-1 in spontaneous labor at term. J Reprod Med 1990;35:235–8. [PubMed] [Google Scholar]
- 240.Romero R , Mazor M , Sepulveda W , Avila C , Copeland D , Williams J. Tumor necrosis factor in preterm and term labor. Am J Obstet Gynecol 1992;166:1576–87. [DOI] [PubMed] [Google Scholar]
- 241.Romero R , Mazor M , Brandt F, et al. Interleukin-1 alpha and interleukin-1 beta in preterm and term human parturition. Am J Reprod Immunol 1992;27:117–23. [DOI] [PubMed] [Google Scholar]
- 242.Hillier SL , Witkin SS , Krohn MA , Watts DH , Kiviat NB , Eschenbach DA. The relationship of amniotic fluid cytokines and preterm delivery, amniotic fluid infection, histologic chorioamnionitis, and chorioamnion infection. Obstet Gynecol 1993;81:941–8. [PubMed] [Google Scholar]
- 243.Gomez R , Ghezzi F , Romero R , Munoz H , Tolosa JE , Rojas I. Premature labor and intra-amniotic infection. Clinical aspects and role of the cytokines in diagnosis and pathophysiology. Clinics in perinatology 1995;22:281–342. [PubMed] [Google Scholar]
- 244.Gonzalez-Bosquet E , Cerqueira MJ , Dominguez C , Gasser I , Bermejo B , Cabero L. Amniotic fluid glucose and cytokines values in the early diagnosis of amniotic infection in patients with preterm labor and intact membranes. J Matern Fetal Med 1999;8:155–8. [DOI] [PubMed] [Google Scholar]
- 245.Fortunato SJ , Menon R , Lombardi SJ. Role of tumor necrosis factor-alpha in the premature rupture of membranes and preterm labor pathways. American journal of obstetrics and gynecology 2002;187:1159–62. [DOI] [PubMed] [Google Scholar]
- 246.Figueroa R , Garry D , Elimian A , Patel K , Sehgal PB , Tejani N. Evaluation of amniotic fluid cytokines in preterm labor and intact membranes. J Matern Fetal Neonatal Med 2005;18:241–7. [DOI] [PubMed] [Google Scholar]
- 247.Sadowsky DW , Adams KM , Gravett MG , Witkin SS , Novy MJ. 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. Am J Obstet Gynecol 2006;195:1578–89. [DOI] [PubMed] [Google Scholar]
- 248.Kumar D , Fung W , Moore RM, et al. Proinflammatory cytokines found in amniotic fluid induce collagen remodeling, apoptosis, and biophysical weakening of cultured human fetal membranes. Biol Reprod 2006;74:29–34. [DOI] [PubMed] [Google Scholar]
- 249.Menon R , Fortunato SJ. Infection and the role of inflammation in preterm premature rupture of the membranes. Best Pract Res Clin Obstet Gynaecol 2007;21:467–78. [DOI] [PubMed] [Google Scholar]
- 250.Marconi C , de Andrade Ramos BR , Peracoli JC , Donders GG , da Silva MG. Amniotic fluid interleukin-1 beta and interleukin-6, but not interleukin-8 correlate with microbial invasion of the amniotic cavity in preterm labor. Am J Reprod Immunol 2011;65:549–56. [DOI] [PubMed] [Google Scholar]
- 251.Romero R , Chaemsaithong P , Korzeniewski SJ, et al. Clinical chorioamnionitis at term II: the intra-amniotic inflammatory response. J Perinat Med 2016;44:5–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Romero R , Chaemsaithong P , Docheva N, et al. Clinical chorioamnionitis at term IV: the maternal plasma cytokine profile. J Perinat Med 2016;44:77–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Atici A , Satar M , Cetíner S , Yaman A. Serum tumor necrosis factor-alpha in neonatal sepsis. Am J Perinatol 1997;14:401–4. [DOI] [PubMed] [Google Scholar]
- 254.Yoder PR , Gibbs RS , Blanco JD , Castaneda YS , St Clair PJ. A prospective, controlled study of maternal and perinatal outcome after intra-amniotic infection at term. Am J Obstet Gynecol 1983;145:695–701. [DOI] [PubMed] [Google Scholar]
- 255.Yancey MK , Duff P , Kubilis P , Clark P , Frentzen BH. Risk factors for neonatal sepsis. Obstet Gynecol 1996;87:188–94. [DOI] [PubMed] [Google Scholar]
- 256.Ladfors L , Tessin I , Mattsson LA , Eriksson M , Seeberg S , Fall O. Risk factors for neonatal sepsis in offspring of women with prelabor rupture of the membranes at 34–42 weeks. J Perinat Med 1998;26:94–101. [DOI] [PubMed] [Google Scholar]
- 257.Benitz WE , Gould JB , Druzin ML. Risk factors for early-onset group B streptococcal sepsis: estimation of odds ratios by critical literature review. Pediatrics 1999;103:e77. [DOI] [PubMed] [Google Scholar]
- 258.Volante E , Moretti S , Pisani F , Bevilacqua G. Early diagnosis of bacterial infection in the neonate. J Matern Fetal Neonatal Med 2004;16 Suppl 2:13–6. [DOI] [PubMed] [Google Scholar]
- 259.Uyemura A , Ameel B , Caughey A. 425. Outcomes of chorioamnionitis in term pregnancies. Am J Obstet Gynecol 2014;210:S215. [Google Scholar]
- 260.Romero R , Hanaoka S , Mazor M, et al. Meconium-stained amniotic fluid: a risk factor for microbial invasion of the amniotic cavity. Am J Obstet Gynecol 1991;164:859–62. [DOI] [PubMed] [Google Scholar]
- 261.Wen TS , Eriksen NL , Blanco JD , Graham JM , Oshiro BT , Prieto JA. Association of clinical intra-amniotic infection and meconium. Am J Perinatol 1993;10:438–40. [DOI] [PubMed] [Google Scholar]
- 262.Cleary GM , Wiswell TE. Meconium-stained amniotic fluid and the meconium aspiration syndrome. An update. Pediatr Clin North Am 1998;45:511–29. [DOI] [PubMed] [Google Scholar]
