Your activity: 20 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Fertility, pregnancy, and nursing in inflammatory bowel disease

Fertility, pregnancy, and nursing in inflammatory bowel disease
Authors:
Mark A Peppercorn, MD
Uma Mahadevan, MD
Section Editors:
Charles J Lockwood, MD, MHCM
Sunanda V Kane, MD, MSPH
Deputy Editor:
Kristen M Robson, MD, MBA, FACG
Literature review current through: Dec 2022. | This topic last updated: Nov 21, 2022.

INTRODUCTION — Inflammatory bowel disease (IBD) can affect patients in their childbearing years and therefore has implications on fertility, pregnancy, and nursing.

This topic will review issues related to fertility in patients with IBD and the diagnosis and management of IBD in pregnant and nursing patients. The medical management of IBD in the general population is discussed in detail separately:

(See "Medical management of low-risk adult patients with mild to moderate ulcerative colitis".)

(See "Management of moderate to severe ulcerative colitis in adults".)

(See "Management of the hospitalized adult patient with severe ulcerative colitis".)

(See "Overview of the medical management of mild (low risk) Crohn disease in adults".)

(See "Overview of medical management of high-risk, adult patients with moderate to severe Crohn disease".)

Our approach is consistent with a multidisciplinary guide to preconception counseling and caring for pregnant patients with IBD [1].

FERTILITY — Patients with quiescent IBD do not have decreased fertility as compared with the age-matched general population [2-6]. Low birth rates among patients with IBD are often due to patient choice rather than disease-related infertility [2,5]. (See "Overview of infertility".)

However, IBD can cause infertility in the following circumstances:

Active inflammation in patients with Crohn disease may result in infertility due to inflammation involving the fallopian tubes and ovaries [6]. Perianal disease can also cause dyspareunia. (See "Causes of female infertility", section on 'Fallopian tube abnormalities/pelvic adhesions'.)

Medications may decrease fertility in male patients. As examples, methotrexate and sulfasalazine can cause reversible oligospermia. In addition, sulfasalazine is associated with reduced sperm motility and abnormal sperm morphology [7-10].

However, studies suggest that anti-tumor necrosis factor (anti-TNF) therapy does not impair sperm quality [11].

Surgery may impact fertility in patients with IBD. Proctocolectomy in males may lead to impotence or ejaculatory difficulties [12]. Restorative proctocolectomy with ileoanal anastomosis has been associated with dyspareunia and a reduction in fertility due to scarring and formation of adnexal adhesions [3,13,14]. (See "Surgical management of Crohn disease", section on 'Surgical approaches' and "Surgical management of ulcerative colitis", section on 'Surgical options'.)

Female IBD patients with infertility may seek assisted reproductive technology (ART) treatment. A cohort study based on Danish health registries evaluated the chance of live birth in patients with IBD who underwent ART treatments [15]. Patients with ulcerative colitis had lower rates of successful pregnancy compared with patients without IBD (odds ratio [OR] 0.73, 95% CI 0.58-0.92). Patients who had surgery for Crohn disease were also less likely to have live birth following ART (adjusted OR 0.51, 95% CI 0.29-0.91).

PRECONCEPTION COUNSELING — Preconception counseling offers an opportunity for the clinician to address specific patient concerns regarding the risk of transmission of IBD or other diseases to their offspring, to optimize control of disease activity and nutritional status, avoid inappropriate medication cessation, and discontinue medications that may adversely affect pregnancy. (See "The preconception office visit".)

IBD risk in offspring — Although IBD follows a non-Mendelian pattern of inheritance, genetically determined factors contribute to IBD susceptibility. First-degree relatives of patients with IBD are approximately 3 to 20 times more likely to develop IBD as compared with the general population [16-19]. In a large national, registry-based cohort study with a median follow-up of 10 years, the risk of developing IBD was increased in offspring born to mothers with ulcerative colitis (HR 4.6, 95% CI 3.5-6.2) and mothers with Crohn disease (HR 7.7, 95% CI 5.7-10.5) compared with mothers without IBD; the impact of paternal IBD was not evaluated [20].

Clinical features of the disease also demonstrate a heritable pattern with concordance in disease location (eg, ileal versus colonic Crohn disease) and type (eg, fibrostenotic Crohn disease, fistulas) [21-26]. Subsequent generations may demonstrate "genetic anticipation" with the development of earlier onset of IBD and more severe disease as compared with their parents [19,27].

The role of genetic factors that increase susceptibility to IBD and genetic syndromes associated with IBD is discussed in detail separately. (See "Genetic factors in inflammatory bowel disease".)

Disease control — Female patients with IBD should attempt conception at a time when the disease is in remission. Patients who have active disease at conception are more likely to have active disease during pregnancy [28]. Data have suggested that patients with ulcerative colitis are more likely to have disease activity during pregnancy than patients with Crohn disease [29]. This may reflect the cytokine production of the placenta and its impact on ulcerative colitis. Having active disease is associated with a significant increase in the rate of preterm birth [30]. The risk of disease activity in the untreated patient should be discussed alongside the potential risk of any medications used during pregnancy to maintain remission. (See "Preterm birth: Definitions of prematurity, epidemiology, and risk factors for infant mortality".)

