Your activity: 46 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Preterm birth: Risk factors, interventions for risk reduction, and maternal prognosis

Preterm birth: Risk factors, interventions for risk reduction, and maternal prognosis
Authors:
Julian N Robinson, MD
Errol R Norwitz, MD, PhD, MBA
Section Editor:
Charles J Lockwood, MD, MHCM
Deputy Editor:
Vanessa A Barss, MD, FACOG
Literature review current through: Dec 2022. | This topic last updated: Jul 26, 2022.

INTRODUCTION — Preterm birth (PTB) refers to a birth that occurs from 20+0 to 36+6 weeks of gestation (table 1). Seventy to 80 percent of PTBs are spontaneous, resulting from preterm labor (40 to 50 percent) or preterm prelabor rupture of membranes (20 to 30 percent); rarely, cervical insufficiency results in spontaneous PTB (sPTB). The remaining 20 to 30 percent of PTBs are initiated by the health care provider because of maternal or fetal issues that jeopardize the health of the mother or fetus (eg, preeclampsia, placenta previa, abruptio placentae, fetal growth restriction, multiple gestation). Complications of pregnancy can lead to both sPTB and provider-initiated PTBs.

PTB is relatively common, occurring in 5 to 18 percent of births worldwide (see "Preterm birth: Definitions of prematurity, epidemiology, and risk factors for infant mortality", section on 'Incidence of prematurity'). There are many risk factors for PTB (table 2) and many pathways from these risk factors to the terminal cascade of events resulting in labor. Preterm labor likely occurs when local uterine factors prematurely stimulate this cascade or suppressive factors that inhibit the cascade and maintain uterine quiescence are withdrawn prematurely. (See "Spontaneous preterm birth: Pathogenesis".)

Ideally, identification of modifiable and nonmodifiable risk factors for PTB before conception or early in pregnancy will lead to interventions that help prevent this complication. However, few interventions have been proven to prolong pregnancy in women at risk. This goal has been elusive for several reasons: two-thirds of PTBs occur among women with no risk factors, causality has been difficult to prove (eg, a cofactor may be required thus complicating the chain of causality), and no adequate animal model exists for study of sPTB.

Risk factors for PTB and potential interventions to mitigate risk, when possible, will be reviewed here. Pathogenesis of PTB and diagnosis and treatment of preterm labor are discussed separately:

(See "Spontaneous preterm birth: Pathogenesis".)

(See "Inhibition of acute preterm labor".)

(See "Preterm labor: Clinical findings, diagnostic evaluation, and initial treatment".)

REPRODUCTIVE HISTORY

History of spontaneous preterm birth — A history of sPTB is the major risk factor for recurrence, and recurrences often occur at the same gestational age [1-3]. Women at highest risk are those with:

No term pregnancy between the previous sPTB and the current pregnancy

A history of multiple sPTBs

In large series, the frequency of recurrent sPTB was 15 to 30 percent after one sPTB and even higher after two sPTBs [4-8]. The odds of recurrent sPTB in three high-income countries (New Zealand, Sweden, Slovenia) ranged from 4.5 to 7.1 in one study [9]. Term births decrease the risk of sPTB in subsequent pregnancies (table 3 and table 4).

The risk of recurrent early sPTB is of particular concern given its high morbidity and mortality. In a large prospective series, approximately 5 percent of women who had an early sPTB at 23 to 27 weeks in their prior pregnancy delivered at <28 weeks in their subsequent pregnancy [4]. By comparison, if there was no previous history of sPTB, then the risk of sPTB <28 weeks was only 0.2 percent.

Other characteristics of the prior sPTB may also predict recurrence risk. In a small retrospective cohort study, women who presented with painless advanced cervical dilation were significantly more likely to have recurrent sPTB than women with a history of preterm prelabor rupture of membranes or preterm labor (55 versus 27 versus 32 percent, respectively) [10]. The increased risk remained after adjustment for gestational age of the last PTB.

Women who were born preterm are at modestly increased risk of having a sPTB compared with women who were born at term. (See 'Genetic factors' below.)

A prior sPTB of twins is associated with an increased risk of sPTB in a subsequent singleton pregnancy. In a meta-analysis assessing the risk of PTB in singleton pregnancies after a previous twin birth, compared with a previous twin term birth, the odds of subsequent preterm singleton birth (all causes) were 2.13 (95% CI 1.21-3.74) if the previous twins were born at 34+0 to 36+6 weeks of gestation, 5.18 (95% CI 2.78-9.64) if they were born at 30+0 to 33+6 weeks, and 9.73 (95% CI 4.99-18.98) if they were born at <30+0 weeks [11]. The odds ratios (ORs) were similar (2.71, 5.74, and 6.54, respectively) when only twin sPTBs were considered.

The overall risk of sPTB in twin pregnancy is significantly higher in multiparous women whose previous singleton pregnancy was a sPTB: 67.3 percent versus 20.9 percent if the previous singleton delivery was at term (OR 7.8, 95% CI 5.5-11.2) [12].

Intervention

Progesterone supplementation appears to reduce the risk of PTB by approximately 20 percent in women with a singleton pregnancy and a history of sPTB or a short cervix. A review of evidence and treatment approaches, including choice of progesterone formulation, is available separately. (See "Progesterone supplementation to reduce the risk of spontaneous preterm labor and birth".)

In women with a history of PTB in whom the diagnosis of cervical insufficiency is uncertain, sonographic evidence of a short cervix in the midtrimester can identify those who may benefit from placement of a cerclage (ultrasound-indicated cerclage). Women with multiple second-trimester pregnancy losses/PTBs associated with painless cervical dilation may benefit from early placement of a cerclage based on this history alone (history-indicated cerclage) (table 5). A review of screening and treatment approaches and supporting evidence is available separately.

(See "Short cervix before 24 weeks: Screening and management in singleton pregnancies", section on 'Parous patients with a prior spontaneous preterm singleton birth'.)

(See "Cervical insufficiency".)

A short interpregnancy interval has been associated with an increased risk of PTB. The March of Dimes encourages women to space pregnancies at least 18 months apart. (See 'Short interpregnancy interval' below.)

Ineffective and unproven interventions

We do not prescribe low-dose aspirin for prevention of sPTB. It has been hypothesized that low-dose aspirin may reduce the risk of sPTB by inhibition of the inflammatory and uteroplacental ischemia pathways leading to this outcome (figure 1). Secondary outcomes from prevention of preeclampsia trials provided the initial support for this hypothesis [13,14]. (See "Spontaneous preterm birth: Pathogenesis".)

Subsequently, the ASPIRIN trial evaluated the use of low-dose aspirin for prevention of PTB as the primary outcome [15]. Nearly 12,000 nulliparous women with singleton pregnancies in six low- and middle-income countries were randomly assigned to receive 81 mg aspirin or placebo beginning at 6+0 to 13+6 weeks of gestation and continuing until 36+6 weeks. Aspirin reduced PTB <37 weeks (11.6 versus 13.1 percent, RR 0.89, 95% CI 0.81-0.98) and <34 weeks (3.3 versus 4.0 percent, RR 0.75, 95% CI 0.61-0.93) and also reduced fetal loss and perinatal mortality; other maternal or neonatal outcomes were similar between groups. These benefits occurred, in part, because of a 60 percent reduction in women who delivered before 34 weeks with a hypertensive disorder of pregnancy (0.1 versus 0.4 percent); the incidence of hypertensive disorders in late pregnancy was not reduced. Limitations of the trial included lack of data on the effect of aspirin in women who are multiparous, carrying a multiple gestation, living in a high-income country, or residing in an area where low-dose aspirin is routinely recommended for women at moderate or high risk for developing preeclampsia. The trial also did not distinguish between sPTB and indicated PTB.

The APRIL trial was the first trial to evaluate aspirin (80 mg) versus placebo in women (n = 387) with singleton pregnancies plus a history of spontaneous preterm birth [16]. The intervention was initiated between 8 and 16 weeks of gestation and continued until 36 weeks or delivery. Aspirin prophylaxis did not result in clear reduction in total PTB <37 weeks (21.2 versus 25.4 percent in the placebo group; RR 0.83, 95% CI 0.58-1.20), including in patients with ≥80 percent medication adherence (19.2 versus 24.8 percent; RR 0.77, 95% CI 0.48-1.25). The two subgroups of spontaneous and indicated preterm births <37 weeks of gestation also did not significantly differ between participants allocated to aspirin versus placebo. A major limitation of the trial was that the PTB rate in both groups was less than anticipated, possibly because nearly two-thirds of participants were prescribed progesterone supplementation and approximately 10 percent underwent cervical cerclage. The findings suggest that a trial would need to be 10-fold larger than APRIL to determine whether aspirin can prevent recurrent PTB [17].

We agree with the American College of Obstetricians and Gynecologists' guidelines, which were published before the ASPIRIN and APRIL trials, that state that low-dose aspirin should not be used in an attempt to prevent sPTB but should be considered for women with risk factors for preeclampsia [18]. Although it is likely that the bulk of the putative beneficial effects of low-dose aspirin on PTB is via the prevention of indicated PTB due to preeclampsia, the possibility of an effect on sPTB as well is plausible and should be studied further.

Although an increase in uterine activity is a prerequisite for labor, randomized trials and a meta-analysis have shown that self-measurement of the frequency of uterine contractions by self-palpation/detection of signs of labor or through use of a home uterine activity monitor does not lead to a reduction in the rate of PTB [19,20]. Moreover, such an approach increases the frequency of unscheduled antenatal visits. The American College of Obstetricians and Gynecologists recommends not using home uterine activity monitoring as a strategy for prediction or prevention of PTB [21].

Prophylactic tocolytic therapy for prevention of PTB in high-risk asymptomatic women is not effective, although few randomized trials have been conducted [22,23]. (See "Management of pregnancy after resolution of an episode of acute idiopathic preterm labor".)

History of indicated preterm birth — A large retrospective cohort study assessed the risk of recurrent PTB by the type of PTB in the previous pregnancy [7]. The rate of recurrent PTB was 23 percent for women with a prior indicated PTB and 31.6 percent for women with a prior sPTB. Women with a prior indicated PTB were at particularly high risk for recurrent indicated PTB (relative risk [RR] 9.10, 95% CI 4.68-17.71) but also at increased risk of sPTB (RR 2.70, 95% CI 2.00-3.65). Women with a prior sPTB were at five- to sixfold increased risk for recurrent sPTB, but also appeared to be at slightly increased risk for indicated PTB (RR 1.61, 95% CI 0.98-2.67).

Intervention

Interventions to reduce the risk for recurrent indicated PTB depend on the indication for PTB. For example, administration of low-dose aspirin to women with a history of early delivery because of preeclampsia with severe features can reduce their risk for recurrent preeclampsia and PTB. (See "Preeclampsia: Prevention", section on 'Low-dose aspirin'.)

History of abortion — In a systematic review of pregnancy outcome after uterine evacuation including over one million women (31 studies involving termination of pregnancy, five studies involving spontaneous abortion), women with a history of surgical uterine evacuation had a small but statistically significant increase in risk for PTB in a subsequent pregnancy compared with controls [24]. Women who underwent medical termination of pregnancy had a similar future risk of PTB as women with no history of pregnancy termination.

Although surgical uterine evacuation appeared to be a risk factor for subsequent PTB, observational studies are flawed because they are subject to recall bias and inadequate adjustment of many of the other risk factors for adverse pregnancy outcome. (See "Overview of pregnancy termination", section on 'Future pregnancies'.)

GENETIC FACTORS — Genetic polymorphisms appear to contribute to length of gestation and a woman's likelihood of sPTB. In a genomewide association study of a large cohort of women of European ancestry, maternal variants at the EBF1, EEFSEC, AGTR2, WNT4, ADCY5, and RAP2C loci were associated with gestational duration and maternal variants at the EBF1, EEFSEC, and AGTR2 loci were associated with PTB; however, birth outcomes were self-reported [25]. Although PTB susceptibility genes have been identified, epigenetic and gene-environmental factors probably play a more important role in PTB than the maternal genotype.

PTBs are more prevalent in some family pedigrees and racial groups, in women who were born preterm themselves, and in women with a first-degree female relative who had a PTB [26,27]. In addition, concordance for timing of parturition is higher in women who are monozygotic twins than in those who are dizygotic twins [3,28-38].

The paternal genotype does not have a significant effect on PTB. (See 'Paternal risk factors' below.)

RACE AND ETHNICITY — PTB rates are higher for non-Hispanic Black and Indigenous women than for White, Asian, or Hispanic women in the United States. In a meta-analysis of eight English-language studies including over 26 million singleton births, the odds of PTB were lowest in couples in which both parents were White and progressively increased with Black parentage: White mother/White father (odds ratio [OR] 1.0), White mother/Black father (OR 1.17), Black mother/White father (OR 1.37), Black mother/Black father (OR 1.78) [39]. This may be related to both biologic and environmental factors (social and economic disadvantage [eg, lower educational attainment, residence in a disadvantaged area, structural racism, lack of access to prenatal care], which can result in chronic stress) [40].

A discrepancy in the risk of recurrent PTB between Black and White populations has also been observed. In Black and White women whose first delivery was at 20 to 31 weeks of gestation, the frequency of a second delivery at the same gestational age range was 13.4 and 8.2 percent, respectively, in one study [4,41]. For the gestational age range 32 to 36 weeks, the frequency of a second delivery at the same gestational age was 3.8 and 1.9 percent, respectively.

Although differences in environmental risk factors account for some of the increased risk in PTB, biologic factors such as polymorphisms in genes for regulation of innate immunity also appear to play a role [42-45]. In addition, women's race/ethnicity seems to influence their microbiome and the impact of vaginal bacteria on PTB [46-49]. One mechanism may involve an enhanced proinflammatory response to normal or altered vaginal microflora, leading to preterm labor or preterm prelabor rupture of membranes (PPROM) [50]. Alternatively, immune hyporesponsiveness may create a permissive environment for ascending infection and its sequelae (premature labor, PPROM) [51,52]. (See "An overview of the innate immune system".)

AGE — The rate of PTB is higher at the extremes of maternal age [53-56]. Physiologic immaturity and socioeconomic factors may increase risk for adolescent mothers; a higher prevalence of preexisting chronic disease and obesity may increase risk for older mothers. Both groups have high rates of unintended pregnancy; prevention of these pregnancies may reduce PTB [57].