- 263.Romero R , Yoon BH , Chaemsaithong P, et al. Bacteria and endotoxin in meconium-stained amniotic fluid at term: could intra-amniotic infection cause meconium passage? J Matern Fetal Neonatal Med 2014;27:775–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Gallo DM , Romero R , Bosco M, et al. Meconium-stained amniotic fluid. Am J Obstet Gynecol 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Adamson SJ , Alessandri LM , Badawi N , Burton PR , Pemberton PJ , Stanley F. Predictors of neonatal encephalopathy in full-term infants. BMJ 1995;311:598–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Willoughby RE Jr., Nelson KB. Chorioamnionitis and brain injury. Clin Perinatol 2002;29:603–21. [DOI] [PubMed] [Google Scholar]
- 267.Chorioamnionitis Cooke R., maternal fever, and neonatal encephalopathy. Dev Med Child Neurol 2008;50:9. [DOI] [PubMed] [Google Scholar]
- 268.Blume HK , Li CI , Loch CM , Koepsell TD. Intrapartum fever and chorioamnionitis as risks for encephalopathy in term newborns: a case-control study. Dev Med Child Neurol 2008;50:19–24. [DOI] [PubMed] [Google Scholar]
- 269.Versland LB , Sommerfelt K , Elgen I. Maternal signs of chorioamnionitis: persistent cognitive impairment in low-birthweight children. Acta Paediatr 2006;95:231–5. [DOI] [PubMed] [Google Scholar]
- 270.Korzeniewski SJ , Romero R , Cortez J, et al. A “multi-hit” model of neonatal white matter injury: cumulative contributions of chronic placental inflammation, acute fetal inflammation and postnatal inflammatory events. J Perinat Med 2014;42:731–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Vander Haar E , Gyamfi-Bannerman C. Chorioamnionitis and Neurocognitive Development at Age 2 Years. Obstet Gynecol 2016;127:437–41. [DOI] [PubMed] [Google Scholar]
- 272.Freud A , Wainstock T , Sheiner E, et al. Maternal chorioamnionitis & long term neurological morbidity in the offspring. Eur J Paediatr Neurol 2019;23:484–90. [DOI] [PubMed] [Google Scholar]
- 273.Tsamantioti E , Lisonkova S , Muraca G , Örtqvist AK , Razaz N. Chorioamnionitis and risk of long-term neurodevelopmental disorders in offspring; a population-based cohort study. Am J Obstet Gynecol 2022. [DOI] [PubMed] [Google Scholar]
- 274.Moyo SR , Hagerstrand I , Nystrom L, et al. Stillbirths and intrauterine infection, histologic chorioamnionitis and microbiological findings. Int J Gynaecol Obstet 1996;54:115–23. [DOI] [PubMed] [Google Scholar]
- 275.Mendez-Figueroa H , Abramovici A , O’Neil AE , Dahlke J , Pedroza C , Chauhan S. 647. Chorioamnionitis without and with neonatal sepsis: newborn and infant outcomes. Am J Obstet Gynecol 2015;212:S318–19. [Google Scholar]
- 276.Romero R , Chaemsaithong P , Docheva N, et al. Clinical chorioamnionitis at term V: umbilical cord plasma cytokine profile in the context of a systemic maternal inflammatory response. J Perinat Med 2016;44:53–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Chaiworapongsa T , Romero R , Kim JC, et al. Evidence for fetal involvement in the pathologic process of clinical chorioamnionitis. Am J Obstet Gynecol 2002;186:1178–82. [DOI] [PubMed] [Google Scholar]
- 278.Zaretsky MV , Alexander JM , Byrd W , Bawdon RE. Transfer of inflammatory cytokines across the placenta. Obstet Gynecol 2004;103:546–50. [DOI] [PubMed] [Google Scholar]
- 279.Gibbs RS , Castillo MS , Rodgers PJ. Management of acute chorioamnionitis. Am J Obstet Gynecol 1980;136:709–13. [DOI] [PubMed] [Google Scholar]
- 280.Hauth JC , Gilstrap LC 3rd , Hankins GD , Connor KD. Term maternal and neonatal complications of acute chorioamnionitis. Obstet Gynecol 1985;66:59–62. [PubMed] [Google Scholar]
- 281.Gibbs RS , Dinsmoor MJ , Newton ER , Ramamurthy RS. A randomized trial of intrapartum versus immediate postpartum treatment of women with intra-amniotic infection. Obstet Gynecol 1988;72:823–8. [DOI] [PubMed] [Google Scholar]
- 282.Gibbs RS , Duff P. Progress in pathogenesis and management of clinical intraamniotic infection. Am J Obstet Gynecol 1991;164:1317–26. [DOI] [PubMed] [Google Scholar]
- 283.Romero R , Chaiworapongsa T , Savasan ZA, et al. Clinical chorioamnionitis is characterized by changes in the expression of the alarmin HMGB1 and one of its receptors, sRAGE. J Matern Fetal Neonatal Med 2012;25:558–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Romero R , Dey SK , Fisher SJ. Preterm labor: one syndrome, many causes. Science 2014;345:760–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Chaiyasit N , Romero R , Chaemsaithong P, et al. Clinical chorioamnionitis at term VIII: a rapid MMP-8 test for the identification of intra-amniotic inflammation. J Perinat Med 2017;45:539–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Aggarwal D , Kanitkar T , Narouz M , Azadian BS , Moore LSP , Mughal N. Clinical utility and cost-effectiveness of bacterial 16S rRNA and targeted PCR based diagnostic testing in a UK microbiology laboratory network. Sci Rep 2020;10:7965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Chaemsaithong P , Romero R , Pongchaikul P, et al. Rapid identification of intra-amniotic infection by nanopore-based sequencing. J Perinat Med 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Duff P , Sanders R , Gibbs RS. The course of labor in term patients with chorioamnionitis. Am J Obstet Gynecol 1983;147:391–5. [DOI] [PubMed] [Google Scholar]