Medications — Medications that may affect fertility or adversely affect pregnancy should be discontinued, when possible, for patients planning pregnancy. The risks and benefits of fetal drug exposure versus discontinuation of medications and its attendant risk of flare should be specifically discussed. These risks and benefits are discussed in detail below. (See 'Medications during pregnancy and lactation' below.)

Nonimmunosuppressive agents – Sulfasalazine causes reversible oligospermia that resolves within discontinuation of therapy. Oligospermia is not associated with 5-aminosalicylic acid (5-ASA) medications (eg, mesalamine). Male patients on sulfasalazine should therefore be transitioned to 5-ASAs four months prior to conception (table 1) [31,32]. (See 'Sulfasalazine and 5-aminosalicylic acid' below.)

Immunosuppressive agents – In female patients, methotrexate is contraindicated because it is an abortifacient and potent teratogen. Methotrexate is associated with oligospermia but has not been associated with adverse fetal outcomes related to paternal exposure [33-35]. Because of the risk of oligospermia, male patients should discontinue methotrexate and use contraception for at least four months prior to conception. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Methotrexate' and 'Medications during pregnancy and lactation' below.)

Data on paternal exposure to immunomodulators and/or biologic agents around the time of conception have suggested that such agents were not associated with increased risk of adverse birth outcomes [36,37]. In a database study including 7453 males with immune-mediated inflammatory diseases (eg, inflammatory bowel disease, psoriasis/psoriatic arthritis) whose partners had conceived, exposure to thiopurines (relative risk [RR], 1.12; 95% CI 0.66-1.76), methotrexate (RR, 0.67; 95% CI 0.21-1.55), anti-tumor necrosis factor (anti-TNF) agents (RR, 1.14; 95% CI 0.81-1.57), or non-TNF targeting biologic agents (eg, vedolizumab, ustekinumab) (RR, 1.75; 95% CI 0.80-3.24) was not associated with increased risk of major congenital anomalies, preterm birth or low birth weight [37]. In a prior meta-analysis, periconception exposure to thiopurines in male patients was not associated with congenital anomalies [36]. These findings lend support for the safety of these biologics and immunomodulators at the time of conception for male patients.

We do not routinely recommend that male patients on thiopurines discontinue treatment unless the couple has a history of infertility or miscarriages that is otherwise unexplained. In such cases, if analysis of male factor infertility suggests abnormal semen quality, and no other factor is identified, a trial of thiopurine discontinuation can be considered. (See 'Azathioprine and mercaptopurine' below and "Causes of male infertility".)

Data on the effect of thiopurines on female fertility are lacking.

Cyclosporine does not appear to reduce male fertility or to have a demonstrable effect on sperm [38]. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Cyclosporine' and 'Cyclosporine' below.)

Nutrition and supplements — Consultation with a nutritionist should be considered to offer advice on eating a well-balanced, healthy diet. We obtain nutrition consultation for patients with risk factors for reduced gestational weight gain such as those with active disease and surgical changes impacting absorption. [39]. Folic acid is important for neural tube development during pregnancy. Folate supplementation (2 mg daily) should be recommended in patients with IBD on low residue diets, with ileal involvement, and patients on medications that interfere with folic acid metabolism (eg, sulfasalazine) [6,40]. (See "Folic acid supplementation in pregnancy".)

Patients with IBD are at risk for iron and vitamin B12 deficiency. Furthermore, iron requirements increase during pregnancy. Iron and B12 levels should therefore be checked in the first trimester and supplementation should be provided as needed. (See "Vitamin and mineral deficiencies in inflammatory bowel disease" and "Nutrition in pregnancy: Dietary requirements and supplements".)

PREGNANCY AND LACTATION — Patients with IBD are at an increased risk for worse obstetric and medical outcomes compared with the age-matched general population [41-43].

Effect of pregnancy on IBD — For patients with quiescent IBD at conception, the course of IBD is approximately the same as in nonpregnant patients [44]. Patients with Crohn disease have a similar disease course during pregnancy and the postpartum as nonpregnant patients with IBD. However, for patients with ulcerative colitis, there is an increased risk of disease flare compared with a patient with Crohn disease. The reason for this is unclear and may be associated with smoking cessation, cytokines secreted by the placenta or undertreatment of ulcerative colitis patients prior to pregnancy [29].

Approximately one-third of patients with quiescent disease at conception relapse during the pregnancy, similar to the general IBD population [45]. Relapses are more common during the first trimester. However, remission achieved during pregnancy is likely to be sustained throughout the remainder of the pregnancy.

In contrast, approximately 70 percent of patients with active disease at conception are likely to have continued or worsening symptoms during pregnancy [46,47]. (See "Clinical manifestations, diagnosis, and prognosis of ulcerative colitis in adults", section on 'Acute complications'.)

The events occurring during one pregnancy do not correlate with the course in subsequent pregnancies [48]. However, pregnancy may be protective in the long term, as suggested by lower relapse rates up to ten years following pregnancy compared with rates prior to pregnancy [49,50]. As parity increases, the risk for surgery to treat IBD also decreases, though this may simply be because healthier patients are more likely to have multiple pregnancies [49].

Effect of IBD on pregnancy — Pregnant patients with IBD may be at increased risk for antepartum hemorrhage, low birth weight infants, and premature delivery [41,43,51-54]. However, the risk of congenital abnormalities does not appear to be increased [41,55,56].