CERVICAL SURGERY AND TRANSCERVICAL PROCEDURES — Cold knife conization and loop electrosurgical excision procedures for treatment of cervical intraepithelial neoplasia have been associated with increased risks for late miscarriage and PTB. Possible mechanisms include loss of tensile strength from loss of cervical stroma, increased susceptibility for infection from loss of cervical glands, and loss of cervical plasticity from cervical scarring. (See "Reproductive effects of cervical excisional and ablative procedures".)

In a meta-analysis of observational studies, women with a history of dilation and curettage (D&C), were at higher risk for PTB compared with those with no such history: PTB <37 weeks, OR 1.29 (95% CI 1.17-1.42); PTB <32 weeks, OR 1.69 (95% CI 1.20-2.38); PTB <28 weeks, OR 1.68 (95% CI 1.47-1.92) [58]. The risk remained increased when the control group was limited to women with a medically managed miscarriage or induced abortion and when only sPTBs were considered, and was highest for women with a history of multiple D&Cs.

Intervention

Women undergoing treatment of cervical intraepithelial neoplasia should have the procedure that best diagnoses or prevents cervical cancer and also incurs the lowest risk of reproductive effects.

Although women who have undergone cervical surgery may develop cervical insufficiency, the pregnancy course and outcome need to be evaluated before making this diagnosis. We perform a single transvaginal ultrasound measurement of cervical length measurement at 18 to 24 weeks in all women and treat those with a short cervix (≤25 mm) and no prior PTB with vaginal progesterone supplementation. (See "Cervical insufficiency", section on 'Patients with no prior second-trimester pregnancy loss or extremely preterm birth, but risk factors for cervical insufficiency'.)

UTERINE MALFORMATIONS

Congenital — In women with congenital uterine malformations, the magnitude of risk for PTB depends upon the specific abnormality [59-61]. (See "Congenital uterine anomalies: Clinical manifestations and diagnosis".)

Intervention

Surgical correction of the abnormality may reduce the risk for PTB. (See "Congenital uterine anomalies: Surgical repair".)

Acquired — Women with fibroids may be at slightly increased risk for pregnancy loss and PTB. A large fibroid (ie, ≥5 to 6 cm) or multiple fibroids appear to be the most important risk factors for PTB; a submucosal location is the most important risk factor for pregnancy loss. (See "Uterine fibroids (leiomyomas): Issues in pregnancy", section on 'Preterm labor and birth'.)

Intervention

Myomectomy before pregnancy may be indicated in women with pregnancy loss or early PTB. Every effort should be made to avoid surgical removal of fibroids during pregnancy because of the risk for significant morbidity (hemorrhage). (See "Uterine fibroids (leiomyomas): Issues in pregnancy".)

CHRONIC MEDICAL DISORDERS — Chronic maternal medical disorders can be associated with maternal or fetal complications necessitating medically indicated PTB as well as an increased risk for sPTB. Examples include women with hypertension, renal insufficiency, type 1 diabetes mellitus, some autoimmune diseases, and nonphysiologic anemia.

Both depression and exposure to selective serotonin reuptake inhibitors have been associated with an increased risk of PTB. (See "Antenatal depression: Pregnancy and neonatal outcomes", section on 'Preterm birth' and "Antenatal use of antidepressants and the potential risk of teratogenicity and adverse pregnancy outcomes: Selective serotonin reuptake inhibitors", section on 'Preterm birth'.)

Intervention

Preconception identification and optimization of chronic medical diseases, such as diabetes and hypertension, can improve maternal health and pregnancy outcome. (See "The preconception office visit".)

Optimization of depression symptoms is also desirable. (See "Unipolar major depression in pregnant women: General principles of treatment".)

PREVIOUS INFANT WITH SUDDEN INFANT DEATH SYNDROME — A history of delivery of an infant who subsequently died from sudden infant death syndrome appears to be a risk factor for PTB in the following pregnancy [62]. (See "Sudden infant death syndrome: Risk factors and risk reduction strategies".)

ASSISTED REPRODUCTION — Pregnancies conceived by assisted reproduction are at higher risk for sPTB, even in the absence of multifetal gestation. The increased risk may be related to baseline maternal factors related to subfertility and/or factors related to assisted reproduction procedures. (See "Assisted reproductive technology: Pregnancy and maternal outcomes", section on 'Low birth weight'.)

MULTIFETAL GESTATION — Multifetal gestation accounts for only 2 to 3 percent of all births but 17 percent of births before 37 weeks of gestation and 23 percent of births before 32 weeks. The widespread availability of assisted reproductive technology resulted in a large increase in the incidence of multiple gestation; this increase, in turn, led to an increase in sPTB and indicated PTB [63]. Subsequently, policies to promote the transfer of only a single embryo into the uterus and other interventions have reduced the frequency of multifetal gestations.

The mechanism for sPTB in multifetal gestations, and particularly higher-order multifetal gestations, may be related to sequelae of increased uterine distension (see "Spontaneous preterm birth: Pathogenesis", section on '#4 Pathologic uterine distention'). The endocrine environment produced by superovulation or the multiple pregnancy may also play a role. As an example, multifetal gestations produce increased amounts of estrogen, progesterone, and sex steroids compared with singleton pregnancies [64,65]. Increased steroid production may be a factor in initiation of labor (see "Physiology of parturition at term"). Higher circulating levels of relaxin associated with super-ovulation may cause cervical insufficiency with subsequent sPTB [66].

Intervention

Prevention and reduction of multifetal gestations, particularly high-order multifetal gestations, appear to improve neonatal outcome. (See "Strategies to control the rate of high order multiple gestation" and "Multifetal pregnancy reduction and selective termination".)

In unselected twin pregnancies, progesterone supplementation, use of a pessary, cerclage, and bed rest/reduction of physical activity do not prolong gestation. In women with a twin pregnancy and a prior singleton sPTB or a short cervix, the use of supplemental progesterone or a pessary is controversial and reviewed separately. (See "Twin pregnancy: Management of pregnancy complications", section on 'Overview' and "Twin pregnancy: Management of pregnancy complications", section on 'Approach to patients with a short cervix' and "Progesterone supplementation to reduce the risk of spontaneous preterm labor and birth", section on 'Twin pregnancy'.)

VAGINAL BLEEDING IN EARLY PREGNANCY — Early pregnancy bleeding is often due to decidual hemorrhage and associated with an increased risk for both subsequent sPTB and indicated PTB. In a large study based on registry data, pregnancies with first-trimester bleeding were at increased risk for preterm prelabor rupture of membranes (PPROM; odds ratio [OR] 1.18, 95% CI 1.01-1.37), placental abruption (OR 1.48, 95% CI 1.30-1.68), and severe preeclampsia (OR 1.25, 95% CI 1.09-1.43) [67]. In this and other studies, the association was stronger for PTB before 34 weeks than late PTB [67,68]. Women with persistent vaginal bleeding and bleeding in the second trimester are at higher risk of these complications than those with an isolated first-trimester event.

Decidual hemorrhage results in release of tissue factor, which can trigger local thrombin formation. Decidual thrombin production has been associated with increased expression of soluble fms-like tyrosine kinase-1 (sFlt-1) and monocyte-recruiting chemokines, factors also associated with subsequent indicated PTB due to preeclampsia, abruption, or fetal growth restriction as well as subsequent sPTB [69]. Later in pregnancy, decidual cell-derived thrombin can inhibit decidual cell progesterone receptor expression, possibly resulting in PTB related to abruption or PPROM [70-72]. (See "Spontaneous preterm birth: Pathogenesis", section on '#3 Decidual hemorrhage'.)

Intervention

Women with a history of PTB may be treated with progesterone to prevent recurrence in a subsequent pregnancy. Those who have vaginal bleeding/abruption in the subsequent pregnancy still appear to respond to hydroxyprogesterone caproate prophylaxis in that pregnancy [73]. (See "Progesterone supplementation to reduce the risk of spontaneous preterm labor and birth".)

There are no effective interventions for improving the prognosis of patients who are experiencing early pregnancy bleeding. Bed rest is not beneficial. Patients often avoid sexual intercourse when they are bleeding, but there is no evidence of harm from sexual activity [74,75].

SHORT CERVIX — There is an inverse relationship between cervical length measured by transvaginal ultrasound at 16 to 28 weeks of gestation and gestational age at delivery (table 6). A high Bishop score on digital examination is also associated with increased odds of PTB [76]. (See "Short cervix before 24 weeks: Screening and management in singleton pregnancies".)

Intervention

For women with singleton pregnancies and no history of prior PTB, we suggest screening for a short cervix (≤25 mm) with a single examination at 18 to 24 weeks, which can be performed in conjunction with the fetal anatomic survey ultrasound examination. Vaginal progesterone supplementation in this population reduces the risk for PTB. A review of evidence and treatment approaches are discussed in detail separately. (See "Progesterone supplementation to reduce the risk of spontaneous preterm labor and birth", section on 'Progesterone preparations' and "Progesterone supplementation to reduce the risk of spontaneous preterm labor and birth", section on 'Candidates for progesterone supplementation'.)

For women with singleton pregnancies and a history of prior PTB, we suggest progesterone supplementation (vaginal or intramuscular) and serial measurements of cervical length, in agreement with most guidelines [21,77]. Cerclage may be indicated for those who develop a short cervix despite progesterone supplementation. We do not prescribe pessaries for women with a short cervix as the body of evidence does not support using a pessary to prolong gestation or improve neonatal outcome. (See "Cervical insufficiency", section on 'Ultrasound-based cervical insufficiency' and "Cervical insufficiency", section on 'Pessary'.)

Bed rest is not helpful — Bed rest improves uteroplacental blood flow and can lead to a slight increase in birth weight, but there is no evidence that it decreases the incidence of PTB [78-80], even in women with a short cervix [81,82]. Although underpowered, the only randomized trial attempting to determine whether hospitalization of women with arrested preterm labor improved outcome found hospitalized women had a similar rate of PTB as those who were discharged home and thus presumably more active [83]. In women with a short cervix, observational studies have reported a higher rate of PTB in those with reduced physical activity [81,84].

Bed rest also has potential harms: it appears to increase the risk of thromboembolic events, has clear negative psychosocial effects, and leads to deconditioning [85-88].

DILATED CERVIX — Cervical dilation ≥1 cm before 24 weeks of gestation is associated with an increased risk of PTB and increasing cervical dilation is associated with increasing risk of PTB.

Intervention

In women with cervical insufficiency based on physical examination, cerclage placement has been associated with a significant increase in prolongation of pregnancy and neonatal survival compared with expectant management. (See "Cervical insufficiency", section on 'Physical examination-based cervical insufficiency'.)

INFECTION — Multiple unrelated studies from varied disciplines (epidemiology, histopathology, microbiology, biochemistry, and maternal-fetal medicine) have reported an association between infection/inflammation and PTB, likely mediated by prostaglandins. The most consistent of these observations were reported by placental pathologists who have described histologic evidence of chorioamnionitis in the placentas of 20 to 75 percent of PTBs and positive membrane cultures in 30 to 60 percent of such patients [89-92]. In the Collaborative Perinatal Project, chorioamnionitis was detected in 6 percent of 43,940 deliveries evaluated, and the rate decreased with increasing gestational age: 15 percent at 28 to 32 weeks, 8 percent at 33 to 36 weeks, and 5 percent after 36 weeks of gestation [93].

Asymptomatic bacteriuria — It is unclear whether asymptomatic bacteriuria is an independent risk factor for PTB [94]. In one of the largest studies, the Cardiff Birth Survey, which prospectively studied over 25,000 births between 1970 and 1979, asymptomatic bacteriuria was not associated with a statistically significant increase in the overall rate of PTB (adjusted odds ratio [aOR] 1.21, 95% CI 0.96-1.53) [95] or sPTB (aOR 1.07, 95% CI 0.78-1.46) [96] when the data were adjusted for demographic and social factors.

Intervention

A first-trimester urine culture should be performed on all pregnant women [97,98], and regular antenatal screening is recommended for women at high risk for asymptomatic bacteriuria (eg, women with sickle cell trait, recurrent urinary tract infections, diabetes mellitus, underlying renal disease). Reliance on symptoms to prompt screening is inadequate because symptoms such as frequency and nocturia are often attributed to the pregnant state.

Pregnant women with asymptomatic bacteriuria should be treated with antibiotics to reduce their risk of developing pyelonephritis and possibly to reduce the risk of PTB. In a meta-analysis (14 randomized trials), treatment of asymptomatic bacteriuria clearly and substantially decreased the frequency of asymptomatic bacteriuria (relative risk [RR] 0.25, 95% CI 0.14-0.48), pyelonephritis (RR 0.23, 95% CI 0.13-0.41) [99], and low birth weight (RR 0.66, 95% CI 0.49-0.89), but a difference in PTB was not established. (See "Urinary tract infections and asymptomatic bacteriuria in pregnancy", section on 'Asymptomatic bacteriuria'.)

Periodontal disease — Periodontal disease is common in adults. Two systematic reviews have reported an association between periodontal disease and adverse pregnancy outcome, such as sPTB, but did not provide conclusive evidence that pregnancy complications, including sPTB, result from periodontal disease [100,101]. The included studies had different designs and used different criteria to diagnose periodontal disease and to define adverse outcome. Moreover, they generally did not adequately adjust for confounders or have adequate sample size to detect significant differences in pregnancy outcome. Oral bacteria that have been associated with both periodontal disease and PTB include Tannerella forsythia, Porphyromonas gingivalis, Actinobacillus actinomycetemcomitans, Treponema denticola, and Fusobacterium nucleatum [102-104].

Several hypotheses have been proposed to explain the association between periodontal disease and sPTB [105-108]. Periodontal flora may seed the fetoplacental unit and cause local inflammation, or inflammatory mediators of periodontal origin may cause systemic inflammation. An alternative, but equally reasonable, explanation is that periodontal disease is a marker of individuals who have a genetic predisposition towards an exaggerated local or systemic inflammatory response to a given stimulus (eg, bacteria), which leads to two separate adverse clinical events: periodontal disease and sPTB. Such individuals may also hyper-respond to vaginal bacteria with enhanced production of cytokines that lead to preterm labor or rupture of membranes. Thus, periodontal disease and preterm labor can be epidemiologically linked but not causally related.