- 289.Satin AJ , Maberry MC , Leveno KJ , Sherman ML , Kline DM. Chorioamnionitis: a harbinger of dystocia. Obstet Gynecol 1992;79:913–5. [PubMed] [Google Scholar]
- 290.Mark SP , Croughan-Minihane MS , Kilpatrick SJ. Chorioamnionitis and uterine function. Obstet Gynecol 2000;95:909–12. [PubMed] [Google Scholar]
- 291.Maberry MC , Ramin SM , Gilstrap LC 3rd , Leveno KJ , Dax JS. Intrapartum asphyxia in pregnancies complicated by intra-amniotic infection. Obstet Gynecol 1990;76:351–4. [PubMed] [Google Scholar]
- 292.Zackler A , Flood P , Dajao R , Maramara L , Goetzl L. Suspected Chorioamnionitis and Myometrial Contractility: Mechanisms for Increased Risk of Cesarean Delivery and Postpartum Hemorrhage. Reprod Sci 2019;26:178–83. [DOI] [PubMed] [Google Scholar]
- 293.Faro J , Romero R , Schwenkel G, et al. Intra-amniotic inflammation induces preterm birth by activating the NLRP3 inflammasome†. Biol Reprod 2019;100:1290–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Bhatti G , Romero R , Rice GE, et al. Compartmentalized profiling of amniotic fluid cytokines in women with preterm labor. PLoS One 2020;15:e0227881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Unal ER , Cierny JT , Roedner C , Newman R , Goetzl L. Maternal inflammation in spontaneous term labor. American journal of obstetrics and gynecology 2011;204:223.e1–5. [DOI] [PubMed] [Google Scholar]
- 296.Cierny JT , Unal ER , Flood P, et al. Maternal inflammatory markers and term labor performance. Am J Obstet Gynecol 2014;210:447.e1–6. [DOI] [PubMed] [Google Scholar]
- 297.Gallo DM , Romero R , Bosco M, et al. Maternal plasma cytokines and the subsequent risk of uterine atony and postpartum hemorrhage. J Perinat Med 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Rouse DJ , Leindecker S , Landon M, et al. The MFMU Cesarean Registry: uterine atony after primary cesarean delivery. Am J Obstet Gynecol 2005;193:1056–60. [DOI] [PubMed] [Google Scholar]
- 299.Mittal P , Romero R , Tarca AL, et al. Characterization of the myometrial transcriptome and biological pathways of spontaneous human labor at term. J Perinat Med 2010;38:617–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Romero R , Tarca AL , Chaemsaithong P, et al. Transcriptome interrogation of human myometrium identifies differentially expressed sense-antisense pairs of protein-coding and long non-coding RNA genes in spontaneous labor at term. J Matern Fetal Neonatal Med 2014;27:1397–408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Hassan SS , Romero R , Haddad R, et al. The transcriptome of the uterine cervix before and after spontaneous term parturition. Am J Obstet Gynecol 2006;195:778–86. [DOI] [PubMed] [Google Scholar]
- 302.Nhan-Chang CL , Romero R , Tarca AL, et al. Characterization of the transcriptome of chorioamniotic membranes at the site of rupture in spontaneous labor at term. Am J Obstet Gynecol 2010;202:462 e1–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Haddad R , Tromp G , Kuivaniemi H , Chaiworapongsa T , Kim YM , Romero R. Spontaneous labor at term is characterized by a genomic signature of acute inflammation in the chorioamniotic membranes but not in the systemic circulation. Am J Obstet Gynecol 2004;191:S138–S38. [Google Scholar]
- 304.Chaemsaithong P , Madan I , Romero R, et al. Characterization of the myometrial transcriptome in women with an arrest of dilatation during labor. J Perinat Med 2013;41:665–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Mittal P , Romero R , Tarca AL, et al. A molecular signature of an arrest of descent in human parturition. Am J Obstet Gynecol 2011;204:177.e15–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Wendel PJ , Cox SM , Roberts SW , Dax J , Gilstrap LC. Chorioamnionitis: association of nonreassuring fetal heart-rate patterns and interval from diagnosis to delivery on neonatal outcome. Infect Dis Obstet Gynecol 1994;2:162–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Sameshima H , Ikenoue T , Ikeda T , Kamitomo M , Ibara S. Association of nonreassuring fetal heart rate patterns and subsequent cerebral palsy in pregnancies with intrauterine bacterial infection. Am J Perinatol 2005;22:181–7. [DOI] [PubMed] [Google Scholar]
- 308.Preti M , Chandraharan E. Importance of fetal heart rate cycling during the interpretation of the cardiotocograph (CTG). 2018. [Google Scholar]
- 309.Galli L , Dall’Asta A , Whelehan V , Archer A , Chandraharan E. Intrapartum cardiotocography patterns observed in suspected clinical and subclinical chorioamnionitis in term fetuses. J Obstet Gynaecol Res 2019;45:2343–50. [DOI] [PubMed] [Google Scholar]
- 310.Odendaal HJ , Crawford JW. Fetal tachycardia and maternal pyrexia during labour. S Afr Med J 1975;49:1873–5. [PubMed] [Google Scholar]
- 311.Herbst A , Wolner-Hanssen P , Ingemarsson I. Maternal fever in term labour in relation to fetal tachycardia, cord artery acidaemia and neonatal infection. BJOG 1997;104:363–66. [DOI] [PubMed] [Google Scholar]
- 312.Aina-Mumuney AJ , Althaus JE , Henderson JL , Blakemore MC , Johnson EA , Graham EM. Intrapartum electronic fetal monitoring and the identification of systemic fetal inflammation. J Reprod Med 2007;52:762–8. [PubMed] [Google Scholar]