The degree of disease activity may account, in part, for the poor pregnancy-related outcomes [55,57]. The risk of poor pregnancy outcomes in patients with IBD is greatest in those who have active disease at the time of conception, and in whom remission may be difficult to achieve during pregnancy [4,55,57-61]. As an example, in a Danish cohort study, the risk of preterm birth increased if the mother was hospitalized during pregnancy (odds ratio [OR] 1.4), and if the first hospitalization for ulcerative colitis occurred during pregnancy (OR 3.4) [55].

Patients with IBD should be followed as high-risk obstetric patients, particularly in the third trimester, and if the disease is active. (See "Overview of antepartum fetal assessment" and "Preterm birth: Risk factors, interventions for risk reduction, and maternal prognosis", section on 'Chronic medical disorders'.)

Effect of lactation on IBD — Breastfeeding does not independently affect the course of disease in IBD after adjusting for an increase in disease activity due to cessation of IBD medications. However, data suggest that over 50 percent of female patients with IBD do not breastfeed their children either because of a clinician recommendation, fear of medication interactions, or personal choice [62]. It is therefore important to discuss the benefits of breastfeeding as well as the compatibility of several anti-inflammatory and immunosuppressive medications with breastfeeding as discussed below. (See "Infant benefits of breastfeeding" and 'Medications during pregnancy and lactation' below and "Breastfeeding: Parental education and support".)

EVALUATING DISEASE ACTIVITY DURING PREGNANCY AND LACTATION

Laboratory studies — Fecal calprotectin is a stool marker of intestinal inflammation that can be measured in pregnant patients to assess disease activity [63-67]. In a cohort study including 85 pregnant patients with IBD, a fecal calprotectin level ≥250 mcg/g during the second trimester was associated with higher rates of low birth weight (35 versus 4 percent) [66]. (See "Clinical manifestations, diagnosis, and prognosis of Crohn disease in adults", section on 'Stool studies'.)

Endoscopy and imaging studies — Endoscopy or imaging studies may be indicated during pregnancy for the diagnosis of suspected IBD or for the assessment of disease severity, or diagnosis and management of complications in patients with established IBD. However, it is important to consider the risks, benefits, and optimal timing of such a diagnostic evaluation in pregnant or nursing patients. (See "Clinical manifestations, diagnosis, and prognosis of ulcerative colitis in adults", section on 'Diagnosis' and "Clinical manifestations, diagnosis, and prognosis of Crohn disease in adults", section on 'Diagnostic evaluation' and "Management of the hospitalized adult patient with severe ulcerative colitis", section on 'Pretreatment evaluation'.)

The safety and efficacy of endoscopic procedures during pregnancy have not been extensively studied. Endoscopy should be performed during pregnancy only if there is a strong indication (eg, significant bleeding) or if the information is critical for making treatment decisions (eg, establish the diagnosis of IBD in a pregnant patient with chronic diarrhea or stage disease if new therapies are needed). Flexible sigmoidoscopy, which can be performed without sedation or colonic preparation, is low risk in pregnancy in any trimester [68]. While there are several reports of colonoscopies being performed during pregnancy without complications, experience is relatively limited compared with sigmoidoscopy [68,69]. Obstetrical staff should be closely involved and the degree of maternal and fetal monitoring should be individualized. Recommendations for procedural sedation for endoscopy in pregnant and nursing patients are discussed in detail separately. (See "Gastrointestinal endoscopy in adults: Procedural sedation administered by endoscopists", section on 'Special populations'.)

Radiographic studies involving ionizing radiation (eg, computed tomography enterography, abdominal radiographs, small bowel follow-through) should be avoided in pregnancy if possible. These studies can be performed if they are necessary for maternal management and if there is no alternative imaging modality available. Several techniques can be used to minimize fetal radiation exposure. Magnetic resonance (MR) enterography is the preferred diagnostic modality in the absence of institutional expertise with ultrasound for IBD because it avoids the ionizing radiation of computed tomography. While MR enterography can be done in any trimester, intravenous gadolinium crosses the placenta [70]. The theoretical risk to the patient needs to be weighed against the additional imaging benefit of using gadolinium. However, oral contrast is routinely used. (See "Diagnostic imaging in pregnant and nursing patients".)

MEDICATIONS DURING PREGNANCY AND LACTATION — The choice of anti-inflammatory and immunosuppressive use during pregnancy and lactation should be based upon their relative safety and indications as well as the risk of relapse of IBD if the medications are discontinued.

Active IBD itself is associated with poor pregnancy outcomes, and therefore the risks associated with discontinuing some medications (eg, azathioprine, anti-tumor necrosis factor [anti-TNF] agents) are higher than the known risks of the medications themselves. (See "Safety of rheumatic disease medication use during pregnancy and lactation".)

The recommendations on lactation are consistent with the Drugs and Lactation Database of the US National Library of Medicine (LactMed) unless noted otherwise. LactMed is a free, online resource for information on maternal and infant levels of drugs, possible effects on breastfed infants and on lactation.

Antibiotics — Antibiotics, metronidazole and ciprofloxacin, are frequently used in the management of patients with inflammatory bowel disease. (See "Overview of the medical management of mild (low risk) Crohn disease in adults", section on 'Antibiotics' and "Overview of medical management of high-risk, adult patients with moderate to severe Crohn disease", section on 'The acutely ill patient with Crohn disease'.)