Intervention

Although treatment of periodontal disease contributes to oral health, there is no strong evidence that treatment of periodontal disease improves pregnancy outcome. A joint consensus report of the European Federation of Periodontology and the American Academy of Periodontology in 2013 concluded that, although periodontal therapy is safe and leads to improved periodontal health in pregnant women, periodontal therapy does not reduce overall rates of PTB and low birth weight [109]. This conclusion was based on meta-analyses limited to higher-quality randomized trials that demonstrated no significant effect of nonsurgical periodontal treatment on rates of PTB or low birth weight [110-113]. Subsequent meta-analyses have reported similar findings [114].

Several hypotheses have been proposed in an attempt to explain why periodontal treatment has not been shown to reduce risk for adverse pregnancy outcomes. Possibilities include [115]:

Lack of causality. Periodontitis may not be a direct or indirect cause of PTB or low birth weight.

Shared risk factors may mitigate the effect of treatment. Some risk factors for both periodontitis and poor pregnancy outcome (eg, smoking) are not affected by periodontal treatment. Furthermore, PTB is likely the end result of a variety of environmental, behavioral, social, biological, and possibly genetic factors so periodontal treatment alone is unlikely to have a major impact on reducing risk.

Underpowered trials. Very large trials would be required to detect significant reductions in very or extreme PTB rates since these are much less common than late PTBs.

Lack of a consistent definition of periodontal disease. This has led to inclusion of women with mild disease whose pregnancies may not benefit from treatment.

Treatment of periodontal disease in the trials was inadequate to affect pregnancy outcome. To demonstrate a beneficial effect, treatment may have to start before pregnancy or very early in pregnancy, continue longer, or be more intense. If PTB is related to changes in the genital tract microbiome induced by changes in the oral microbiome, local treatment of oral inflammation may not reverse genital tract changes.

Genital tract infection/colonization — Multiple studies have reported an association between preterm labor/delivery and various genital tract infections/colonization (table 7), including group B streptococci (GBS) [116], Chlamydia trachomatis [117-120], bacterial vaginosis (BV) [121-123], Neisseria gonorrhea [124], syphilis [125], Trichomonas vaginalis [126], Ureaplasma species [127], unencapsulated Haemophilus influenzae [128], and possibly persistent human papillomavirus 16/18 [129,130]. Mycoplasma hominis in cervicovaginal fluid does not appear to be a risk factor for sPTB [131-134].

In a population-based retrospective cohort study including over 14 million singleton live births in the United States, the PTB rate associated with chlamydia, gonorrhea, and syphilis infection was 9.9, 12.2, and 13.3 percent, respectively [135]. Compared with pregnancies without these infections, the odds of PTB were: chlamydia OR 1.03 (95% CI 1.02-1.04), gonorrhea OR 1.11 (95% CI 1.08-1.15), syphilis OR 1.17 (95% CI 1.11-1.22), and any of these sexually transmitted infections OR 1.06 (95% CI 1.05-1.07), after adjustment for sociodemographic, medical, and/or health factors. Limitations of the study included lack of information about gestational age at diagnosis, treatment, type of PTB (spontaneous, indicated, or after PPROM), and misclassification bias.

A positive culture correlates with the presence of histologic chorioamnionitis; however, causal relationships for most of these infections and PTB have not been proven and are controversial [116,136,137].

Candida species colonization is not a risk factor for PTB [138].

Vaginal microbiome — Emerging research has found that pregnancy alters the vaginal microbiome profile to be more hospitable to Lactobacillus and less favorable to G. vaginalis and other taxa associated with BV, with the exception of BV-associated bacterium 1 (BVAB1), which tends to remain stable [139]. In addition, there is increasing evidence that some vaginal microbiomes are associated with an increased risk for sPTB, and the prevalence of these microbiomes varies across populations [140-142]. As an example, carriage of BVAB1 is positively associated with PTB and more prevalent in pregnant women of African ancestry, who are known to have an increased risk of PTB compared with those of European ancestry, whereas carriage of L. crispatus is protective against PTB and more prevalent in pregnant women of European ancestry [140]. Whether interventions designed to favorably alter the vaginal microbiome will abrogate the risk of PTB is not known.

Intervention

Role of routine screening – Some trials have reported a reduction in PTB with routine screening and treatment for infection in the early second trimester [143], while others have reported no benefit [144,145]. Discordant findings may be due to confounding by recolonization or reinfection after therapy, intercurrent use of nonprotocol antibiotics, and failure to culture fastidious bacteria (eg, Mycoplasma hominis, Ureaplasma urealyticum) leading to misclassifications of women as noninfected.

Role of empiric antibiotic therapy – Empiric antibiotic therapy does not reduce PTB. A meta-analysis of 17 randomized trials evaluated use of prophylactic antibiotics for prevention of PTB based on abnormal vaginal flora (12 trials), a previous PTB (three trials), and a positive fetal fibronectin test result (two trials) [146]. There was no significant association between antibiotic treatment and reduction in PTB regardless of the criteria used to assess risk, the antimicrobial drug administered, or gestational age at the time of treatment (overall combined random effect for delivery at less than 37 weeks RR 1.03, 95% CI 0.86-1.24). Another meta-analysis of randomized trials limited studies of antibiotic prophylaxis in the second or third trimester also found that the intervention did not significantly reduce the risk of PTB (RR 0.85, 95% CI 0.64-1.14, five trials, 1480 women) or preterm prelabor rupture of membranes (RR 0.31, 95% CI 0.06-1.49, one trial, 229 women); however, the included studies were of low methodologic quality [147]. Subsequent randomized trials have also not shown a benefit [148].

Chlamydia, gonorrhea, syphilis – There is no evidence that treatment of chlamydia, gonorrhea, or syphilis prolongs gestation. The only controlled trial that evaluated the effect of treatment of chlamydia on gestational duration did not show a reduction in PTB [149]. However, screening for and treatment of these infections is recommended to prevent other maternal and neonatal sequelae. (See "Clinical manifestations and diagnosis of Neisseria gonorrhoeae infection in adults and adolescents" and "Treatment of uncomplicated Neisseria gonorrhoeae infections" and "Syphilis in pregnancy" and "Treatment of Chlamydia trachomatis infection".)

Bacterial vaginosis, Ureaplasma, GBS – Prospective controlled studies and meta-analyses have reported either a modest or no effect of antibiotic treatment on prolonging gestation in asymptomatic women who screened positive for BV [145,150-153], Ureaplasma urealyticum [154,155], or GBS [156], although these studies are undoubtedly confounded by recolonization or reinfection after therapy and intercurrent use of nonprotocol antibiotics. GBS screening in late pregnancy and chemoprophylaxis for prevention of early-onset neonatal GBS infection is recommended. (See "Prevention of early-onset group B streptococcal disease in neonates".)

Women with BV and a previous PTB may benefit from BV screening and treatment, but there are insufficient data to recommend this as a routine practice. This is discussed in detail separately. (See "Bacterial vaginosis: Initial treatment", section on 'Asymptomatic pregnant persons'.)

Trichomonas – Screening and treatment of asymptomatic Trichomonas infection in HIV-negative women is not recommended during pregnancy because there is no convincing evidence that it reduces the risk for PTB [144,157-161]. By contrast, screening and treatment are recommended for HIV-positive women to reduce the risks for pelvic inflammatory disease and vertical transmission of HIV. (See "Trichomoniasis: Clinical manifestations and diagnosis", section on 'Diagnostic evaluation' and "Trichomoniasis: Treatment", section on 'Pregnant'.)

Malaria — Malaria is associated with PTB, low birth weight, and other maternal and neonatal morbidities [162]. (See "Malaria in pregnancy: Epidemiology, clinical manifestations, diagnosis, and outcome".)

Intervention

Prevention of malaria infection and treatment of established malaria infection can reduce the risk for PTB [163-165]. (See "Malaria in pregnancy: Prevention and treatment".)

BEHAVIOR

Short interpregnancy interval — A short interpregnancy interval has been associated with an increased risk for PTB, even if the previous delivery was at term [166]. The risk is highest in women with a previous PTB. In a study of 263 women with consecutive sPTBs and 299 women with consecutive term births, an interpregnancy interval ≤6 months more than tripled the risk for sPTB less than 34 weeks in the second pregnancy after adjustment of confounders; the risk for late PTB was not affected [167]. (See "Interpregnancy interval: Optimizing time between pregnancies".)

Intervention

Increasing the interval between pregnancies to at least 12 months may reduce a woman's risk for sPTB. In a large cohort study that examined the impact of postpartum contraceptive coverage and use within 18 months of birth in preventing PTB, postpartum contraceptive coverage was protective against PTB [168]. For every month of contraceptive coverage, odds of PTB <37 weeks decreased by 1.1 percent.

The March of Dimes encourages women to space pregnancies at least 18 months apart. However, the interval between pregnancies should not be excessive. A meta-analysis calculated that an interval ≥60 months also increased the risk for PTB (odds ratio [OR] 1.20, 95% CI 1.17-1.24) [169].

Occupational physical activity — A modest relationship between maternal physical activity related to working during pregnancy and PTB has been consistently noted in meta-analyses, but has not been clearly established because available evidence is generally of low quality [170-173]. Factors that have been evaluated include a high cumulative work fatigue score; standing and walking at work for more than three or four hours per day; lifting and carrying >5 kg or ≥11 kg; lifting objects for a combined weight of ≥100 kg per day; lifting and carrying in the third trimester; having a job that required physical effort or physical exertion; and working rotating shifts, fixed night shifts, or longer hours (>40/week). The ORs ranged from 1.1 to 1.6 for all of these associations, and some dose-response patterns were observed.

It is probably important to quantitate all of the factors involved in work-related exertion, as well as the mother's ability to handle stress and fatigue, to gain insight into this controversy. In addition, a "healthy worker" effect is likely present in many studies whereby healthier workers are more likely to continue to work, work longer hours, and work in more demanding jobs, thus biasing outcomes. (See "Working during pregnancy", section on 'Work on pregnancy and child development'.)

Intervention

Women with uncomplicated pregnancies who are employed where there are no greater potential hazards than those encountered in routine daily life may continue to work without interruption until the onset of labor. Nevertheless, the physical demands of the woman's job should be considered, especially in women at high risk of PTB, and working hours and occupational physical activity during pregnancy should be limited using common sense and local guidelines [174-177].

The effects of reducing occupational fatigue have not been evaluated in randomized trials. Maternity legislation in many European countries has regulated work schedules and working conditions for pregnant women; however, none of the European countries except France have experienced a reduction in PTB rates [178]. Nevertheless, paid maternity leave, guaranteed job protection, and regulation of hazardous working conditions remain desirable societal goals.

Bed rest is not helpful. (See 'Bed rest is not helpful' above.)

Exercise — In randomized trials of women with uncomplicated pregnancies, exercise during pregnancy did not increase the risk for PTB [179]. A systematic review of prospective cohort, case-cohort, nested case-control or randomized study design found that exercise (leisure time physical activity) was not associated with an increased risk of PTB, and may decrease the risk by 10 to 14 percent compared with physical inactivity [180]. The optimum time appeared to be two to four hours of physical activity/week. As discussed above, a "healthy exerciser" effect likely exists whereby healthier women and those at low risk of PTB are more likely to continue to exercise during pregnancy. However, it has also been hypothesized that exercise may reduce the risk of PTB by reducing oxidative stress or increasing placental vascularization [181]. (See "Exercise during pregnancy and the postpartum period".)

Coitus — Sexual intercourse is not a risk factor for PTB; therefore, abstinence after pregnancy has been achieved has no role in strategies for prevention of PTB [182-185].

Smoking — Cigarette smoking has a modest dose-dependent relationship with the risk for PTB [3,95,186-192]. This effect may be explained by increased rates of smoking-related complications of pregnancy, such as placental abruption, placenta previa, prelabor rupture of membranes, and fetal growth restriction. However, the association still exists when adjustment is made for these possible confounding factors, suggesting that there may be a direct effect of cigarette smoking on spontaneous preterm labor and delivery [192]. (See "Cigarette and tobacco products in pregnancy: Impact on pregnancy and the neonate".)

Intervention

Smoking cessation should always be encouraged for its general health benefits. It is likely that smokers who decrease or stop cigarette smoking will reduce their risk for PTB, but this has not been proven. (See "Cigarette and tobacco products in pregnancy: Impact on pregnancy and the neonate", section on 'Preterm birth' and "Tobacco and nicotine use in pregnancy: Cessation strategies and treatment options".)

Since 2010, in the United States, Medicaid programs are required to cover tobacco-cessation counseling and drug therapy for pregnant women without cost sharing, which might increase utilization of these services [193].

Substance use — Maternal substance use increases the risk of PTB, but it is difficult to separate the risk attributable to the substance from other risk factors, which are common in these patients [95,189-192,194-198]. In one study, women with cocaine-positive urine samples were at fourfold increased risk of developing preterm labor [196]. Another series found positive urine toxicology in 24 of 141 (17 percent) of women with preterm labor compared with 3 of 108 (2.8 percent) controls with uncomplicated labor at term [195]. Cocaine was the most common substance identified and was detected in approximately 60 percent of women in preterm labor with positive toxicology tests. Alcohol [197,199] and toluene [198] are additional substances associated with an increased risk of preterm labor and birth. In women who use multiple drugs, risk of PTB has been reported to range from 25 to 63 percent [200,201]. (See "Substance use during pregnancy: Screening and prenatal care" and "Alcohol intake and pregnancy".)

Intervention

Health care providers should attempt to identify maternal substance use, provide information on the maternal and fetal risks associated with this practice, and help patients to stop using these drugs. It is likely that this will reduce their risk for PTB, but this has not been proven. (See "Substance use during pregnancy: Screening and prenatal care" and "Overview of management of opioid use disorder during pregnancy" and "Methadone and buprenorphine pharmacotherapy of opioid use disorder during pregnancy".)

DIET — Women with adequate nutrition and a normal body mass index have better pregnancy outcomes than other women, which suggests that nutritional interventions may have a role in preventing PTB in selected populations.

There is some evidence supporting the hypothesis that maternal undernutrition in pregnancy results in PTB [202]. In sheep, moderate maternal undernutrition around the time of conception results in accelerated maturation of the fetal hypothalamic-pituitary-adrenal axis, a precocious fetal cortisol surge, and PTB [203,204]. In Gambian women, pregnancies conceived during the rainy season when food is scarce were significantly shorter than those conceived when food was more plentiful [205]. Observations of shorter gestational length with early pregnancy exposure to the Dutch famine also support this hypothesis [206]. Thus, focusing on dietary events around the time of conception may be important in prevention of some cases of PTB.