- 313.Buhimschi CS , Abdel-Razeq S , Cackovic M, et al. Fetal heart rate monitoring patterns in women with amniotic fluid proteomic profiles indicative of inflammation. Am J Perinatol 2008;25:359–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314.Schiano MA , Hauth JC , Gilstrap III LC. Second-stage fetal tachycardia and neonatal infection. Am J Obstet Gynecol 1984;148:779–81. [DOI] [PubMed] [Google Scholar]
- 315.Day D , Ugol JH , French JI , Haverkamp A , Wall RE , McGregor JA. Fetal monitoring in perinatal sepsis. Am J Perinatol 1992;9:28–33. [DOI] [PubMed] [Google Scholar]
- 316.Samueloff A , Langer O , Berkus MD , Field NT , Xenakis EM , Piper JM. The effect of clinical chorioamnionitis on cord blood gas at term. Eur J Obstet Gynecol Reprod Biol 1994;54:87–91. [DOI] [PubMed] [Google Scholar]
- 317.Yanowitz TD , Jordan JA , Gilmour CH, et al. Hemodynamic disturbances in premature infants born after chorioamnionitis: association with cord blood cytokine concentrations. Pediatr Res 2002;51:310–6. [DOI] [PubMed] [Google Scholar]
- 318.Burnham P , Gomez-Lopez N , Heyang M, et al. Separating the signal from the noise in metagenomic cell-free DNA sequencing. Microbiome 2020;8:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Chaiworapongsa T , Erez O , Kusanovic JP, et al. Amniotic fluid heat shock protein 70 concentration in histologic chorioamnionitis, term and preterm parturition. J Matern Fetal Neonatal Med 2008;21:449–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.Romero R , Chaiworapongsa T , Alpay Savasan Z, et al. Damage-associated molecular patterns (DAMPs) in preterm labor with intact membranes and preterm PROM: a study of the alarmin HMGB1. J Matern Fetal Neonatal Med 2011;24:1444–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321.Gomez-Lopez N , Romero R , Plazyo O, et al. Intra-Amniotic Administration of HMGB1 Induces Spontaneous Preterm Labor and Birth. Am J Reprod Immunol 2016;75:3–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322.Plazyo O , Romero R , Unkel R, et al. HMGB1 Induces an Inflammatory Response in the Chorioamniotic Membranes That Is Partially Mediated by the Inflammasome. Biol Reprod 2016;95:130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323.Shimizu H , Sakimoto T , Yamagami S. Pro-inflammatory role of NLRP3 inflammasome in experimental sterile corneal inflammation. Sci Rep 2019;9:9596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 324.Martinon F , Pétrilli V , Mayor A , Tardivel A , Tschopp J. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 2006;440:237–41. [DOI] [PubMed] [Google Scholar]
- 325.Schauer C , Janko C , Munoz LE, et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat Med 2014;20:511–7. [DOI] [PubMed] [Google Scholar]
- 326.Li Y , Cao X , Liu Y , Zhao Y , Herrmann M. Neutrophil Extracellular Traps Formation and Aggregation Orchestrate Induction and Resolution of Sterile Crystal-Mediated Inflammation. Front Immunol 2018;9:1559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327.Oppenheim JJ , Yang D. Alarmins: chemotactic activators of immune responses. Curr Opin Immunol 2005;17:359–65. [DOI] [PubMed] [Google Scholar]
- 328.Bianchi ME. DAMPs, PAMPs and alarmins: all we need to know about danger. J Leukoc Biol 2007;81:1–5. [DOI] [PubMed] [Google Scholar]
- 329.Chen GY , Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol 2010;10:826–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330.Davalos AR , Kawahara M , Malhotra GK, et al. p53-dependent release of Alarmin HMGB1 is a central mediator of senescent phenotypes. J Cell Biol 2013;201:613–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331.Fasching G , Haeusler M , Mayr J , Schimpl G , Haas J , Puerstner P. Can levels of interleukins and matrix metalloproteinases in the amniotic fluid predict postnatal bowel function in fetuses with gastroschisis? J Pediatr Surg 2005;40:1887–91. [DOI] [PubMed] [Google Scholar]
- 332.Romero R , Grivel JC , Tarca AL, et al. Evidence of perturbations of the cytokine network in preterm labor. Am J Obstet Gynecol 2015;213:836.e1–36.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 333.Gomez-Lopez N , Romero R , Garcia-Flores V, et al. Inhibition of the NLRP3 inflammasome can prevent sterile intra-amniotic inflammation, preterm labor/birth, and adverse neonatal outcomesdagger. Biol Reprod 2019;100:1306–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334.Galaz J , Romero R , Arenas-Hernandez M, et al. Clarithromycin prevents preterm birth and neonatal mortality by dampening alarmin-induced maternal-fetal inflammation in mice. BMC Pregnancy Childbirth 2022;22:503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335.Galaz J , Romero R , Arenas-Hernandez M , Panaitescu B , Para R , Gomez-Lopez N. Betamethasone as a potential treatment for preterm birth associated with sterile intra-amniotic inflammation: a murine study. J Perinat Med 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336.Butwick AJ , Bentley J , Wong CA , Snowden JM , Sun E , Guo N. United States State-Level Variation in the Use of Neuraxial Analgesia During Labor for Pregnant Women. JAMA network open 2018;1:e186567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 337.Anim-Somuah M , Smyth RM , Cyna AM , Cuthbert A. Epidural versus non-epidural or no analgesia for pain management in labour. The Cochrane database of systematic reviews 2018;5:Cd000331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338.Kearns RJ , Shaw M , Gromski PS , Iliodromiti S , Lawlor DA , Nelson SM. Association of Epidural Analgesia in Women in Labor With Neonatal and Childhood Outcomes in a Population Cohort. JAMA network open 2021;4:e2131683–e83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339.Ramin SM , Gambling DR , Lucas MJ , Sharma SK , Sidawi JE , Leveno KJ. Randomized trial of epidural versus intravenous analgesia during labor. Obstet Gynecol 1995;86:783–9. [DOI] [PubMed] [Google Scholar]