The use of metronidazole should be limited to short courses. A number of studies have suggested that metronidazole is not associated with an increased risk of congenital anomaly or cancer [71,72]. However, long-term use in pregnancy remains controversial because it is carcinogenic in rodents and mutagenic in bacteria [73,74]. In a study of 922 patients exposed to metronidazole in pregnancy, there was no association with congenital anomalies, preterm birth, or low birth weight [75].

Metronidazole is excreted into breast milk and is a possible mutagen. Breastfeeding should be avoided for 12 to 24 hours after a single oral dose. [76]. Ciprofloxacin is not recommended in pregnant patients and in children under age 18 as it affects growing cartilage and can cause arthropathy. However, among 200 pregnancies exposed to fluoroquinolones (including ciprofloxacin), the incidence of adverse events were similar to controls [77].

Ciprofloxacin is excreted into breast milk, but short courses of ciprofloxacin are probably safe during breastfeeding due to the low concentration in breast milk [76].

Amoxicillin-clavulanic acid has a favorable safety profile and is considered acceptable for use during pregnancy and lactation [78]. Rifaximin has no published safety data in human pregnancy or lactation. Per their label, animal studies have revealed evidence of teratogenicity at doses about 0.9 to 5 times (rats) and 0.7 to 33 times (rabbits) the recommended human doses of 600 to 1650 mg per day [79]. It should be avoided in pregnancy. There are no published pregnancy safety data for probiotics in IBD. However, there are several large ongoing trials regarding the use of specific probiotics in all pregnancies to improve outcomes including preterm birth and infant infections and allergies [80].

Sulfasalazine and 5-aminosalicylic acid — Sulfasalazine and 5-aminosalicylic acid (5-ASA) drugs can be used during pregnancy and lactation.

The incidence of decreased birth weight, prematurity, spontaneous abortion, stillbirths, or congenital anomaly is similar in children born to mothers taking sulfasalazine compared with the general population [81]. Sulfasalazine and its sulfapyridine metabolite can be found in umbilical cord blood at similar concentrations to maternal blood. However, these concentrations do not cause significant displacement of bilirubin from albumin [82,83]. As a result, sulfasalazine does not have to be discontinued during pregnancy.

Folate supplementation (2 mg daily) should be recommended in patients on sulfasalazine as it interferes with folic acid metabolism [6]. (See 'Nutrition and supplements' above.)

Studies suggest that 5-ASA agents by themselves are safe during pregnancy [84-87]. Enteric coating of mesalamine with dibutyl phthalate (DBP) has been associated with skeletal malformations and adverse effects on the male reproductive system in fetal animal models exposed to high doses of DBP [88]. However, at this time there is no commercial preparation with DBP in the coating (table 1). (See "Sulfasalazine and 5-aminosalicylates in the treatment of inflammatory bowel disease" and "Occupational and environmental risks to reproduction in females: Specific exposures and impact", section on 'Bisphenol A and other phenols'.)

Sulfasalazine and 5-ASAs are excreted in breast milk in low concentrations. Infants whose mothers are on sulfasalazine and 5-ASAs should be monitored for the development of diarrhea, a rare complication of breastfeeding [76,89,90].

Glucocorticoids — Glucocorticoids are considered low risk in pregnancy and should be administered for the same indications as a nonpregnant patient with IBD. The lowest possible dose of systemic glucocorticoids should be used and they should not be considered an alternative to standard maintenance therapy. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Glucocorticoids' and "Preterm birth: Risk factors, interventions for risk reduction, and maternal prognosis".)

Although older studies have demonstrated a threefold increase in the risk of cleft palate after maternal exposure to glucocorticoids, the absolute risk is low (0.2 to 0.4 percent) [91] and multiple other studies have not confirmed that risk [92]. In addition, as palatal closure is usually complete by the 12th week of pregnancy, the risk is limited to administration during the first trimester. (See "Etiology, prenatal diagnosis, obstetric management, and recurrence of cleft lip and/or palate".)

Fetal adrenal insufficiency and low birth weight have also been associated with glucocorticoids [93]. However, adrenal insufficiency of the fetus following maternal administration of glucocorticoids is rare as rapid maternal metabolism of prednisolone, binding to serum proteins, and conversion to inactive metabolites by placental 11 beta-hydroxysteroid dehydrogenase results in relatively low fetal concentrations [94,95]. However, long-term administration of high doses of glucocorticoids (>20 mg) is associated with adrenal insufficiency and warrants close neonatal monitoring. (See "Overview of antepartum fetal assessment".)

Glucocorticoids have the potential for exacerbating pregnancy-induced hypertension, gestational diabetes, and preterm delivery from premature rupture of membranes [96]. Therefore, pregnant patients on glucocorticoids should be appropriately monitored for these complications [91,97]. (See "Gestational hypertension" and "Gestational diabetes mellitus: Glucose management and maternal prognosis" and "Gestational diabetes mellitus: Obstetric issues and management" and "Preterm prelabor rupture of membranes: Clinical manifestations and diagnosis".)

Although low levels of prednisone and its metabolite, prednisolone, can be measured in breast milk, these are unlikely to be clinically significant. In one study, following a dose of prednisolone 50 mg IV, an average of 0.025 percent was recovered from breast milk [98].