Intervention

In systematic reviews, isocaloric protein supplements [207], balanced protein/energy supplements [208], and high protein supplements [208] did not reduce the rate of PTB. Most studies show that vitamin supplements during pregnancy do not reduce the risk of PTB [209-215], although they have other benefits. There may be potential benefits of micronutrient supplementation in specific subpopulations of pregnant women, such as those who are undernourished or HIV-infected [216].

In a 2018 meta-analysis of placebo-controlled randomized trials of omega-3 long-chain polyunsaturated fatty acids (n-3 PUFA) supplements or dietary additions during pregnancy, n-3 PUFA reduced PTB <37 and <34 weeks, with a corresponding trend for reduction in perinatal death [217]. These and subsequent discordant data and recommendations regarding fish consumption and n-3 PUFA supplementation during pregnancy are discussed in detail separately. (See "Fish consumption and marine omega-3 fatty acid supplementation in pregnancy".)

WEIGHT AND WEIGHT CHANGES — Extremes of prepregnancy weight and/or body mass index have been associated with increased rates of PTB [218-222]. The strength of this association is not well-defined because the effect is bimodal as opposed to linear and because of interdependent variables [223]. For example, low prepregnancy weight may be confounded by socioeconomic status, race/ethnicity, and even weight gain in pregnancy.

Obese pregnant women are at increased risk of iatrogenic PTB resulting from medical complications. Obesity also appears to increase the risk for preterm prelabor rupture of membranes (PPROM) and may increase the risk of sPTB without PPROM. A potential effect on sPTB is hypothesized to be mediated by the inflammatory state, but data are weak. (See "Obesity in pregnancy: Complications and maternal management", section on 'Indicated and spontaneous preterm birth'.)

Low and high weight gain during pregnancy have also been associated with PTB [224-226]. These issues are discussed in detail separately. (See "Gestational weight gain".)

Intervention

Although some evidence suggests that weight loss in obese women before pregnancy and appropriate weight gain in pregnancy can reduce the risk for PTB, the evidence is not definitive. In a meta-analysis of randomized trials of the effects of dietary and lifestyle interventions in pregnancy on maternal weight and obstetric outcomes, the reduction in PTB was not statistically significant (relative risk 0.78, 95% CI 0.60-1.02, 13 trials, 2652 women) [227]. However, these trials had significant heterogeneity and were of low quality. (See "Obesity in pregnancy: Complications and maternal management" and "Gestational weight gain".)

Regardless of its effect on pregnancy, weight loss before pregnancy should be recommended for obese women because of general health benefits. (See "Overweight and obesity in adults: Health consequences".)

Women with eating disorders can also benefit from intervention to achieve a normal weight. (See "Eating disorders in pregnancy".)

HEIGHT — Women with shorter stature appear to be at increased risk for PTB and taller women appear to be decreased risk [228-230].

STRESS — Most women report experiencing at least one stressful life event in the year before giving birth [231]. An association between stress (including posttraumatic stress disorder) and PTB is biologically plausible. There is evidence that maternal and fetal stress activates cells in the placenta, decidua, and fetal membranes to produce corticotropin-releasing hormone (CRH) [232]. CRH can enhance local prostaglandin production, which initiates contractions. However, studies have not consistently demonstrated a relationship between maternal stress, CRH concentration, and PTB [233-235]. (See "Spontaneous preterm birth: Pathogenesis", section on '#1 Stress-induced premature activation of the HPA axis'.)

When maternal psychosocial stress has been associated with an increased risk of PTB, the risk was modest: odds ratio 1.42 (95% CI 1.05-1.91) in cohort studies [236]. Analysis of data is complicated by difficulty defining and measuring maternal stress, assessments at different times during pregnancy, variations in adjustment of confounders, lack of differentiation between acute and chronic stressors, and discordant baseline characteristics of the populations studied [237].

Intervention

Although social support during pregnancy has resulted in improvements in immediate psychosocial outcome, it has not been shown to significantly reduce the rate of PTB in stressed gravida. A systematic review concluded that social support was not sufficiently powerful to improve the obstetric outcome of the pregnancy in which it was provided, possibly because of the immense social deprivation experienced by most of the women in the trials examined [238].

There are limited data on other interventions for reducing stress in pregnant women (eg, relaxation or mind-body therapies [eg, meditation, massage, yoga, breathing exercises, music therapy, aromatherapy]). Available trials are small and of poor quality; clear effects on birth outcomes have not been proven [239].

ENVIRONMENT — Systematic reviews have reported an association between PTB and fine particulate matter and ozone in the air, high environmental temperature, and phthalate exposure [240-247]. Although the effects were small and limited by differences in study designs, particularly assessment of exposure, a causal effect is possible. (See "Occupational and environmental risks to reproduction in females: Specific exposures and impact".)

SUBOPTIMAL PRENATAL CARE — The absence of prenatal care has been consistently identified as a risk factor for preterm labor and delivery, but it is less clear whether this association is causal or a marker for other factors that contribute to PTB. (See "Prenatal care: Initial assessment" and "Prenatal care: Second and third trimesters".)

Intervention

Regular prenatal care should be encouraged and improves perinatal outcome in women with underlying medical disorders (eg, diabetes, chronic hypertension, thyroid disease) or pregnancy-related conditions (eg, preeclampsia); however, the March of Dimes trial discussed below suggests enhanced care is unlikely to decrease the incidence of PTB.

Retrospective studies cannot be adequately controlled to adjust for confounding factors, while randomized trials (no prenatal care versus standard care) would be unethical. Therefore, the only well-designed studies on the effect of prenatal care on PTB compare standard with enhanced care (ie, some combination of patient education, case management, home visits, nutrition counseling, and extra prenatal visits and cervical examinations).

The March of Dimes Multicenter Prematurity Prevention Trial assigned 2395 women with singleton or multiple gestations at high risk for PTB to either standard of care or an enhanced care intervention (more frequent prenatal visits, improved patient education regarding symptoms and signs of preterm labor, and weekly pelvic examinations after 20 to 24 weeks of gestation) [19]. There was no significant difference between the two groups in sPTB rates. A meta-analysis that included three trials of women with singleton pregnancies also found no clear evidence that specialized antenatal clinics reduced PTB [248].

PRETERM LABOR — Uterine contractions are an essential component of labor, but mild irregular contractions are a normal finding at all stages of pregnancy, thereby adding to the challenge of distinguishing true labor (contractions that result in cervical change) from false labor (contractions that do not result in cervical change [ie, Braxton-Hicks contractions]). Only 13 percent of women presenting at <34 weeks of gestation and meeting explicit contraction criteria for preterm labor deliver within one week. (See "Preterm labor: Clinical findings, diagnostic evaluation, and initial treatment".)

Intervention

Tocolytic therapy of an acute episode of idiopathic preterm labor often abolishes contractions temporarily but does not remove the underlying stimulus that initiated the process of parturition (eg, infection, inflammation, uterine overdistention, decidual hemorrhage, cervical insufficiency) or reverse parturitional changes in the uterus. The net effect is that tocolytics are unlikely to prolong pregnancy by weeks or months. However, short-term tocolysis is endorsed by most guidelines since delivery can often be delayed for at least 48 hours, thus enabling maternal administration of glucocorticoids to achieve the maximum fetal effect [77]. (See "Inhibition of acute preterm labor" and "Antenatal corticosteroid therapy for reduction of neonatal respiratory morbidity and mortality from preterm delivery".)

FETAL FACTORS — Male sex is a risk factor for sPTB [249-252]. Certain congenital anomalies [253,254] and growth restriction [255-260] are risk factors for sPTB and indicated PTB.

PATERNAL RISK FACTORS — No paternal risk factors for development of PTB in their partners have been identified [261]. PTB risk does not appear to be affected by the father's history of preterm children with other women or PTBs to members of the father's family [262].

PREDICTING RISK FOR PRETERM BIRTH

Risk scoring systems — Risk scoring is a quantitative method used to identify women at increased risk for PTB. Proposed systems typically calculate an additive score based on points assigned to arbitrarily selected or weighted epidemiologic, historical, and clinical risk factors [95,191,192,194].

Systematic reviews have concluded that there is no effective risk scoring system for prediction of PTB [263,264]. This is due to our lack of knowledge regarding the cause(s) of PTB in most women and because the most powerful risk factor is previous PTB, which is not applicable to nulliparous women. The positive predictive value (the percent of women defined as high risk that actually go on to have a PTB) of most risk scoring systems is low, 20 to 30 percent, and varies according to the population studied [195].

Biomarkers — Cervicovaginal fetal fibronectin (fFN) can be a useful biomarker for predicting PTB within 7 to 14 days in women with contractions and mild cervical dilation and effacement, particularly when combined with ultrasound assessment of cervical length and when a quantitative measurement is available. The predictive value of fFN for PTB more than 14 days after testing is poor. (See "Preterm labor: Clinical findings, diagnostic evaluation, and initial treatment", section on '<34 weeks of gestation'.)

Fetal fibronectin may be useful for predicting risk of PTB in asymptomatic high-risk women (eg, previous PTB). A fFN ≥50 ng/mL at 22 to 27+6 weeks of gestation had sensitivity 55 percent and positive predictive value 27 percent for prediction of PTB <34 weeks in one study [265]. An algorithm combining quantitative fFN (not available in the United States) and cervical length, demographic information, and obstetric history (whether previous sPTB/preterm prelabor rupture of membranes or current suspected preterm labor) has been incorporated into an App (QUiPP) for prediction of sPTB in Europe [266,267]. In a randomized multicenter trial involving nearly 1900 pregnant patients between 23+0 and 34+6 weeks of gestation with symptoms of preterm labor, the QUiPP app did not reduce unnecessary treatments compared with routine management [268].

By contrast, fFN is not useful as a screening test for predicting risk of PTB in asymptomatic nulliparous women. In the largest prospective cohort study of use of fFN in asymptomatic low-risk nulliparous women with singleton pregnancies and cervical length >15 mm (n = 9410), the sensitivity and positive predictive value of fFN ≥50 ng/mL at 22 to 30 weeks of gestation for PTB <32 weeks was 32.1 and 3.1 percent, respectively [269]. Using a lower or higher threshold did not significantly improve overall test performance.

A test (PreTRM Test) for two serum proteins, insulin-like growth factor-binding protein 4 and sex hormone-binding globulin, is available for clinical use to predict PTB. In a study to develop and validate a mass spectrometry-based serum test to predict spontaneous preterm delivery in asymptomatic pregnant women, the test had sensitivity and specificity of 0.75 and 0.74, respectively, for predicting PTB <37 weeks [270]. We recommend not moving forward with serum screening for PTB until such screening has been adequately tested and validated.

Over 30 other biomarkers have been studied for identification of asymptomatic women at high risk of PTB. A systematic review of these biomarkers included 72 observational studies involving almost 90,000 women and concluded that none of these other biomarkers (alone or in combination) was clinically useful for predicting sPTB in asymptomatic women [271]. The markers included inflammation-related biomarkers, placental protein/hormone-related biomarkers, angiogenesis-related biomarkers, coagulation-related biomarkers, genetic-biomarkers, and proteomic-related biomarkers.

PROGNOSIS

Long-term maternal consequences

Cardiovascular disease – Women who deliver preterm are at increased risk for cardiovascular morbidity and mortality years after the delivery. It is unclear why sPTB appears to be a marker for later cardiovascular disease or whether women who deliver preterm should be identified by primary care providers and encouraged to optimize modifiable risk factors for cardiovascular disease more so than women without this history. (See "Overview of primary prevention of cardiovascular disease".)

In a meta-analysis of 10 cohort studies, women who had a sPTB were at higher risk for the following cardiovascular events when compared with women who delivered at term and followed for 12 to 35 years postpartum [272]:

-Fatal and nonfatal ischemic heart disease (hazard ratio [HR] 1.38, 95% CI 1.22-1.57)

-Fatal and nonfatal stroke (HR 1.71, 95% CI 1.53-1.91)

-Fatal and nonfatal overall cardiovascular disease (HR 2.01, 95% CI 1.52-2.65)

Others have shown that the risk increases with earlier PTB. In a national cohort study from Sweden, women who delivered preterm or extremely preterm had 2.5- and 4-fold increased risks of ischemic heart disease in the next 10 years compared with those who delivered full-term, even after adjusting for other maternal factors including preeclampsia, diabetes, obesity, and smoking [273].

All-cause mortality – In a national cohort study with over 50 million person-years of follow-up and over 76,000 deaths (median age at death 57.6 years), the adjusted HR (aHR) for all-cause mortality associated with preterm versus full term delivery (39 to 41 weeks) was aHR 1.73, 95% CI 1.61-1.87 during the 10 years after delivery, and increased with decreasing gestational age at preterm delivery (aHR 2.20 for preterm delivery at 22 to 27 weeks, 2.28 for preterm delivery at 28 to 33 weeks, 1.52 for preterm delivery at 34 to 36 weeks, and 1.19 for early term delivery at 37 to 38 weeks) [274]. The risks declined but remained significantly raised up to 40 years later. Several causes were identified, including cardiovascular and respiratory disorders, diabetes, and cancer, and were independent of shared genetic or environmental factors within families.

Offspring prognosis

(See "Short-term complications of the preterm infant".)

(See "Preterm birth: Definitions of prematurity, epidemiology, and risk factors for infant mortality".)

(See "Late preterm infants".)

(See "Long-term outcome of the preterm infant".)

(See "Long-term neurodevelopmental impairment in infants born preterm: Epidemiology and risk factors".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Preterm labor and birth".)

INFORMATION FOR PATIENTS — UpToDate offers two types of patient education materials, "The Basics" and "Beyond the Basics." The Basics patient education pieces are written in plain language, at the 5th to 6th grade reading level, and they answer the four or five key questions a patient might have about a given condition. These articles are best for patients who want a general overview and who prefer short, easy-to-read materials. Beyond the Basics patient education pieces are longer, more sophisticated, and more detailed. These articles are written at the 10th to 12th grade reading level and are best for patients who want in-depth information and are comfortable with some medical jargon.

Here are the patient education articles that are relevant to this topic. We encourage you to print or e-mail these topics to your patients. (You can also locate patient education articles on a variety of subjects by searching on "patient info" and the keyword(s) of interest.)