- 340.Sharma SK , Sidawi JE , Ramin SM , Lucas MJ , Leveno KJ , Cunningham FG. Cesarean delivery: a randomized trial of epidural versus patient-controlled meperidine analgesia during labor. Anesthesiology 1997;87:487–94. [DOI] [PubMed] [Google Scholar]
- 341.Lucas MJ , Sharma SK , McIntire DD, et al. A randomized trial of labor analgesia in women with pregnancy-induced hypertension. Am J Obstet Gynecol 2001;185:970–5. [DOI] [PubMed] [Google Scholar]
- 342.Sharma SK , Alexander JM , Messick G, et al. Cesarean delivery: a randomized trial of epidural analgesia versus intravenous meperidine analgesia during labor in nulliparous women. Anesthesiology 2002;96:546–51. [DOI] [PubMed] [Google Scholar]
- 343.Halpern SH , Muir H , Breen TW, et al. A multicenter randomized controlled trial comparing patient-controlled epidural with intravenous analgesia for pain relief in labor. Anesth Analg 2004;99:1532–38. [DOI] [PubMed] [Google Scholar]
- 344.de Orange FA , Passini R Jr., Amorim MM, Almeida T, Barros A. Combined spinal and epidural anaesthesia and maternal intrapartum temperature during vaginal delivery: a randomized clinical trial. Br J Anaesth 2011;107:762–8. [DOI] [PubMed] [Google Scholar]
- 345.Sultan P , David AL , Fernando R , Ackland GL. Inflammation and Epidural-Related Maternal Fever: Proposed Mechanisms. Anesth Analg 2016;122:1546–53. [DOI] [PubMed] [Google Scholar]
- 346.Fusi L , Steer PJ , Maresh MJ , Beard RW. Maternal pyrexia associated with the use of epidural analgesia in labour. Lancet 1989;1:1250–2. [DOI] [PubMed] [Google Scholar]
- 347.Camann WR , Hortvet LA , Hughes N , Bader AM , Datta S. Maternal temperature regulation during extradural analgesia for labour. Br J Anaesth 1991;67:565–8. [DOI] [PubMed] [Google Scholar]
- 348.Yancey MK , Zhang J , Schwarz J , Dietrich CS 3rd, Klebanoff M. Labor epidural analgesia and intrapartum maternal hyperthermia. Obstet Gynecol 2001;98:763–70. [DOI] [PubMed] [Google Scholar]
- 349.Goetzl L , Rivers J , Zighelboim I , Wali A , Badell M , Suresh MS. Intrapartum epidural analgesia and maternal temperature regulation. Obstet Gynecol 2007;109:687–90. [DOI] [PubMed] [Google Scholar]
- 350.Zhao BS , Li B , Wang QN , Jia JX , Song XR. Time- and dose-dependent correlations between patient-controlled epidural analgesia and intrapartum maternal fever. BMC Anesthesiol 2021;21:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 351.Morton S , Kua J , Mullington CJ. Epidural analgesia, intrapartum hyperthermia, and neonatal brain injury: a systematic review and meta-analysis. Br J Anaesth 2021;126:500–15. [DOI] [PubMed] [Google Scholar]
- 352.Larue F , Benhamou D , Jullien P , Labaille T , Champagne C. Temperature monitoring during epidural anesthesia for cesarean delivery. Reg Anesth 1991;16:322–4. [PubMed] [Google Scholar]
- 353.Segal S Labor epidural analgesia and maternal fever. Anesth Analg 2010;111:1467–75. [DOI] [PubMed] [Google Scholar]
- 354.Sultan P , Segal S. Epidural-Related Maternal Fever: Still a Hot Topic, But What Are the Burning Issues? Anesth Analg 2020;130:318–20. [DOI] [PubMed] [Google Scholar]
- 355.Riley LE , Celi AC , Onderdonk AB, et al. Association of epidural-related fever and noninfectious inflammation in term labor. Obstet Gynecol 2011;117:588–95. [DOI] [PubMed] [Google Scholar]
- 356.Jansen S , Lopriore E , Naaktgeboren C, et al. Epidural-Related Fever and Maternal and Neonatal Morbidity: A Systematic Review and Meta-Analysis. Neonatology 2020;117:259–70. [DOI] [PubMed] [Google Scholar]
- 357.De Jongh RF , Bosmans EP , Puylaert MJ , Ombelet WU , Vandeput HJ , Berghmans RA. The influence of anaesthetic techniques and type of delivery on peripartum serum interleukin-6 concentrations. Acta Anaesthesiol Scand 1997;41:853–60. [DOI] [PubMed] [Google Scholar]
- 358.Goetzl L , Evans T , Rivers J , Suresh MS , Lieberman E. Elevated maternal and fetal serum interleukin-6 levels are associated with epidural fever. Am J Obstet Gynecol 2002;187:834–8. [DOI] [PubMed] [Google Scholar]
- 359.Eltzschig HK , Lieberman ES , Camann WR. Regional anesthesia and analgesia for labor and delivery. N Engl J Med 2003;348:319–32. [DOI] [PubMed] [Google Scholar]
- 360.Igarashi T , Hirabayashi Y , Shimizu R , Saitoh K , Fukuda H , Suzuki H. The fiberscopic findings of the epidural space in pregnant women. Anesthesiology 2000;92:1631–6. [DOI] [PubMed] [Google Scholar]
- 361.Hirabayashi Y , Shimizu R , Fukuda H , Saitoh K , Igarashi T. Effects of the pregnant uterus on the extradural venous plexus in the supine and lateral positions, as determined by magnetic resonance imaging. Br J Anaesth 1997;78:317–9. [DOI] [PubMed] [Google Scholar]