Azathioprine and mercaptopurine — Thiopurines, azathioprine and mercaptopurine, should be continued during pregnancy if they are monotherapy [99,100]. In a meta-analysis of 3045 patients with IBD, thiopurine exposure was associated with preterm birth (odds ratio [OR] 1.7, 95% CI 1.3-2.2), but not with congenital abnormalities or low birth weight [36]. In addition, discontinuing azathioprine and mercaptopurine is associated with a high rate of relapse and active IBD is itself associated with an increased risk of low birth weight [100-107].

Azathioprine and mercaptopurine are detectable in breast milk but studies have demonstrated that levels in breast milk are low and metabolites are undetectable in breastfed neonates [108,109]. Although data are limited, these studies suggest that breastfeeding is safe. Breastfeeding can be continued while on azathioprine and mercaptopurine after a detailed discussion of the risks and benefits of breastfeeding [6]. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Azathioprine and 6-mercaptopurine'.)

Cyclosporine — Cyclosporine is effective at inducing remission in patients with severe steroid refractory ulcerative colitis during pregnancy [57]. When the administration of cyclosporine is necessary during pregnancy, the minimum dose should be used. (See "Management of the hospitalized adult patient with severe ulcerative colitis".)

In a meta-analysis, cyclosporine was not associated with an increase in risk of congenital malformations but was associated with increased rates of prematurity [110]. A higher incidence of small for gestational age infants has also been reported. However, it is unclear if fetal growth restriction and prematurity were due to cyclosporine or due to the mother’s underlying disease.

Cyclosporine is excreted in breast milk, and therapeutic levels have been reported in breastfed infants [111,112]. However, as the amount of transfer is minimal, cyclosporine can be used in the breastfeeding mother and infant levels of drug can be monitored if concern for toxicity. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Cyclosporine'.)

Methotrexate — Methotrexate is contraindicated in pregnancy as it is a potent abortifacient, and its use during pregnancy is associated with multiple skeletal abnormalities. Female patients who plan to conceive should, therefore, discontinue methotrexate and use contraception for at least three months, and ideally six months, prior to conception.

The rate of congenital malformations due to maternal methotrexate use is estimated to be 9 to 17 percent [113,114]. The risk of methotrexate-induced developmental toxicity is highest at 8 to 10 weeks of gestation and with doses ≥10 mg per week.

Breastfeeding is contraindicated in patients on methotrexate as it is excreted into breast milk and can accumulate in neonatal tissues [6,115]. (See "Safety of rheumatic disease medication use during pregnancy and lactation", section on 'Methotrexate'.)

Anti-tumor necrosis factor agents — Experience with anti-TNF agents used to treat IBD during pregnancy is reassuring [116-121]. The Toronto Consensus on Management of IBD in pregnancy recommended that pregnant patients on anti-TNF monotherapy for maintenance should continue therapy throughout pregnancy [122]. Switching from a combination of a biologic plus thiopurine therapy to anti-TNF monotherapy may be considered in very select low-risk patients. Low-risk patients are those in sustained remission prior to pregnancy (ie, symptomatic, glucocorticoid-free remission with endoscopic healing for at least three months duration) and no history of multiple medication failures, significant perianal disease, or multiple surgeries.

Data have consistently suggested that the use of anti-TNF agents throughout pregnancy may lower the risk of a disease flare [121,123]. In a population-based study including 5293 pregnancies exposed to anti-TNF agents from conception, use of anti-TNF therapy beyond 24 weeks was associated with lower risk of a maternal IBD flare compared with stopping anti-TNF therapy, after adjusting for factors such as IBD type and glucocorticoid use (35.8 versus 39 percent; adjusted risk ratio [aRR] 0.93, 95% CI 0.86-0.99) [121]. In addition, continuing anti-TNF agents beyond 24 weeks was not associated with increased risk of adverse pregnancy outcomes (ie, stillbirth, low birth weight, preterm birth).

The use of anti-TNF agents (alone or in combination with thiopurines) during pregnancy does not appear to increase the risk of severe infection for the child [118,121]. In a study with mean follow-up of four years that included 841 children born to mothers with IBD, the rate of severe infection was similar in children exposed to anti-TNF agents in utero compared with unexposed children (1.6 versus 2.8 percent per person-year; HR 1.2, 95% CI 0.8-1.8) [118].

Use of anti-TNF therapy during pregnancy has not been associated with an increased risk of adverse pregnancy outcomes, and continuing anti-TNF therapy beyond 24 weeks may lower the risk of preterm birth [121,124-128]. In a study including 5293 pregnancies exposed to anti-TNF agents from conception, use of anti-TNF therapy beyond 24 weeks was associated with lower risk of preterm birth compared with stopping anti-TNF therapy (7.6 versus 8.9 percent; aRR 0.82, 95% CI 0.68-0.99) [121]. In an analysis of data from a prospective cohort (Pregnancy in IBD and Neonatal Outcomes Registry [PIANO]) including 1490 pregnant patients (ie, 242 patients exposed to azathioprine, 642 patients exposed to biologics, 227 patients exposed to combination therapy, and 379 patients unexposed), use of biologic, thiopurine, or combination therapy was not associated with increased risk for congenital malformations, spontaneous abortion, preterm birth, low birth weight, or infections at one year [128]. Patients with ulcerative colitis tended to have higher rates of disease activity compared with patients with Crohn disease. (See "Safety of rheumatic disease medication use during pregnancy and lactation".)