Basics topic (see "Patient education: Preterm labor (The Basics)")

Beyond the Basics topics (see "Patient education: Preterm labor (Beyond the Basics)" and "Patient education: Bacterial vaginosis (Beyond the Basics)" and "Patient education: Management of a cervical biopsy with precancerous cells (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

Overview – There are many risk factors for preterm labor and delivery (table 2). Some are reversible, others are permanent. Identification of risk factors for spontaneous preterm birth (sPTB) before conception or early in pregnancy ideally would lead to interventions that could help prevent this complication. (See 'Introduction' above.)

Previous PTB

Prior PTB is the strongest risk factor for future PTB, and recurrences often occur at the same gestational age. The risk of recurrent PTB is higher in patients with earlier gestational age at PTB and those with multiple PTBs (ie, 15 to 30 percent after one PTB and up to 60 percent after two PTBs). Term births decrease the risk of PTB in subsequent pregnancies (table 3 and table 4). (See 'History of spontaneous preterm birth' above.)

For women with a history of sPTB, progesterone supplementation reduces the risk of recurrent sPTB by approximately 30 percent. (See "Progesterone supplementation to reduce the risk of spontaneous preterm labor and birth", section on 'Candidates for progesterone supplementation'.)

Short cervix – Short cervical length (≤25 mm) on transvaginal ultrasound examination between 18 and 24 weeks of gestation in the current pregnancy is a risk factor for PTB and is the basis for screening for a short cervix in the midtrimester. (See 'Short cervix' above.)

For women with no previous history of sPTB who develop a short cervix, vaginal progesterone supplementation may prolong gestation. (See "Progesterone supplementation to reduce the risk of spontaneous preterm labor and birth", section on 'Candidates for progesterone supplementation'.)

For women with a history of sPTB who develop a short cervix despite progesterone supplementation, placement of a cerclage may prolong gestation. (See "Cervical insufficiency".)

Smoking cessation, treatment of drug misuse, treatment of asymptomatic bacteriuria, and achievement/maintenance of a normal body mass index – These interventions have health benefits and may also reduce the risk of PTB. (See 'Smoking' above and 'Substance use' above and 'Asymptomatic bacteriuria' above and 'Weight and weight changes' above.)

Interpregnancy interval – Avoiding an interpregnancy interval of less than six months, and ideally less than 12 months, may reduce a woman's risk for sPTB. (See 'Short interpregnancy interval' above.)

Multiple gestation – Singleton gestations are less likely to deliver preterm than multiple gestations. When fertility enhancing interventions are used, prevention and reduction of multifetal gestations, particularly high-order multifetal gestations, can reduce the risk of PTB. (See 'Multifetal gestation' above.)

Low-dose aspirin – We do not prescribe low-dose aspirin for prevention of sPTB, in agreement with the American College of Obstetricians and Gynecologists' guidelines. We prescribe low-dose aspirin for pregnant women with moderate or high risk factors for preeclampsia to reduce their risk for that disorder and its sequelae, which includes PTB. (See 'Ineffective and unproven interventions' above and "Preeclampsia: Prevention", section on 'Low-dose aspirin'.)

Biomarkers – No biomarker performs well as a screening test for predicting sPTB in asymptomatic low risk women. (See 'Biomarkers' above.)

Long-term maternal cardiovascular risk – Women who deliver preterm are at increased risk for cardiovascular morbidity and mortality and all-cause mortality for years after the delivery. (See 'Long-term maternal consequences' above.)