- 362.Wohlrab P , Boehme S , Kaun C, et al. Ropivacaine Activates Multiple Proapoptotic and Inflammatory Signaling Pathways That Might Subsume to Trigger Epidural-Related Maternal Fever. Anesth Analg 2020;130:321–31. [DOI] [PubMed] [Google Scholar]
- 363.Del Arroyo AG , Sanchez J , Patel S, et al. Role of leucocyte caspase-1 activity in epidural-related maternal fever: a single-centre, observational, mechanistic cohort study. Br J Anaesth 2019;122:92–102. [DOI] [PubMed] [Google Scholar]
- 364.Mullington CJ , Malhotra S. Hyperthermia after epidural analgesia in obstetrics. BJA education 2021;21:26–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 365.Goetzl L Maternal Fever in Labor: Etiologies, Consequences and Clinical Management. Am J Obstet Gynecol 2023. [DOI] [PubMed] [Google Scholar]
- 366.Patel. S , Ciechanowicz. S , Blumenfeld. Y, Sultan. P. Epidural-related maternal fever: incidence, pathophysiology, outcomes and management. Am J Obstet Gynecol 2023. [DOI] [PubMed] [Google Scholar]
- 367.Looff JD , Hager WD. Management of chorioamnionitis. Surg Gynecol Obstet 1984;158:161–6. [PubMed] [Google Scholar]
- 368.Conde-Agudelo A , Romero R , Jung EJ , Garcia Sánchez Á J. Management of clinical chorioamnionitis: an evidence-based approach. Am J Obstet Gynecol 2020;223:848–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 369.Czikk MJ , McCarthy FP , Murphy KE. Chorioamnionitis: from pathogenesis to treatment. Clin Microbiol Infect 2011;17:1304–11. [DOI] [PubMed] [Google Scholar]
- 370.Fishman SG , Gelber SE. Evidence for the clinical management of chorioamnionitis. Semin Fetal Neonatal Med 2012;17:46–50. [DOI] [PubMed] [Google Scholar]
- 371.Committee Opinion No. 712: Intrapartum Management of Intraamniotic Infection. Obstet Gynecol 2017;130:e95–e101. [DOI] [PubMed] [Google Scholar]
- 372.Johnson CT , Adami RR , Farzin A. Antibiotic Therapy for Chorioamnionitis to Reduce the Global Burden of Associated Disease. Front Pharmacol 2017;8:97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 373.Gilstrap LC 3rd , Leveno KJ , Cox SM , Burris JS , Mashburn M , Rosenfeld CR. Intrapartum treatment of acute chorioamnionitis: impact on neonatal sepsis. Am J Obstet Gynecol 1988;159:579–83. [DOI] [PubMed] [Google Scholar]
- 374.Sperling RS , Ramamurthy RS , Gibbs RS. A comparison of intrapartum versus immediate postpartum treatment of intra-amniotic infection. Obstet Gynecol 1987;70:861–5. [PubMed] [Google Scholar]
- 375.Rand KH , Beal SG , Rivera K , Allen B , Payton T , Lipori GP. Hourly Effect of Pretreatment With IV Antibiotics on Blood Culture Positivity Rate in Emergency Department Patients. Open forum infectious diseases 2019;6:ofz179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 376.Scalambrino S , Mangioni C , Milani R, et al. Sulbactam/ampicillin versus cefotetan in the treatment of obstetric and gynecologic infections. Suppl Int J Gynecol Obstet 1989;2:21–7. [DOI] [PubMed] [Google Scholar]
- 377.Maberry MC , Gilstrap LC 3rd , Bawdon R , Little BB , Dax J. Anaerobic coverage for intra-amnionic infection: maternal and perinatal impact. Am J Perinatol 1991;8:338–41. [DOI] [PubMed] [Google Scholar]
- 378.Locksmith GJ , Chin A , Vu T , Shattuck KE , Hankins GD. High compared with standard gentamicin dosing for chorioamnionitis: a comparison of maternal and fetal serum drug levels. Obstet Gynecol 2005;105:473–9. [DOI] [PubMed] [Google Scholar]
- 379.Lyell DJ , Pullen K , Fuh K, et al. Daily compared with 8-hour gentamicin for the treatment of intrapartum chorioamnionitis: a randomized controlled trial. Obstet Gynecol 2010;115:344–49. [DOI] [PubMed] [Google Scholar]
- 380.Patangia DV , Ryan CA , Dempsey E , Stanton C , Ross RP. Vertical transfer of antibiotics and antibiotic resistant strains across the mother/baby axis. Trends Microbiol 2022;30:47–56. [DOI] [PubMed] [Google Scholar]
- 381.Bokulich NA , Chung J , Battaglia T, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med 2016;8:343ra82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 382.Waites KB , Katz B , Schelonka RL. Mycoplasmas and ureaplasmas as neonatal pathogens. Clin Microbiol Rev 2005;18:757–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 383.Tantengco OAG , Yanagihara I. Current understanding and treatment of intra-amniotic infection with Ureaplasma spp. J Obstet Gynaecol Res 2019;45:1796–808. [DOI] [PubMed] [Google Scholar]
- 384.Beeton ML , Chalker VJ , Jones LC , Maxwell NC , Spiller OB. Antibiotic Resistance among Clinical Ureaplasma Isolates Recovered from Neonates in England and Wales between 2007 and 2013. Antimicrob Agents Chemother 2016;60:52–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 385.Koh E , Kim S , Kim I-S , Maeng K-Y , Lee S. Antimicrobial susceptibilities of Ureaplasma urealyticum and Mycoplasma hominis in pregnant women. Korean Journal of Clinical Microbiology 2009;12:159–62. [Google Scholar]
- 386.Abdel Rahman SM , Hassan RA , Sakna NA. Antimicrobial susceptibility pattern of genital Mycoplasmas among a group of pregnant women. Alexandria journal of medicine 2016;52:353–58. [Google Scholar]