Infliximab — Infliximab is compatible with use during conception in male and female patients and throughout pregnancy [117,122]. Rates of live births, miscarriages, and therapeutic terminations do not appear to be significantly different in patients exposed to infliximab during pregnancy compared with the general population [125,129-131].

Infliximab should be continued throughout pregnancy to avoid flares. Discontinuation of therapy can lead to increased disease activity and increased adverse outcomes to the child [121,123]. The dose should ideally be given so that the patient is due for the next dose soon after delivery. For example, if the dosing schedule is every eight weeks, the infliximab dose is timed to be given between weeks 30 to 34; alternatively, if the dosing schedule is every four weeks, the dose is timed to be given between weeks 35 to 37. Infliximab is considered compatible with breastfeeding as it is excreted in trace amounts in human milk and is poorly absorbed orally, thus reducing the potential for systemic adverse effects [6,132].

Infliximab crosses the placenta [133]. The exact timing is unclear, but in general, maternal antibodies are actively transported across the placenta by the neonatal Fc receptor (FcRn) beginning in the early to mid-second trimester, with the highest fetal concentration in the third trimester near term. High levels of infliximab have been detected in the cord blood of newborns [133,134], and this raises concern about an increased risk of infection and a suboptimal response to vaccinations in the newborn [135]. However, in a cohort study including 179 pregnant patients with IBD, infants born to mothers on biologic therapy did not have lower rates of adequate serologic response to Haemophilus influenza B (HiB) or tetanus toxin following vaccination compared with infants unexposed to biologic therapy (HiB group: 71 versus 50 percent, and tetanus toxoid group: 80 versus 75 percent) [136]. In addition, the median cord blood concentrations of infliximab were similar in infants with an adequate serologic response to the vaccines compared with infants without an adequate response.

Live vaccines should not be given to infants exposed to infliximab, adalimumab, golimumab, or natalizumab, as newborns may have detectable serum levels of drug for up to nine months [122,133,137]. All other vaccinations can be given on schedule. (See "Standard immunizations for children and adolescents: Overview", section on 'Routine schedule'.)

Adalimumab — Adalimumab is considered compatible with use during conception and throughout pregnancy [122]. In observational studies, the incidence of congenital malformations, spontaneous abortion, stillbirth, and preterm delivery was not increased in pregnant patients exposed to adalimumab [133,134,138]. Adalimumab actively crosses the placenta as well [133].

Limited evidence suggests that adalimumab is compatible with breastfeeding. While it can be detected in breast milk, levels are low and it has not been demonstrated to adversely affect the nursing infant [139].

Certolizumab — Certolizumab is a PEGylated Fab fragment of a humanized anti-TNF-alpha monoclonal antibody. Unlike other anti-TNF agents, which are actively transferred across the placenta in the third trimester, there is minimal placental transfer of certolizumab to the infant during pregnancy as it does not have an Fc component to bind to the FcRn on the placenta [133].

Certolizumab can therefore be used through conception and continued throughout pregnancy until delivery. Although there are limited data, certolizumab pegol may also be compatible with breastfeeding as it is minimally excreted into breast milk [117]. There is no need to adjust vaccine schedules in infants exposed to certolizumab in utero.

Other anti-TNF agents — We typically continue golimumab in patients with ulcerative colitis who are pregnant or breastfeeding. (See "Overview of dosing and monitoring of biologic agents and small molecules for treating ulcerative colitis in adults", section on 'Tumor necrosis factor inhibitors'.)

Other biologic agents

Vedolizumab – We typically continue vedolizumab in pregnant patients to avoid disease flares. Limited data on vedolizumab use during pregnancy suggested that vedolizumab was not associated with an increased risk of miscarriage, stillbirth, or congenital anomalies [140,141]. In an observational, case-control study including 73 patients with IBD (79 pregnancies), miscarriage rates were not significantly different for patients given vedolizumab compared with patients given anti-TNF agents or a control group who were not exposed to biologics or immunomodulators (16 versus 13 and 10 percent, respectively) [140]. Stillbirth rates were not significantly different for patients treated with vedolizumab compared with anti-TNF agents or with an unexposed control group. In addition, data from 25 pregnancies in patients with IBD found no increase in congenital anomalies [141].

For patients who are breastfeeding, we also continue vedolizumab therapy. Vedolizumab is detectable at very low levels in breastmilk, which is similar to other biologic therapies [142]. (See 'Anti-tumor necrosis factor agents' above.)

Ustekinumab – There are limited safety data in Crohn disease, but safety registries in psoriasis report a 1.7 percent rate of fetal malformations, similar to what would be expected in the general population [143]. There are limited human data on lactation safety, but again, it is the author's practice to continue use during breastfeeding in the healthy term infant.

Risankizumab – We typically continue risankizumab in pregnant patients to reduce the risk of a disease flare. Limited studies did not show an increased risk of adverse pregnancy outcomes or infant adverse effects with use of risankizumab during pregnancy or lactation [144].

Natalizumab – There are limited safety data on the use of natalizumab during pregnancy and lactation. The risk of congenital malformations was not increased in 164 pregnancies in patients with Crohn disease or multiple sclerosis on natalizumab during the first trimester [145].