  1. Bloom SL, Yost NP, McIntire DD, Leveno KJ. Recurrence of preterm birth in singleton and twin pregnancies. Obstet Gynecol 2001; 98:379.
  2. Esplin MS, O'Brien E, Fraser A, et al. Estimating recurrence of spontaneous preterm delivery. Obstet Gynecol 2008; 112:516.
  3. Bhattacharya S, Raja EA, Mirazo ER, et al. Inherited predisposition to spontaneous preterm delivery. Obstet Gynecol 2010; 115:1125.
  4. Mercer BM, Goldenberg RL, Moawad AH, et al. The preterm prediction study: effect of gestational age and cause of preterm birth on subsequent obstetric outcome. National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Am J Obstet Gynecol 1999; 181:1216.
  5. Lykke JA, Paidas MJ, Langhoff-Roos J. Recurring complications in second pregnancy. Obstet Gynecol 2009; 113:1217.
  6. Kristensen J, Langhoff-Roos J, Kristensen FB. Implications of idiopathic preterm delivery for previous and subsequent pregnancies. Obstet Gynecol 1995; 86:800.
  7. Laughon SK, Albert PS, Leishear K, Mendola P. The NICHD Consecutive Pregnancies Study: recurrent preterm delivery by subtype. Am J Obstet Gynecol 2014; 210:131.e1.
  8. Yamashita M, Hayashi S, Endo M, et al. Incidence and risk factors for recurrent spontaneous preterm birth: A retrospective cohort study in Japan. J Obstet Gynaecol Res 2015; 41:1708.
  9. Ferrero DM, Larson J, Jacobsson B, et al. Cross-Country Individual Participant Analysis of 4.1 Million Singleton Births in 5 Countries with Very High Human Development Index Confirms Known Associations but Provides No Biologic Explanation for 2/3 of All Preterm Births. PLoS One 2016; 11:e0162506.
  10. Drassinower D, Običan SG, Siddiq Z, et al. Does the clinical presentation of a prior preterm birth predict risk in a subsequent pregnancy? Am J Obstet Gynecol 2015; 213:686.e1.
  11. Menzies R, Li ALK, Melamed N, et al. Risk of singleton preterm birth after prior twin preterm birth: a systematic review and meta-analysis. Am J Obstet Gynecol 2020; 223:204.e1.
  12. Schaaf JM, Hof MH, Mol BW, et al. Recurrence risk of preterm birth in subsequent twin pregnancy after preterm singleton delivery. BJOG 2012; 119:1624.
  13. van Vliet EO, Askie LA, Mol BW, Oudijk MA. Antiplatelet Agents and the Prevention of Spontaneous Preterm Birth: A Systematic Review and Meta-analysis. Obstet Gynecol 2017; 129:327.
  14. Andrikopoulou M, Purisch SE, Handal-Orefice R, Gyamfi-Bannerman C. Low-dose aspirin is associated with reduced spontaneous preterm birth in nulliparous women. Am J Obstet Gynecol 2018; 219:399.e1.
  15. Hoffman MK, Goudar SS, Kodkany BS, et al. Low-dose aspirin for the prevention of preterm delivery in nulliparous women with a singleton pregnancy (ASPIRIN): a randomised, double-blind, placebo-controlled trial. Lancet 2020; 395:285.
  16. Landman AJEMC, de Boer MA, Visser L, et al. Evaluation of low-dose aspirin in the prevention of recurrent spontaneous preterm labour (the APRIL study): A multicentre, randomised, double-blinded, placebo-controlled trial. PLoS Med 2022; 19:e1003892.
  17. Hodgetts Morton V, Stock SJ. Low-dose aspirin for the prevention of preterm birth: More questions than answers. PLoS Med 2022; 19:e1003908.
  18. American College of Obstetricians and Gynecologists' Practice Advisory: Low-Dose Aspirin Use for the Prevention of Preeclampsia and Related Morbidity and Mortality. December 2021. https://www.acog.org/clinical/clinical-guidance/practice-advisory/articles/2021/12/low-dose-aspirin-use-for-the-prevention-of-preeclampsia-and-related-morbidity-and-mortality (Accessed on December 21, 2021).
  19. Multicenter randomized, controlled trial of a preterm birth prevention program. Collaborative Group on Preterm Birth Prevention. Am J Obstet Gynecol 1993; 169:352.
  20. Urquhart C, Currell R, Harlow F, Callow L. Home uterine monitoring for detecting preterm labour. Cochrane Database Syst Rev 2017; 2:CD006172.
  21. American College of Obstetricians and Gynecologists' Committee on Practice Bulletins—Obstetrics. Prediction and Prevention of Spontaneous Preterm Birth: ACOG Practice Bulletin, Number 234. Obstet Gynecol 2021; 138:e65.
  22. Whitworth M, Quenby S. Prophylactic oral betamimetics for preventing preterm labour in singleton pregnancies. Cochrane Database Syst Rev 2008; :CD006395.
  23. Khanprakob T, Laopaiboon M, Lumbiganon P, Sangkomkamhang US. Cyclo-oxygenase (COX) inhibitors for preventing preterm labour. Cochrane Database Syst Rev 2012; 10:CD007748.
  24. Saccone G, Perriera L, Berghella V. Prior uterine evacuation of pregnancy as independent risk factor for preterm birth: a systematic review and metaanalysis. Am J Obstet Gynecol 2016; 214:572.
  25. Zhang G, Feenstra B, Bacelis J, et al. Genetic Associations with Gestational Duration and Spontaneous Preterm Birth. N Engl J Med 2017; 377:1156.
  26. Boivin A, Luo ZC, Audibert F, et al. Risk for preterm and very preterm delivery in women who were born preterm. Obstet Gynecol 2015; 125:1177.
  27. Koire A, Chu DM, Aagaard K. Family history is a predictor of current preterm birth. Am J Obstet Gynecol MFM 2021; 3:100277.
  28. Ward K, Argyle V, Meade M, Nelson L. The heritability of preterm delivery. Obstet Gynecol 2005; 106:1235.
  29. Clausson B, Lichtenstein P, Cnattingius S. Genetic influence on birthweight and gestational length determined by studies in offspring of twins. BJOG 2000; 107:375.
  30. Hao K, Wang X, Niu T, et al. A candidate gene association study on preterm delivery: application of high-throughput genotyping technology and advanced statistical methods. Hum Mol Genet 2004; 13:683.
  31. Treloar SA, Macones GA, Mitchell LE, Martin NG. Genetic influences on premature parturition in an Australian twin sample. Twin Res 2000; 3:80.
  32. Annells MF, Hart PH, Mullighan CG, et al. Interleukins-1, -4, -6, -10, tumor necrosis factor, transforming growth factor-beta, FAS, and mannose-binding protein C gene polymorphisms in Australian women: Risk of preterm birth. Am J Obstet Gynecol 2004; 191:2056.
  33. Winkvist A, Mogren I, Högberg U. Familial patterns in birth characteristics: impact on individual and population risks. Int J Epidemiol 1998; 27:248.
  34. Macones GA, Parry S, Elkousy M, et al. A polymorphism in the promoter region of TNF and bacterial vaginosis: preliminary evidence of gene-environment interaction in the etiology of spontaneous preterm birth. Am J Obstet Gynecol 2004; 190:1504.
  35. Kistka ZA, DeFranco EA, Ligthart L, et al. Heritability of parturition timing: an extended twin design analysis. Am J Obstet Gynecol 2008; 199:43.e1.
  36. Velez DR, Fortunato S, Thorsen P, et al. Spontaneous preterm birth in African Americans is associated with infection and inflammatory response gene variants. Am J Obstet Gynecol 2009; 200:209.e1.
  37. Svensson AC, Sandin S, Cnattingius S, et al. Maternal effects for preterm birth: a genetic epidemiologic study of 630,000 families. Am J Epidemiol 2009; 170:1365.
  38. Wilcox AJ, Skjaerven R, Lie RT. Familial patterns of preterm delivery: maternal and fetal contributions. Am J Epidemiol 2008; 167:474.
  39. Srinivasjois RM, Shah S, Shah PS, Knowledge Synthesis Group on Determinants Of Preterm/LBW Births. Biracial couples and adverse birth outcomes: a systematic review and meta-analyses. Acta Obstet Gynecol Scand 2012; 91:1134.
  40. Grobman WA, Parker CB, Willinger M, et al. Racial Disparities in Adverse Pregnancy Outcomes and Psychosocial Stress. Obstet Gynecol 2018; 131:328.
  41. Iams JD, Goldenberg RL, Mercer BM, et al. The Preterm Prediction Study: recurrence risk of spontaneous preterm birth. National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Am J Obstet Gynecol 1998; 178:1035.
  42. Manuck TA, Lai Y, Meis PJ, et al. Admixture mapping to identify spontaneous preterm birth susceptibility loci in African Americans. Obstet Gynecol 2011; 117:1078.
  43. Genc MR, Onderdonk A. Endogenous bacterial flora in pregnant women and the influence of maternal genetic variation. BJOG 2011; 118:154.
  44. Tsai HJ, Hong X, Chen J, et al. Role of African ancestry and gene-environment interactions in predicting preterm birth. Obstet Gynecol 2011; 118:1081.
  45. Frey HA, Stout MJ, Pearson LN, et al. Genetic variation associated with preterm birth in African-American women. Am J Obstet Gynecol 2016; 215:235.e1.
  46. Wen A, Srinivasan U, Goldberg D, et al. Selected vaginal bacteria and risk of preterm birth: an ecological perspective. J Infect Dis 2014; 209:1087.
  47. Fettweis JM, Brooks JP, Serrano MG, et al. Differences in vaginal microbiome in African American women versus women of European ancestry. Microbiology 2014; 160:2272.
  48. Hyman RW, Fukushima M, Jiang H, et al. Diversity of the vaginal microbiome correlates with preterm birth. Reprod Sci 2014; 21:32.
  49. Vinturache AE, Gyamfi-Bannerman C, Hwang J, et al. Maternal microbiome - A pathway to preterm birth. Semin Fetal Neonatal Med 2016; 21:94.
  50. Genc MR, Witkin SS, Delaney ML, et al. A disproportionate increase in IL-1beta over IL-1ra in the cervicovaginal secretions of pregnant women with altered vaginal microflora correlates with preterm birth. Am J Obstet Gynecol 2004; 190:1191.
  51. Simhan HN, Caritis SN, Krohn MA, et al. Decreased cervical proinflammatory cytokines permit subsequent upper genital tract infection during pregnancy. Am J Obstet Gynecol 2003; 189:560.
  52. Simhan HN, Krohn MA. First-trimester cervical inflammatory milieu and subsequent early preterm birth. Am J Obstet Gynecol 2009; 200:377.e1.
  53. Fuchs F, Monet B, Ducruet T, et al. Effect of maternal age on the risk of preterm birth: A large cohort study. PLoS One 2018; 13:e0191002.
  54. Lawlor DA, Mortensen L, Andersen AM. Mechanisms underlying the associations of maternal age with adverse perinatal outcomes: a sibling study of 264 695 Danish women and their firstborn offspring. Int J Epidemiol 2011; 40:1205.
  55. Fraser AM, Brockert JE, Ward RH. Association of young maternal age with adverse reproductive outcomes. N Engl J Med 1995; 332:1113.
  56. Mayo JA, Lu Y, Stevenson DK, et al. Parental age and preterm birth: a population-based cohort of nearly 3 million California livebirths from 2007 to 2012. J Perinatol 2021; 41:2156.
  57. Shapiro-Mendoza CK, Barfield WD, Henderson Z, et al. CDC Grand Rounds: Public Health Strategies to Prevent Preterm Birth. MMWR Morb Mortal Wkly Rep 2016; 65:826.
  58. Lemmers M, Verschoor MA, Hooker AB, et al. Dilatation and curettage increases the risk of subsequent preterm birth: a systematic review and meta-analysis. Hum Reprod 2016; 31:34.
  59. Koike T, Minakami H, Kosuge S, et al. Uterine leiomyoma in pregnancy: its influence on obstetric performance. J Obstet Gynaecol Res 1999; 25:309.
  60. Davis JL, Ray-Mazumder S, Hobel CJ, et al. Uterine leiomyomas in pregnancy: a prospective study. Obstet Gynecol 1990; 75:41.
  61. Rice JP, Kay HH, Mahony BS. The clinical significance of uterine leiomyomas in pregnancy. Am J Obstet Gynecol 1989; 160:1212.
  62. Smith GC, Wood AM, Pell JP, Dobbie R. Sudden infant death syndrome and complications in other pregnancies. Lancet 2005; 366:2107.
  63. Kiely JL. What is the population-based risk of preterm birth among twins and other multiples? Clin Obstet Gynecol 1998; 41:3.
  64. TambyRaja RL, Ratnam SS. Plasma steroid changes in twin pregnancies. Prog Clin Biol Res 1981; 69A:189.
  65. Muechler EK, Huang KE. Plasma estrogen and progesterone in quintuplet pregnancy induced with menotropins. Am J Obstet Gynecol 1983; 147:105.
  66. Weiss G, Goldsmith LT, Sachdev R, et al. Elevated first-trimester serum relaxin concentrations in pregnant women following ovarian stimulation predict prematurity risk and preterm delivery. Obstet Gynecol 1993; 82:821.
  67. Lykke JA, Dideriksen KL, Lidegaard O, Langhoff-Roos J. First-trimester vaginal bleeding and complications later in pregnancy. Obstet Gynecol 2010; 115:935.
  68. Szymusik I, Bartnik P, Wypych K, et al. The association of first trimester bleeding with preterm delivery. J Perinat Med 2015; 43:525.
  69. Lockwood CJ. Risk factors for preterm birth and new approaches to its early diagnosis. J Perinat Med 2015; 43:499.
  70. Lockwood CJ, Kayisli UA, Stocco C, et al. Abruption-induced preterm delivery is associated with thrombin-mediated functional progesterone withdrawal in decidual cells. Am J Pathol 2012; 181:2138.
  71. Mackenzie AP, Schatz F, Krikun G, et al. Mechanisms of abruption-induced premature rupture of the fetal membranes: Thrombin enhanced decidual matrix metalloproteinase-3 (stromelysin-1) expression. Am J Obstet Gynecol 2004; 191:1996.
  72. Lockwood CJ, Toti P, Arcuri F, et al. Mechanisms of abruption-induced premature rupture of the fetal membranes: thrombin-enhanced interleukin-8 expression in term decidua. Am J Pathol 2005; 167:1443.
  73. Manuck TA, Esplin MS, Biggio J, et al. Predictors of response to 17-alpha hydroxyprogesterone caproate for prevention of recurrent spontaneous preterm birth. Am J Obstet Gynecol 2016; 214:376.e1.
  74. Moscrop A. Can sex during pregnancy cause a miscarriage? A concise history of not knowing. Br J Gen Pract 2012; 62:e308.
  75. Ribeiro MC, de Tubino Scanavino M, do Amaral MLS, et al. Beliefs About Sexual Activity During Pregnancy: A Systematic Review of the Literature. J Sex Marital Ther 2017; 43:822.
  76. Newman RB, Goldenberg RL, Iams JD, et al. Preterm prediction study: comparison of the cervical score and Bishop score for prediction of spontaneous preterm delivery. Obstet Gynecol 2008; 112:508.
  77. Medley N, Poljak B, Mammarella S, Alfirevic Z. Clinical guidelines for prevention and management of preterm birth: a systematic review. BJOG 2018; 125:1361.
  78. Goldenberg RL, Cliver SP, Bronstein J, et al. Bed rest in pregnancy. Obstet Gynecol 1994; 84:131.
  79. Sciscione AC. Maternal activity restriction and the prevention of preterm birth. Am J Obstet Gynecol 2010; 202:232.e1.
  80. Sosa CG, Althabe F, Belizán JM, Bergel E. Bed rest in singleton pregnancies for preventing preterm birth. Cochrane Database Syst Rev 2015; :CD003581.
  81. Grobman WA, Gilbert SA, Iams JD, et al. Activity restriction among women with a short cervix. Obstet Gynecol 2013; 121:1181.
  82. Fox NS, Jean-Pierre C, Predanic M, Chasen ST. Does hospitalization prevent preterm delivery in the patient with a short cervix? Am J Perinatol 2007; 24:49.
  83. Yost NP, Bloom SL, McIntire DD, Leveno KJ. Hospitalization for women with arrested preterm labor: a randomized trial. Obstet Gynecol 2005; 106:14.
  84. Zemet R, Schiff E, Manovitch Z, et al. Quantitative assessment of physical activity in pregnant women with sonographic short cervix and the risk for preterm delivery: A prospective pilot study. PLoS One 2018; 13:e0198949.
  85. Kovacevich GJ, Gaich SA, Lavin JP, et al. The prevalence of thromboembolic events among women with extended bed rest prescribed as part of the treatment for premature labor or preterm premature rupture of membranes. Am J Obstet Gynecol 2000; 182:1089.
  86. Crowther CA, Han S. Hospitalisation and bed rest for multiple pregnancy. Cochrane Database Syst Rev 2010; :CD000110.
  87. Abdul Sultan A, West J, Tata LJ, et al. Risk of first venous thromboembolism in pregnant women in hospital: population based cohort study from England. BMJ 2013; 347:f6099.
  88. Convertino VA, Bloomfield SA, Greenleaf JE. An overview of the issues: physiological effects of bed rest and restricted physical activity. Med Sci Sports Exerc 1997; 29:187.
  89. Norwitz ER, Robinson JN, Challis JR. The control of labor. N Engl J Med 1999; 341:660.
  90. Salafia CM, Vogel CA, Vintzileos AM, et al. Placental pathologic findings in preterm birth. Am J Obstet Gynecol 1991; 165:934.
  91. Klein LL, Gibbs RS. Use of microbial cultures and antibiotics in the prevention of infection-associated preterm birth. Am J Obstet Gynecol 2004; 190:1493.
  92. Goldenberg RL, Andrews WW, Hauth JC. Choriodecidual infection and preterm birth. Nutr Rev 2002; 60:S19.