- 387.Lee MY , Kim MH , Lee WI , Kang SY , Jeon YL. Prevalence and Antibiotic Susceptibility of Mycoplasma hominis and Ureaplasma urealyticum in Pregnant Women. Yonsei Med J 2016;57:1271–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 388.Grigsby PL , Novy MJ , Sadowsky DW, et al. Maternal azithromycin therapy for Ureaplasma intraamniotic infection delays preterm delivery and reduces fetal lung injury in a primate model. Am J Obstet Gynecol 2012;207:475.e1–75.e14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 389.Acosta EP , Grigsby PL , Larson KB, et al. Transplacental transfer of Azithromycin and its use for eradicating intra-amniotic ureaplasma infection in a primate model. J Infect Dis 2014;209:898–904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 390.Romero R , Hagay Z , Nores J , Sepulveda W , Mazor M. Eradication of Ureaplasma urealyticum from the amniotic fluid with transplacental antibiotic treatment. Am J Obstet Gynecol 1992;166:618–20. [DOI] [PubMed] [Google Scholar]
- 391.Hassan S , Romero R , Hendler I, et al. A sonographic short cervix as the only clinical manifestation of intra-amniotic infection. J Perinat Med 2006;34:13–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 392.Lee J , Romero R , Kim SM , Chaemsaithong P , Yoon BH. A new antibiotic regimen treats and prevents intra-amniotic inflammation/infection in patients with preterm PROM. J Matern Fetal Neonatal Med 2016;29:2727–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 393.Lee J , Romero R , Kim SM, et al. A new anti-microbial combination prolongs the latency period, reduces acute histologic chorioamnionitis as well as funisitis, and improves neonatal outcomes in preterm PROM. J Matern Fetal Neonatal Med 2016;29:707–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 394.Yoon BH , Romero R , Park JY, et al. Antibiotic administration can eradicate intra-amniotic infection or intra-amniotic inflammation in a subset of patients with preterm labor and intact membranes. Am J Obstet Gynecol 2019;221:142.e1–42.e22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 395.Oh KJ , Romero R , Park JY, et al. Evidence that antibiotic administration is effective in the treatment of a subset of patients with intra-amniotic infection/inflammation presenting with cervical insufficiency. Am J Obstet Gynecol 2019;221:140.e1–40.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 396.Yeo L , Romero R , Chaiworapongsa T, et al. Resolution of acute cervical insufficiency after antibiotics in a case with amniotic fluid sludge. J Matern Fetal Neonatal Med 2021:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 397.Kacerovsky M , Romero R , Stepan M, et al. Antibiotic administration reduces the rate of intraamniotic inflammation in preterm prelabor rupture of the membranes. Am J Obstet Gynecol 2020;223:114.e1–14.e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 398.Oh KJ , Romero R , Kim HJ, et al. The role of intraamniotic inflammation in threatened midtrimester miscarriage. Am J Obstet Gynecol 2022;227:895.e1–95.e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 399.Tita ATN , Szychowski JM , Boggess K, et al. Adjunctive Azithromycin Prophylaxis for Cesarean Delivery. N Engl J Med 2016;375:1231–41.27682034 [Google Scholar]
- 400.ACOG Practice Bulletin No. 199: Use of Prophylactic Antibiotics in Labor and Delivery. Obstet Gynecol 2018;132:e103–e19. [DOI] [PubMed] [Google Scholar]
- 401.Witt A , Sommer EM , Cichna M, et al. Placental passage of clarithromycin surpasses other macrolide antibiotics. Am J Obstet Gynecol 2003;188:816–9. [DOI] [PubMed] [Google Scholar]
- 402.De Francesco MA , Caracciolo S , Bonfanti C , Manca N. Incidence and antibiotic susceptibility of Mycoplasma hominis and Ureaplasma urealyticum isolated in Brescia, Italy, over 7 years. J Infect Chemother 2013;19:621–7. [DOI] [PubMed] [Google Scholar]
- 403.Kafetzis DA , Brater DC , Fanourgakis JE , Voyatzis J , Georgakopoulos P. Ceftriaxone distribution between maternal blood and fetal blood and tissues at parturition and between blood and milk postpartum. Antimicrob Agents Chemother 1983;23:870–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 404.Lamb HM , Ormrod D , Scott LJ , Figgitt DP. Ceftriaxone: an update of its use in the management of community-acquired and nosocomial infections. Drugs 2002;62:1041–89. [DOI] [PubMed] [Google Scholar]
- 405.Freeman CD , Klutman NE , Lamp KC. Metronidazole. A therapeutic review and update. Drugs 1997;54:679–708. [DOI] [PubMed] [Google Scholar]
- 406.Spectrum Brook I. and treatment of anaerobic infections. J Infect Chemother 2016;22:1–13. [DOI] [PubMed] [Google Scholar]
- 407.WHO recommendations for prevention and treatment of maternal peripartum infections. World Health Organization. 2016. [PubMed] [Google Scholar]
- 408.Turnquest MA , How HY , Cook CR, et al. Chorioamnionitis: is continuation of antibiotic therapy necessary after cesarean section? Am J Obstet Gynecol 1998;179:1261–6. [DOI] [PubMed] [Google Scholar]
- 409.Edwards RK , Duff P. Single additional dose postpartum therapy for women with chorioamnionitis. Obstet Gynecol 2003;102:957–61. [DOI] [PubMed] [Google Scholar]
- 410.Black LP , Hinson L , Duff P. Limited course of antibiotic treatment for chorioamnionitis. Obstet Gynecol 2012;119:1102–5. [DOI] [PubMed] [Google Scholar]
- 411.Shanks AL , Mehra S , Gross G , Colvin R , Harper LM , Tuuli MG. Treatment Utility of Postpartum Antibiotics in Chorioamnionitis Study. Am J Perinatol 2016;33:732–7. [DOI] [PubMed] [Google Scholar]