Small molecules

Tofacitinib – There are limited data on use of tofacitinib, and we avoid its use, particularly in the first trimester, in pregnant patients and breastfeeding mothers given teratogenicity documented in animals.

Upadacitinib – We avoid the use of upadacitinib in pregnant patients. Upadacitinib crosses the placenta in the first trimester, and animal data show teratogenicity at doses equal to and less than the daily human dose [146].

Antidiarrheal drugs — Antidiarrheal drugs should be avoided, especially early in pregnancy. Antidiarrheals should only be used for severe diarrhea that cannot be controlled with dietary manipulation and bulking agents (eg, kaolin/pectin, psyllium).

Diphenoxylate with atropine and loperamide are not teratogenic in animal studies but there are no controlled studies in humans. Case reports have reported fetal malformations in infants exposed to diphenoxylate with atropine during the first trimester, but it is unclear if this was causal [147,148].

Loperamide is compatible with nursing [76]. However, there are no data for diphenoxylate. Diphenoxylate-atropine should therefore be used only in short, infrequent courses, at low doses while breastfeeding an older infant.

Anticoagulants — Pregnancy and the puerperium, as well as a diagnosis of IBD, are well-established risk factors for deep vein thrombosis (DVT) and pulmonary embolism (PE), which are collectively referred to as venous thromboembolic disease (VTE). Patients should receive VTE prophylaxis during any hospitalization and after cesarean delivery [1]. Prevention of VTE and the use of anticoagulants in pregnant patients are discussed separately. (See "Deep vein thrombosis and pulmonary embolism in pregnancy: Prevention" and "Use of anticoagulants during pregnancy and postpartum".)

SURGERY — Complications associated with IBD that warrant surgery in a pregnant patient include acute refractory colitis, perforation, abscesses, severe hemorrhage, and obstruction [6]. There are limited data pertaining to the risk of surgery in pregnant patients with IBD [149,150]. Surgery has been associated with preterm labor and spontaneous abortions, possibly related to inadvertent uterine manipulation, but complications are rare [6,151,152]. When possible, medical treatment should be intensified prior to considering surgery [153]. (See "Clinical manifestations, diagnosis, and prognosis of ulcerative colitis in adults", section on 'Acute complications' and "Surgical management of Crohn disease".)

Nonurgent surgery that cannot wait until delivery is ideally performed in the second trimester. The timing of nonobstetric surgery for pregnant patients is discussed separately. (See "Nonobstetric surgery in pregnant patients: Patient counseling, surgical considerations, and obstetric management", section on 'Timing of surgery'.)

MODE OF DELIVERY — Patients with active perineal disease or rectal involvement with Crohn disease should undergo cesarean delivery in order to avoid perineal trauma from vaginal birth that can trigger or worsen existing perineal disease [122,154]. The mode of delivery in all other patients with inflammatory bowel disease should be dictated by obstetric necessity. In patients with an ileoanal anastomosis or J pouch, many advocate planned cesarean delivery as well, though data suggest no long-term worsening of pouch function [154].

Vaginal delivery can be attempted in patients with a colostomy, ileostomy, or continent ileostomy and has not been associated with an increased risk of ostomy complications [6,155]. (See "Ileostomy or colostomy care and complications".)

POSTPARTUM CARE — Some studies have reported an increased risk of postpartum flares [29,156,157]. Risk factors for disease flares may include hormonal fluctuations, discontinuing IBD medications during lactation, and resuming tobacco use [157]. For patients with a clear pregnancy plan who continue medications during lactation, follow-up should occur within six months following delivery or sooner if symptoms develop or worsen prior to the visit.

Normal physiologic changes and routine maternal care in the postpartum period are discussed separately. (See "Overview of the postpartum period: Normal physiology and routine maternal care".)

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: Ulcerative colitis in adults" and "Society guideline links: Crohn disease in adults".)

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.)

Beyond the Basics topics (see "Patient education: Inflammatory bowel disease and pregnancy (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

Background – IBD can affect patients in their childbearing years and therefore has implications for fertility, pregnancy, and nursing. (See 'Introduction' above.)

Patients with quiescent IBD do not have decreased fertility compared with the general population. However, fertility in patients with IBD may be affected by active inflammation, medications, and prior surgery. (See 'Fertility' above.)

During pregnancy, patients with ulcerative colitis have more disease activity than patients with Crohn disease. Pregnant patients with IBD are at an increased risk for worse obstetric and pregnancy-related outcomes compared with the general population, even with disease in remission. (See 'Pregnancy and lactation' above.)

Preconception counseling – The course of IBD during pregnancy is determined in part by the activity of the disease at conception. Patients in remission at the time of conception are likely to remain in remission during pregnancy. In contrast, up to 70 percent of patients with active disease at conception are likely to have continued or worsening symptoms during pregnancy. (See 'Disease control' above.)

Diagnostic evaluation during pregnancy – Endoscopy should be performed during pregnancy only if there is a strong indication or if the information is critical for making treatment decisions. An unsedated flexible sigmoidoscopy is considered low risk. Magnetic resonance enterography is the preferred diagnostic modality in pregnant patients with IBD in the absence of institutional expertise with ultrasound for IBD. (See 'Evaluating disease activity during pregnancy and lactation' above.)