  93. Williams MC, O'Brien WF, Nelson RN, Spellacy WN. Histologic chorioamnionitis is associated with fetal growth restriction in term and preterm infants. Am J Obstet Gynecol 2000; 183:1094.
  94. Schnarr J, Smaill F. Asymptomatic bacteriuria and symptomatic urinary tract infections in pregnancy. Eur J Clin Invest 2008; 38 Suppl 2:50.
  95. Meis PJ, Michielutte R, Peters TJ, et al. Factors associated with preterm birth in Cardiff, Wales. I. Univariable and multivariable analysis. Am J Obstet Gynecol 1995; 173:590.
  96. Meis PJ, Michielutte R, Peters TJ, et al. Factors associated with preterm birth in Cardiff, Wales. II. Indicated and spontaneous preterm birth. Am J Obstet Gynecol 1995; 173:597.
  97. Connolly A, Thorp JM Jr. Urinary tract infections in pregnancy. Urol Clin North Am 1999; 26:779.
  98. Uncu Y, Uncu G, Esmer A, Bilgel N. Should asymptomatic bacteriuria be screened in pregnancy? Clin Exp Obstet Gynecol 2002; 29:281.
  99. Smaill F, Vazquez JC. Antibiotics for asymptomatic bacteriuria in pregnancy. Cochrane Database Syst Rev 2007; :CD000490.
  100. Xiong X, Buekens P, Fraser WD, et al. Periodontal disease and adverse pregnancy outcomes: a systematic review. BJOG 2006; 113:135.
  101. Vergnes JN, Sixou M. Preterm low birth weight and maternal periodontal status: a meta-analysis. Am J Obstet Gynecol 2007; 196:135.e1.
  102. Offenbacher S, Jared HL, O'Reilly PG, et al. Potential pathogenic mechanisms of periodontitis associated pregnancy complications. Ann Periodontol 1998; 3:233.
  103. Newnham JP, Shub A, Jobe AH, et al. The effects of intra-amniotic injection of periodontopathic lipopolysaccharides in sheep. Am J Obstet Gynecol 2005; 193:313.
  104. Han YW, Redline RW, Li M, et al. Fusobacterium nucleatum induces premature and term stillbirths in pregnant mice: implication of oral bacteria in preterm birth. Infect Immun 2004; 72:2272.
  105. Kornman KS, di Giovine FS. Genetic variations in cytokine expression: a risk factor for severity of adult periodontitis. Ann Periodontol 1998; 3:327.
  106. Galbraith GM, Steed RB, Sanders JJ, Pandey JP. Tumor necrosis factor alpha production by oral leukocytes: influence of tumor necrosis factor genotype. J Periodontol 1998; 69:428.
  107. Kornman KS, Crane A, Wang HY, et al. The interleukin-1 genotype as a severity factor in adult periodontal disease. J Clin Periodontol 1997; 24:72.
  108. Boggess KA, Moss K, Madianos P, et al. Fetal immune response to oral pathogens and risk of preterm birth. Am J Obstet Gynecol 2005; 193:1121.
  109. Sanz M, Kornman K, Working group 3 of joint EFP/AAP workshop. Periodontitis and adverse pregnancy outcomes: consensus report of the Joint EFP/AAP Workshop on Periodontitis and Systemic Diseases. J Clin Periodontol 2013; 40 Suppl 14:S164.
  110. Polyzos NP, Polyzos IP, Zavos A, et al. Obstetric outcomes after treatment of periodontal disease during pregnancy: systematic review and meta-analysis. BMJ 2010; 341:c7017.
  111. Uppal A, Uppal S, Pinto A, et al. The effectiveness of periodontal disease treatment during pregnancy in reducing the risk of experiencing preterm birth and low birth weight: a meta-analysis. J Am Dent Assoc 2010; 141:1423.
  112. Chambrone L, Pannuti CM, Guglielmetti MR, Chambrone LA. Evidence grade associating periodontitis with preterm birth and/or low birth weight: II: a systematic review of randomized trials evaluating the effects of periodontal treatment. J Clin Periodontol 2011; 38:902.
  113. Kim AJ, Lo AJ, Pullin DA, et al. Scaling and root planing treatment for periodontitis to reduce preterm birth and low birth weight: a systematic review and meta-analysis of randomized controlled trials. J Periodontol 2012; 83:1508.
  114. Schwendicke F, Karimbux N, Allareddy V, Gluud C. Periodontal treatment for preventing adverse pregnancy outcomes: a meta- and trial sequential analysis. PLoS One 2015; 10:e0129060.
  115. Michalowicz BS, Gustafsson A, Thumbigere-Math V, Buhlin K. The effects of periodontal treatment on pregnancy outcomes. J Clin Periodontol 2013; 40 Suppl 14:S195.
  116. Valkenburg-van den Berg AW, Sprij AJ, Dekker FW, et al. Association between colonization with Group B Streptococcus and preterm delivery: a systematic review. Acta Obstet Gynecol Scand 2009; 88:958.
  117. Martin DH, Koutsky L, Eschenbach DA, et al. Prematurity and perinatal mortality in pregnancies complicated by maternal Chlamydia trachomatis infections. JAMA 1982; 247:1585.
  118. Andrews WW, Goldenberg RL, Mercer B, et al. The Preterm Prediction Study: association of second-trimester genitourinary chlamydia infection with subsequent spontaneous preterm birth. Am J Obstet Gynecol 2000; 183:662.
  119. Ryan GM Jr, Abdella TN, McNeeley SG, et al. Chlamydia trachomatis infection in pregnancy and effect of treatment on outcome. Am J Obstet Gynecol 1990; 162:34.
  120. Cohen I, Veille JC, Calkins BM. Improved pregnancy outcome following successful treatment of chlamydial infection. JAMA 1990; 263:3160.
  121. Hillier SL, Nugent RP, Eschenbach DA, et al. Association between bacterial vaginosis and preterm delivery of a low-birth-weight infant. The Vaginal Infections and Prematurity Study Group. N Engl J Med 1995; 333:1737.
  122. Meis PJ, Goldenberg RL, Mercer B, et al. The preterm prediction study: significance of vaginal infections. National Institute of Child Health and Human Development Maternal-Fetal Medicine Units Network. Am J Obstet Gynecol 1995; 173:1231.
  123. Bretelle F, Rozenberg P, Pascal A, et al. High Atopobium vaginae and Gardnerella vaginalis vaginal loads are associated with preterm birth. Clin Infect Dis 2015; 60:860.
  124. Edwards LE, Barrada MI, Hamann AA, Hakanson EY. Gonorrhea in pregnancy. Am J Obstet Gynecol 1978; 132:637.
  125. Watson-Jones D, Changalucha J, Gumodoka B, et al. Syphilis in pregnancy in Tanzania. I. Impact of maternal syphilis on outcome of pregnancy. J Infect Dis 2002; 186:940.
  126. Van Gerwen OT, Craig-Kuhn MC, Jones AT, et al. Trichomoniasis and adverse birth outcomes: a systematic review and meta-analysis. BJOG 2021; 128:1907.
  127. Sweeney EL, Kallapur SG, Gisslen T, et al. Placental Infection With Ureaplasma species Is Associated With Histologic Chorioamnionitis and Adverse Outcomes in Moderately Preterm and Late-Preterm Infants. J Infect Dis 2016; 213:1340.
  128. Collins S, Ramsay M, Slack MP, et al. Risk of invasive Haemophilus influenzae infection during pregnancy and association with adverse fetal outcomes. JAMA 2014; 311:1125.
  129. Kalliala I, Eriksson T, Aro K, et al. Preterm birth rate after bivalent HPV vaccination: Registry-based follow-up of a randomized clinical trial. Prev Med 2021; 146:106473.
  130. Niyibizi J, Mayrand MH, Audibert F, et al. Association Between Human Papillomavirus Infection Among Pregnant Women and Preterm Birth. JAMA Netw Open 2021; 4:e2125308.
  131. Cunha G, Bastos LB, Freitas SF, et al. Genital mycoplasma infection and spontaneous preterm birth outcome: a prospective cohort study. BJOG 2022; 129:273.
  132. Kataoka S, Yamada T, Chou K, et al. Association between preterm birth and vaginal colonization by mycoplasmas in early pregnancy. J Clin Microbiol 2006; 44:51.
  133. Capoccia R, Greub G, Baud D. Ureaplasma urealyticum, Mycoplasma hominis and adverse pregnancy outcomes. Curr Opin Infect Dis 2013; 26:231.
  134. Lee SE, Romero R, Kim EC, Yoon BH. A high Nugent score but not a positive culture for genital mycoplasmas is a risk factor for spontaneous preterm birth. J Matern Fetal Neonatal Med 2009; 22:212.
  135. Gao R, Liu B, Yang W, et al. Association of Maternal Sexually Transmitted Infections With Risk of Preterm Birth in the United States. JAMA Netw Open 2021; 4:e2133413.
  136. Andrews WW, Klebanoff MA, Thom EA, et al. Midpregnancy genitourinary tract infection with Chlamydia trachomatis: association with subsequent preterm delivery in women with bacterial vaginosis and Trichomonas vaginalis. Am J Obstet Gynecol 2006; 194:493.
  137. Mancuso MS, Figueroa D, Szychowski JM, et al. Midtrimester bacterial vaginosis and cervical length in women at risk for preterm birth. Am J Obstet Gynecol 2011; 204:342.e1.
  138. Schuster HJ, de Jonghe BA, Limpens J, et al. Asymptomatic vaginal Candida colonization and adverse pregnancy outcomes including preterm birth: a systematic review and meta-analysis. Am J Obstet Gynecol MFM 2020; 2:100163.
  139. Serrano MG, Parikh HI, Brooks JP, et al. Racioethnic diversity in the dynamics of the vaginal microbiome during pregnancy. Nat Med 2019; 25:1001.
  140. Fettweis JM, Serrano MG, Brooks JP, et al. The vaginal microbiome and preterm birth. Nat Med 2019; 25:1012.
  141. Elovitz MA, Gajer P, Riis V, et al. Cervicovaginal microbiota and local immune response modulate the risk of spontaneous preterm delivery. Nat Commun 2019; 10:1305.
  142. Callahan BJ, DiGiulio DB, Goltsman DSA, et al. Replication and refinement of a vaginal microbial signature of preterm birth in two racially distinct cohorts of US women. Proc Natl Acad Sci U S A 2017; 114:9966.
  143. Kiss H, Petricevic L, Husslein P. Prospective randomised controlled trial of an infection screening programme to reduce the rate of preterm delivery. BMJ 2004; 329:371.
  144. Klebanoff MA, Carey JC, Hauth JC, et al. Failure of metronidazole to prevent preterm delivery among pregnant women with asymptomatic Trichomonas vaginalis infection. N Engl J Med 2001; 345:487.
  145. Subtil D, Brabant G, Tilloy E, et al. Early clindamycin for bacterial vaginosis in pregnancy (PREMEVA): a multicentre, double-blind, randomised controlled trial. Lancet 2018; 392:2171.
  146. Simcox R, Sin WT, Seed PT, et al. Prophylactic antibiotics for the prevention of preterm birth in women at risk: a meta-analysis. Aust N Z J Obstet Gynaecol 2007; 47:368.
  147. Thinkhamrop J, Hofmeyr GJ, Adetoro O, et al. Antibiotic prophylaxis during the second and third trimester to reduce adverse pregnancy outcomes and morbidity. Cochrane Database Syst Rev 2015; 1:CD002250.
  148. Bellad MB, Hoffman MK, Mallapur AA, et al. Clindamycin to reduce preterm birth in a low resource setting: a randomised placebo-controlled clinical trial. BJOG 2018; 125:1601.
  149. Martin DH, Eschenbach DA, Cotch MF, et al. Double-Blind Placebo-Controlled Treatment Trial of Chlamydia trachomatis Endocervical Infections in Pregnant Women. Infect Dis Obstet Gynecol 1997; 5:10.
  150. Leitich H, Brunbauer M, Bodner-Adler B, et al. Antibiotic treatment of bacterial vaginosis in pregnancy: a meta-analysis. Am J Obstet Gynecol 2003; 188:752.
  151. Carey JC, Klebanoff MA, Hauth JC, et al. Metronidazole to prevent preterm delivery in pregnant women with asymptomatic bacterial vaginosis. National Institute of Child Health and Human Development Network of Maternal-Fetal Medicine Units. N Engl J Med 2000; 342:534.
  152. Brocklehurst P, Hannah M, McDonald H. Interventions for treating bacterial vaginosis in pregnancy. Cochrane Database Syst Rev 2000; :CD000262.
  153. Haahr T, Ersbøll AS, Karlsen MA, et al. Treatment of bacterial vaginosis in pregnancy in order to reduce the risk of spontaneous preterm delivery - a clinical recommendation. Acta Obstet Gynecol Scand 2016; 95:850.
  154. Eschenbach DA, Nugent RP, Rao AV, et al. A randomized placebo-controlled trial of erythromycin for the treatment of Ureaplasma urealyticum to prevent premature delivery. The Vaginal Infections and Prematurity Study Group. Am J Obstet Gynecol 1991; 164:734.
  155. Raynes-Greenow CH, Roberts CL, Bell JC, et al. Antibiotics for ureaplasma in the vagina in pregnancy. Cochrane Database Syst Rev 2004; :CD003767.
  156. Klebanoff MA, Regan JA, Rao AV, et al. Outcome of the Vaginal Infections and Prematurity Study: results of a clinical trial of erythromycin among pregnant women colonized with group B streptococci. Am J Obstet Gynecol 1995; 172:1540.
  157. Koss CA, Baras DC, Lane SD, et al. Investigation of metronidazole use during pregnancy and adverse birth outcomes. Antimicrob Agents Chemother 2012; 56:4800.
  158. Kigozi GG, Brahmbhatt H, Wabwire-Mangen F, et al. Treatment of Trichomonas in pregnancy and adverse outcomes of pregnancy: a subanalysis of a randomized trial in Rakai, Uganda. Am J Obstet Gynecol 2003; 189:1398.
  159. Mann JR, McDermott S, Zhou L, et al. Treatment of trichomoniasis in pregnancy and preterm birth: An observational study. J Womens Health (Larchmt) 2009; 18:493.
  160. Gülmezoglu AM, Azhar M. Interventions for trichomoniasis in pregnancy. Cochrane Database Syst Rev 2011; :CD000220.
  161. Stringer E, Read JS, Hoffman I, et al. Treatment of trichomoniasis in pregnancy in sub-Saharan Africa does not appear to be associated with low birth weight or preterm birth. S Afr Med J 2010; 100:58.
  162. Aidoo M, McElroy PD, Kolczak MS, et al. Tumor necrosis factor-alpha promoter variant 2 (TNF2) is associated with pre-term delivery, infant mortality, and malaria morbidity in western Kenya: Asembo Bay Cohort Project IX. Genet Epidemiol 2001; 21:201.
  163. Luntamo M, Kulmala T, Mbewe B, et al. Effect of repeated treatment of pregnant women with sulfadoxine-pyrimethamine and azithromycin on preterm delivery in Malawi: a randomized controlled trial. Am J Trop Med Hyg 2010; 83:1212.
  164. Wolfe EB, Parise ME, Haddix AC, et al. Cost-effectiveness of sulfadoxine-pyrimethamine for the prevention of malaria-associated low birth weight. Am J Trop Med Hyg 2001; 64:178.
  165. Garner P, Gülmezoglu AM. Drugs for preventing malaria-related illness in pregnant women and death in the newborn. Cochrane Database Syst Rev 2003; :CD000169.
  166. Wong LF, Wilkes J, Korgenski K, et al. Risk factors associated with preterm birth after a prior term delivery. BJOG 2016; 123:1772.
  167. Rodrigues T, Barros H. Short interpregnancy interval and risk of spontaneous preterm delivery. Eur J Obstet Gynecol Reprod Biol 2008; 136:184.
  168. Rodriguez MI, Chang R, Thiel de Bocanegra H. The impact of postpartum contraception on reducing preterm birth: findings from California. Am J Obstet Gynecol 2015; 213:703.e1.
  169. Conde-Agudelo A, Rosas-Bermúdez A, Kafury-Goeta AC. Birth spacing and risk of adverse perinatal outcomes: a meta-analysis. JAMA 2006; 295:1809.
  170. Mozurkewich EL, Luke B, Avni M, Wolf FM. Working conditions and adverse pregnancy outcome: a meta-analysis. Obstet Gynecol 2000; 95:623.
  171. van Beukering MD, van Melick MJ, Mol BW, et al. Physically demanding work and preterm delivery: a systematic review and meta-analysis. Int Arch Occup Environ Health 2014; 87:809.
  172. Cai C, Vandermeer B, Khurana R, et al. The impact of occupational shift work and working hours during pregnancy on health outcomes: a systematic review and meta-analysis. Am J Obstet Gynecol 2019; 221:563.
  173. Cai C, Vandermeer B, Khurana R, et al. The impact of occupational activities during pregnancy on pregnancy outcomes: a systematic review and metaanalysis. Am J Obstet Gynecol 2020; 222:224.
  174. ACOG Committee Opinion No. 733: Employment Considerations During Pregnancy and the Postpartum Period. Obstet Gynecol 2018; 131:e115.
  175. https://www.eeoc.gov/eeoc/publications/pregnant_workers.cfm (Accessed on March 03, 2020).
  176. https://www.hse.gov.uk/mothers/law.htm (Accessed on March 03, 2020).
  177. https://handbookgermany.de/en/live/pregnancy.html (Accessed on March 03, 2020).
  178. Berkowitz GS, Papiernik E. Working conditions, maternity legislation, and preterm birth. Semin Perinatol 1995; 19:272.
  179. Di Mascio D, Magro-Malosso ER, Saccone G, et al. Exercise during pregnancy in normal-weight women and risk of preterm birth: a systematic review and meta-analysis of randomized controlled trials. Am J Obstet Gynecol 2016; 215:561.
  180. Aune D, Schlesinger S, Henriksen T, et al. Physical activity and the risk of preterm birth: a systematic review and meta-analysis of epidemiological studies. BJOG 2017; 124:1816.