- 412.Goldberg A , Malik M. Post-partum management of chorioamnionitis: efficacy of continuation of antibiotics after delivery [13OP]. Obstet Gynecol 2017;129:5S. [Google Scholar]
- 413.Berry C , Hansen KA , McCaul JF. Abbreviated antibiotic therapy for clinical chorioamnionitis: a randomized trial. J Matern Fetal Med 1994;3:216–18. [Google Scholar]
- 414.Adashek J , Asarat T , Lagrew D , Rumney P , Adhoot D , Iriye B. The effect of antibiotics after a vaginal delivery complicated by chorioamnionitis in preventing the development of postpartum endomyometritis: a prospectively randomized double-blind placebo-controlled trial. Am J Obstet Gynecol 1998;178:S212. [Google Scholar]
- 415.Shai D , Orvieto R , Touval O, et al. Fertility and pregnancy complications following chorioamnionitis. Hum Fertil (Camb) 2021:1–4. [DOI] [PubMed] [Google Scholar]
- 416.Morishima HO , Glaser B , Niemann WH , James LS. Increased uterine activity and fetal deterioration during maternal hyperthermia. Am J Obstet Gynecol 1975;121:531–8. [DOI] [PubMed] [Google Scholar]
- 417.Cefalo RC , Hellegers AE. The effects of maternal hyperthermia on maternal and fetal cardiovascular and respiratory function. Am J Obstet Gynecol 1978;131:687–94. [DOI] [PubMed] [Google Scholar]
- 418.Lieberman E , Lang J , Richardson DK , Frigoletto FD , Heffner LJ , Cohen A. Intrapartum maternal fever and neonatal outcome. Pediatrics 2000;105:8–13. [DOI] [PubMed] [Google Scholar]
- 419.Greenwell EA , Wyshak G , Ringer SA , Johnson LC , Rivkin MJ , Lieberman E. Intrapartum temperature elevation, epidural use, and adverse outcome in term infants. Pediatrics 2012;129:e447–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 420.Dior UP , Kogan L , Eventov-Friedman S, et al. Very High Intrapartum Fever in Term Pregnancies and Adverse Obstetric and Neonatal Outcomes. Neonatology 2016;109:62–8. [DOI] [PubMed] [Google Scholar]
- 421.Hochler H , Lipschuetz M , Guedalia J, et al. The impact of peak and duration of maternal intrapartum fever on perinatal outcomes. Am J Obstet Gynecol MFM 2021;3:100390. [DOI] [PubMed] [Google Scholar]
- 422.Hensel D , Zhang F , Carter EB, et al. Severity of intrapartum fever and neonatal outcomes. American journal of obstetrics and gynecology 2022;227:513.e1–13.e8. [DOI] [PubMed] [Google Scholar]
- 423.Grether JK , Nelson KB. Maternal infection and cerebral palsy in infants of normal birth weight. JAMA 1997;278:207–11. [PubMed] [Google Scholar]
- 424.Lieberman E , Eichenwald E , Mathur G , Richardson D , Heffner L , Cohen A. Intrapartum fever and unexplained seizures in term infants. Pediatrics 2000;106:983–8. [DOI] [PubMed] [Google Scholar]
- 425.Impey L , Greenwood C , MacQuillan K , Reynolds M , Sheil O. Fever in labour and neonatal encephalopathy: a prospective cohort study. BJOG 2001;108:594–7. [DOI] [PubMed] [Google Scholar]
- 426.Impey LW , Greenwood CE , Black RS , Yeh PS , Sheil O , Doyle P. The relationship between intrapartum maternal fever and neonatal acidosis as risk factors for neonatal encephalopathy. Am J Obstet Gynecol 2008;198:49.e1–6. [DOI] [PubMed] [Google Scholar]
- 427.Petrova A , Demissie K , Rhoads GG , Smulian JC , Marcella S , Ananth CV. Association of maternal fever during labor with neonatal and infant morbidity and mortality. Obstet Gynecol 2001;98:20–7. [DOI] [PubMed] [Google Scholar]
- 428.Nitsche JF , Patil AS , Langman LJ, et al. Transplacental Passage of Acetaminophen in Term Pregnancy. Am J Perinatol 2017;34:541–43. [DOI] [PubMed] [Google Scholar]
- 429.Kirshon B , Moise KJ Jr., Wasserstrum N. Effect of acetaminophen on fetal acid-base balance in chorioamnionitis. J Reprod Med 1989;34:955–9. [PubMed] [Google Scholar]
- 430.Reilly DR , Oppenheimer LW. Fever in term labour. J Obstet Gynaecol Can 2005;27:218–23. [DOI] [PubMed] [Google Scholar]
- 431.Lavesson T , Åkerman F , Källén K , Olofsson P. Effects on fetal and maternal temperatures of paracetamol administration during labour: a case-control study. Eur J Obstet Gynecol Reprod Biol 2013;168:138–44. [DOI] [PubMed] [Google Scholar]
- 432.Morrison CL , Glenn LL. Statistical significance of paracetamol administration in fetal and maternal body temperatures. Eur J Obstet Gynecol Reprod Biol 2013;171:191. [DOI] [PubMed] [Google Scholar]
- 433.Burgess APH , Katz JE , Moretti M , Lakhi N. Risk Factors for Intrapartum Fever in Term Gestations and Associated Maternal and Neonatal Sequelae. Gynecol Obstet Invest 2017;82:508–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 434.Holmér Pettersson P , Owall A , Jakobsson J. Early bioavailability of paracetamol after oral or intravenous administration. Acta Anaesthesiol Scand 2004;48:867–70. [DOI] [PubMed] [Google Scholar]
- 435.Ope-Adenuga S , Burgess APH , Reilly JG , Moretti M , Lakhi N. Intravenous acetaminophen for the treatment of intrapartum fever and resolution of fetal tachycardia: a novel use for an old medication. Clin Exp Obstet Gynecol 2017;44:166–68. [PubMed] [Google Scholar]
- 436.Reduction of intrapartum fever with intravenous acetaminophen (RIFIVA). Available at: https://clinicaltrials-gov.proxy.lib.wayne.edu/ct2/show/NCT02625454. Accessed June 25, 2020.