  181. Armson BA. Physical activity and preterm birth: risk factor or benefit? BJOG 2017; 124:1827.
  182. Berghella V, Klebanoff M, McPherson C, et al. Sexual intercourse association with asymptomatic bacterial vaginosis and Trichomonas vaginalis treatment in relationship to preterm birth. Am J Obstet Gynecol 2002; 187:1277.
  183. Read JS, Klebanoff MA. Sexual intercourse during pregnancy and preterm delivery: effects of vaginal microorganisms. The Vaginal Infections and Prematurity Study Group. Am J Obstet Gynecol 1993; 168:514.
  184. Mills JL, Harlap S, Harley EE. Should coitus late in pregnancy be discouraged? Lancet 1981; 2:136.
  185. Yost NP, Owen J, Berghella V, et al. Effect of coitus on recurrent preterm birth. Obstet Gynecol 2006; 107:793.
  186. SIMPSON WJ. A preliminary report on cigarette smoking and the incidence of prematurity. Am J Obstet Gynecol 1957; 73:807.
  187. Meyer MB, Tonascia JA. Maternal smoking, pregnancy complications, and perinatal mortality. Am J Obstet Gynecol 1977; 128:494.
  188. Shiono PH, Klebanoff MA, Rhoads GG. Smoking and drinking during pregnancy. Their effects on preterm birth. JAMA 1986; 255:82.
  189. Berkowitz GS, Papiernik E. Epidemiology of preterm birth. Epidemiol Rev 1993; 15:414.
  190. Cnattingius S, Forman MR, Berendes HW, et al. Effect of age, parity, and smoking on pregnancy outcome: a population-based study. Am J Obstet Gynecol 1993; 168:16.
  191. Harlow BL, Frigoletto FD, Cramer DW, et al. Determinants of preterm delivery in low-risk pregnancies. The RADIUS Study Group. J Clin Epidemiol 1996; 49:441.
  192. Kyrklund-Blomberg NB, Cnattingius S. Preterm birth and maternal smoking: risks related to gestational age and onset of delivery. Am J Obstet Gynecol 1998; 179:1051.
  193. http://www.cdc.gov/mmwr/volumes/65/wr/mm6532a4.htm?s_cid=mm6532a4_e.
  194. Nicholson W, Croughan-Minihane M, Posner S, et al. Preterm delivery in patients admitted with preterm labor: a prediction study. J Matern Fetal Med 2001; 10:102.
  195. Ney JA, Dooley SL, Keith LG, et al. The prevalence of substance abuse in patients with suspected preterm labor. Am J Obstet Gynecol 1990; 162:1562.
  196. Spence MR, Williams R, DiGregorio GJ, et al. The relationship between recent cocaine use and pregnancy outcome. Obstet Gynecol 1991; 78:326.
  197. Borges G, Lopez-Cervantes M, Medina-Mora ME, et al. Alcohol consumption, low birth weight, and preterm delivery in the National Addiction Survey (Mexico). Int J Addict 1993; 28:355.
  198. Wilkins-Haug L, Gabow PA. Toluene abuse during pregnancy: obstetric complications and perinatal outcomes. Obstet Gynecol 1991; 77:504.
  199. Ikehara S, Kimura T, Kakigano A, et al. Association between maternal alcohol consumption during pregnancy and risk of preterm delivery: the Japan Environment and Children's Study. BJOG 2019; 126:1448.
  200. Boer K, Smit BJ, van Huis AM, Hogerzeil HV. Substance use in pregnancy: do we care? Acta Paediatr Suppl 1994; 404:65.
  201. Almario CV, Seligman NS, Dysart KC, et al. Risk factors for preterm birth among opiate-addicted gravid women in a methadone treatment program. Am J Obstet Gynecol 2009; 201:326.e1.
  202. Villar J, Gülmezoglu AM, de Onis M. Nutritional and antimicrobial interventions to prevent preterm birth: an overview of randomized controlled trials. Obstet Gynecol Surv 1998; 53:575.
  203. Bloomfield FH, Oliver MH, Hawkins P, et al. A periconceptional nutritional origin for noninfectious preterm birth. Science 2003; 300:606.
  204. Kumarasamy V, Mitchell MD, Bloomfield FH, et al. Effects of periconceptional undernutrition on the initiation of parturition in sheep. Am J Physiol Regul Integr Comp Physiol 2005; 288:R67.
  205. Rayco-Solon P, Fulford AJ, Prentice AM. Maternal preconceptional weight and gestational length. Am J Obstet Gynecol 2005; 192:1133.
  206. Stein Z, Susser M. The Dutch famine, 1944-1945, and the reproductive process. I. Effects on six indices at birth. Pediatr Res 1975; 9:70.
  207. Kramer MS. Isocaloric balanced protein supplementation in pregnancy. Cochrane Database Syst Rev 2000; :CD000118.
  208. Ota E, Tobe-Gai R, Mori R, Farrar D. Antenatal dietary advice and supplementation to increase energy and protein intake. Cochrane Database Syst Rev 2012; :CD000032.
  209. Vahratian A, Siega-Riz AM, Savitz DA, Thorp JM Jr. Multivitamin use and the risk of preterm birth. Am J Epidemiol 2004; 160:886.
  210. Fawzi WW, Msamanga GI, Urassa W, et al. Vitamins and perinatal outcomes among HIV-negative women in Tanzania. N Engl J Med 2007; 356:1423.
  211. Spinnato JA 2nd, Freire S, Pinto e Silva JL, et al. Antioxidant supplementation and premature rupture of the membranes: a planned secondary analysis. Am J Obstet Gynecol 2008; 199:433.e1.
  212. Rumbold A, Ota E, Nagata C, et al. Vitamin C supplementation in pregnancy. Cochrane Database Syst Rev 2015; :CD004072.
  213. Rumbold A, Ota E, Hori H, et al. Vitamin E supplementation in pregnancy. Cochrane Database Syst Rev 2015; :CD004069.
  214. Saccone G, Berghella V. Folic acid supplementation in pregnancy to prevent preterm birth: a systematic review and meta-analysis of randomized controlled trials. Eur J Obstet Gynecol Reprod Biol 2016; 199:76.
  215. Pérez-López FR, Pasupuleti V, Mezones-Holguin E, et al. Effect of vitamin D supplementation during pregnancy on maternal and neonatal outcomes: a systematic review and meta-analysis of randomized controlled trials. Fertil Steril 2015; 103:1278.
  216. Catov JM, Bodnar LM, Ness RB, et al. Association of periconceptional multivitamin use and risk of preterm or small-for-gestational-age births. Am J Epidemiol 2007; 166:296.
  217. Middleton P, Gomersall JC, Gould JF, et al. Omega-3 fatty acid addition during pregnancy. Cochrane Database Syst Rev 2018; 11:CD003402.
  218. Han Z, Mulla S, Beyene J, et al. Maternal underweight and the risk of preterm birth and low birth weight: a systematic review and meta-analyses. Int J Epidemiol 2011; 40:65.
  219. McDonald SD, Han Z, Mulla S, et al. Overweight and obesity in mothers and risk of preterm birth and low birth weight infants: systematic review and meta-analyses. BMJ 2010; 341:c3428.
  220. Cnattingius S, Villamor E, Johansson S, et al. Maternal obesity and risk of preterm delivery. JAMA 2013; 309:2362.
  221. Girsen AI, Mayo JA, Carmichael SL, et al. Women's prepregnancy underweight as a risk factor for preterm birth: a retrospective study. BJOG 2016; 123:2001.
  222. Salmon C, Thibon P, Prime L, et al. Impact of maternal underweight on obstetric and neonatal prognosis: A retrospective study. Eur J Obstet Gynecol Reprod Biol 2021; 260:6.
  223. Honest H, Bachmann LM, Ngai C, et al. The accuracy of maternal anthropometry measurements as predictor for spontaneous preterm birth--a systematic review. Eur J Obstet Gynecol Reprod Biol 2005; 119:11.
  224. Carmichael SL, Abrams B. A critical review of the relationship between gestational weight gain and preterm delivery. Obstet Gynecol 1997; 89:865.
  225. Dietz PM, Callaghan WM, Cogswell ME, et al. Combined effects of prepregnancy body mass index and weight gain during pregnancy on the risk of preterm delivery. Epidemiology 2006; 17:170.
  226. Schieve LA, Cogswell ME, Scanlon KS, et al. Prepregnancy body mass index and pregnancy weight gain: associations with preterm delivery. The NMIHS Collaborative Study Group. Obstet Gynecol 2000; 96:194.
  227. Thangaratinam S, Rogozinska E, Jolly K, et al. Effects of interventions in pregnancy on maternal weight and obstetric outcomes: meta-analysis of randomised evidence. BMJ 2012; 344:e2088.
  228. Myklestad K, Vatten LJ, Magnussen EB, et al. Do parental heights influence pregnancy length?: A population-based prospective study, HUNT 2. BMC Pregnancy Childbirth 2013; 13:33.
  229. Derraik JG, Lundgren M, Cutfield WS, Ahlsson F. Maternal Height and Preterm Birth: A Study on 192,432 Swedish Women. PLoS One 2016; 11:e0154304.
  230. Han Z, Lutsiv O, Mulla S, McDonald SD. Maternal height and the risk of preterm birth and low birth weight: a systematic review and meta-analyses. J Obstet Gynaecol Can 2012; 34:721.
  231. http://www.cdc.gov/mmwr/preview/mmwrhtml/mm6409a3.htm?s_cid=mm6409a3_e.
  232. Lockwood CJ. Stress-associated preterm delivery: the role of corticotropin-releasing hormone. Am J Obstet Gynecol 1999; 180:S264.
  233. Petraglia F, Hatch MC, Lapinski R, et al. Lack of effect of psychosocial stress on maternal corticotropin-releasing factor and catecholamine levels at 28 weeks' gestation. J Soc Gynecol Investig 2001; 8:83.
  234. Lu MC, Chen B. Racial and ethnic disparities in preterm birth: the role of stressful life events. Am J Obstet Gynecol 2004; 191:691.
  235. Kramer MS, Lydon J, Séguin L, et al. Stress pathways to spontaneous preterm birth: the role of stressors, psychological distress, and stress hormones. Am J Epidemiol 2009; 169:1319.
  236. Lima SAM, El Dib RP, Rodrigues MRK, et al. Is the risk of low birth weight or preterm labor greater when maternal stress is experienced during pregnancy? A systematic review and meta-analysis of cohort studies. PLoS One 2018; 13:e0200594.
  237. Chen MJ, Grobman WA, Gollan JK, Borders AE. The use of psychosocial stress scales in preterm birth research. Am J Obstet Gynecol 2011; 205:402.
  238. Hodnett ED, Fredericks S. Support during pregnancy for women at increased risk of low birthweight babies. Cochrane Database Syst Rev 2003; :CD000198.
  239. Khianman B, Pattanittum P, Thinkhamrop J, Lumbiganon P. Relaxation therapy for preventing and treating preterm labour. Cochrane Database Syst Rev 2012; :CD007426.
  240. Li X, Huang S, Jiao A, et al. Association between ambient fine particulate matter and preterm birth or term low birth weight: An updated systematic review and meta-analysis. Environ Pollut 2017; 227:596.
  241. Sun X, Luo X, Zhao C, et al. The association between fine particulate matter exposure during pregnancy and preterm birth: a meta-analysis. BMC Pregnancy Childbirth 2015; 15:300.
  242. Melody SM, Ford J, Wills K, et al. Maternal exposure to short-to medium-term outdoor air pollution and obstetric and neonatal outcomes: A systematic review. Environ Pollut 2019; 244:915.
  243. Yuan L, Zhang Y, Gao Y, Tian Y. Maternal fine particulate matter (PM2.5) exposure and adverse birth outcomes: an updated systematic review based on cohort studies. Environ Sci Pollut Res Int 2019; 26:13963.
  244. Porpora MG, Piacenti I, Scaramuzzino S, et al. Environmental Contaminants Exposure and Preterm Birth: A Systematic Review. Toxics 2019; 7.
  245. Bekkar B, Pacheco S, Basu R, DeNicola N. Association of Air Pollution and Heat Exposure With Preterm Birth, Low Birth Weight, and Stillbirth in the US: A Systematic Review. JAMA Netw Open 2020; 3:e208243.
  246. Chersich MF, Pham MD, Areal A, et al. Associations between high temperatures in pregnancy and risk of preterm birth, low birth weight, and stillbirths: systematic review and meta-analysis. BMJ 2020; 371:m3811.
  247. Radke EG, Glenn BS, Braun JM, Cooper GS. Phthalate exposure and female reproductive and developmental outcomes: a systematic review of the human epidemiological evidence. Environ Int 2019; 130:104580.
  248. Whitworth M, Quenby S, Cockerill RO, Dowswell T. Specialised antenatal clinics for women with a pregnancy at high risk of preterm birth (excluding multiple pregnancy) to improve maternal and infant outcomes. Cochrane Database Syst Rev 2011; :CD006760.
  249. Zeitlin J, Saurel-Cubizolles MJ, De Mouzon J, et al. Fetal sex and preterm birth: are males at greater risk? Hum Reprod 2002; 17:2762.
  250. Ingemarsson I. Gender aspects of preterm birth. BJOG 2003; 110 Suppl 20:34.
  251. Zeitlin J, Ancel PY, Larroque B, et al. Fetal sex and indicated very preterm birth: results of the EPIPAGE study. Am J Obstet Gynecol 2004; 190:1322.
  252. McGregor JA, Leff M, Orleans M, Baron A. Fetal gender differences in preterm birth: findings in a North American cohort. Am J Perinatol 1992; 9:43.
  253. Dolan SM, Gross SJ, Merkatz IR, et al. The contribution of birth defects to preterm birth and low birth weight. Obstet Gynecol 2007; 110:318.
  254. Purisch SE, DeFranco EA, Muglia LJ, et al. Preterm birth in pregnancies complicated by major congenital malformations: a population-based study. Am J Obstet Gynecol 2008; 199:287.e1.
  255. Lackman F, Capewell V, Richardson B, et al. The risks of spontaneous preterm delivery and perinatal mortality in relation to size at birth according to fetal versus neonatal growth standards. Am J Obstet Gynecol 2001; 184:946.
  256. Morken NH, Källen K, Jacobsson B. Fetal growth and onset of delivery: a nationwide population-based study of preterm infants. Am J Obstet Gynecol 2006; 195:154.
  257. Ott WJ. Intrauterine growth retardation and preterm delivery. Am J Obstet Gynecol 1993; 168:1710.
  258. Zeitlin J, Ancel PY, Saurel-Cubizolles MJ, Papiernik E. The relationship between intrauterine growth restriction and preterm delivery: an empirical approach using data from a European case-control study. BJOG 2000; 107:750.
  259. Gardosi JO. Prematurity and fetal growth restriction. Early Hum Dev 2005; 81:43.
  260. Wadhwa PD, Garite TJ, Porto M, et al. Placental corticotropin-releasing hormone (CRH), spontaneous preterm birth, and fetal growth restriction: a prospective investigation. Am J Obstet Gynecol 2004; 191:1063.
  261. Basso O, Olsen J, Christensen K. Low birthweight and prematurity in relation to paternal factors: a study of recurrence. Int J Epidemiol 1999; 28:695.
  262. Boyd HA, Poulsen G, Wohlfahrt J, et al. Maternal contributions to preterm delivery. Am J Epidemiol 2009; 170:1358.
  263. Meertens LJE, van Montfort P, Scheepers HCJ, et al. Prediction models for the risk of spontaneous preterm birth based on maternal characteristics: a systematic review and independent external validation. Acta Obstet Gynecol Scand 2018; 97:907.
  264. Honest H, Bachmann LM, Sundaram R, et al. The accuracy of risk scores in predicting preterm birth--a systematic review. J Obstet Gynaecol 2004; 24:343.
  265. Hezelgrave NL, Abbott DS, Radford SK, et al. Quantitative Fetal Fibronectin at 18 Weeks of Gestation to Predict Preterm Birth in Asymptomatic High-Risk Women. Obstet Gynecol 2016; 127:255.
  266. Kuhrt K, Smout E, Hezelgrave N, et al. Development and validation of a tool incorporating cervical length and quantitative fetal fibronectin to predict spontaneous preterm birth in asymptomatic high-risk women. Ultrasound Obstet Gynecol 2016; 47:104.
  267. www.QUiPP.org.
  268. Watson HA, Carlisle N, Seed PT, et al. Evaluating the use of the QUiPP app and its impact on the management of threatened preterm labour: A cluster randomised trial. PLoS Med 2021; 18:e1003689.
  269. Esplin MS, Elovitz MA, Iams JD, et al. Predictive Accuracy of Serial Transvaginal Cervical Lengths and Quantitative Vaginal Fetal Fibronectin Levels for Spontaneous Preterm Birth Among Nulliparous Women. JAMA 2017; 317:1047.
  270. Saade GR, Boggess KA, Sullivan SA, et al. Development and validation of a spontaneous preterm delivery predictor in asymptomatic women. Am J Obstet Gynecol 2016; 214:633.e1.
  271. Conde-Agudelo A, Papageorghiou AT, Kennedy SH, Villar J. Novel biomarkers for the prediction of the spontaneous preterm birth phenotype: a systematic review and meta-analysis. BJOG 2011; 118:1042.
  272. Heida KY, Velthuis BK, Oudijk MA, et al. Cardiovascular disease risk in women with a history of spontaneous preterm delivery: A systematic review and meta-analysis. Eur J Prev Cardiol 2016; 23:253.
  273. Crump C, Sundquist J, Howell EA, et al. Pre-Term Delivery and Risk of Ischemic Heart Disease in Women. J Am Coll Cardiol 2020; 76:57.
  274. Crump C, Sundquist J, Sundquist K. Preterm delivery and long term mortality in women: national cohort and co-sibling study. BMJ 2020; 370:m2533.
Topic 6761 Version 131.0

References