Medication use – The choice of anti-inflammatory and immunosuppressive medications during pregnancy and lactation should be based upon their relative safety and indications as well as the risk of relapse of IBD if the medications are discontinued. (See 'Medications during pregnancy and lactation' above.)

Mode of delivery – Patients with perineal disease or rectal involvement with Crohn disease should undergo a cesarean delivery. The mode of delivery in all other patients with IBD should be dictated by obstetric necessity. (See 'Mode of delivery' above.)

Surgery in pregnant patients – Complications associated with IBD that warrant surgery in a pregnant patient include acute refractory colitis, perforation, abscesses, severe hemorrhage, and obstruction. Although data are limited, surgery is associated with premature labor or spontaneous abortion, possibly related to inadvertent uterine manipulation. (See 'Surgery' above.)

ACKNOWLEDGMENT — We are saddened by the death of Paul Rutgeerts, MD, who passed away in September 2020. UpToDate gratefully acknowledges Dr. Rutgeerts' work as our Section Editor for Gastroenterology.

  1. Mahadevan U, Robinson C, Bernasko N, et al. Inflammatory Bowel Disease in Pregnancy Clinical Care Pathway: A Report From the American Gastroenterological Association IBD Parenthood Project Working Group. Gastroenterology 2019; 156:1508.
  2. Baird DD, Narendranathan M, Sandler RS. Increased risk of preterm birth for women with inflammatory bowel disease. Gastroenterology 1990; 99:987.
  3. Hudson M, Flett G, Sinclair TS, et al. Fertility and pregnancy in inflammatory bowel disease. Int J Gynaecol Obstet 1997; 58:229.
  4. Woolfson K, Cohen Z, McLeod RS. Crohn's disease and pregnancy. Dis Colon Rectum 1990; 33:869.
  5. Tavernier N, Fumery M, Peyrin-Biroulet L, et al. Systematic review: fertility in non-surgically treated inflammatory bowel disease. Aliment Pharmacol Ther 2013; 38:847.
  6. Van Assche G, Dignass A, Reinisch W, et al. The second European evidence-based Consensus on the diagnosis and management of Crohn's disease: Special situations. J Crohns Colitis 2010; 4:63.
  7. Birnie GG, McLeod TI, Watkinson G. Incidence of sulphasalazine-induced male infertility. Gut 1981; 22:452.
  8. O'Moráin C, Smethurst P, Doré CJ, Levi AJ. Reversible male infertility due to sulphasalazine: studies in man and rat. Gut 1984; 25:1078.
  9. Toovey S, Hudson E, Hendry WF, Levi AJ. Sulphasalazine and male infertility: reversibility and possible mechanism. Gut 1981; 22:445.
  10. Ley D, Jones J, Parrish J, et al. Methotrexate Reduces DNA Integrity in Sperm From Men With Inflammatory Bowel Disease. Gastroenterology 2018; 154:2064.
  11. Grosen A, Bungum M, Christensen LA, et al. Semen Quality and Sperm DNA Integrity in Patients With Severe Active Inflammatory Bowel Disease and Effects of Tumour Necrosis Factor-alpha Inhibitors. J Crohns Colitis 2019; 13:564.
  12. Narendranathan M, Sandler RS, Suchindran CM, Savitz DA. Male infertility in inflammatory bowel disease. J Clin Gastroenterol 1989; 11:403.
  13. Ørding Olsen K, Juul S, Berndtsson I, et al. Ulcerative colitis: female fecundity before diagnosis, during disease, and after surgery compared with a population sample. Gastroenterology 2002; 122:15.
  14. Waljee A, Waljee J, Morris AM, Higgins PD. Threefold increased risk of infertility: a meta-analysis of infertility after ileal pouch anal anastomosis in ulcerative colitis. Gut 2006; 55:1575.
  15. Nørgård BM, Larsen PV, Fedder J, et al. Live birth and adverse birth outcomes in women with ulcerative colitis and Crohn's disease receiving assisted reproduction: a 20-year nationwide cohort study. Gut 2016; 65:767.
  16. Laharie D, Debeugny S, Peeters M, et al. Inflammatory bowel disease in spouses and their offspring. Gastroenterology 2001; 120:816.
  17. Roth MP, Petersen GM, McElree C, et al. Familial empiric risk estimates of inflammatory bowel disease in Ashkenazi Jews. Gastroenterology 1989; 96:1016.
  18. Yang H, McElree C, Roth MP, et al. Familial empirical risks for inflammatory bowel disease: differences between Jews and non-Jews. Gut 1993; 34:517.
  19. Satsangi J, Grootscholten C, Holt H, Jewell DP. Clinical patterns of familial inflammatory bowel disease. Gut 1996; 38:738.
  20. Jølving LR, Nielsen J, Beck-Nielsen SS, et al. The Association Between Maternal Chronic Inflammatory Bowel Disease and Long-term Health Outcomes in Children-A Nationwide Cohort Study. Inflamm Bowel Dis 2017; 23:1440.
  21. Bayless TM, Tokayer AZ, Polito JM 2nd, et al. Crohn's disease: concordance for site and clinical type in affected family members--potential hereditary influences. Gastroenterology 1996; 111:573.
  22. Colombel JF, Grandbastien B, Gower-Rousseau C, et al. Clinical characteristics of Crohn's disease in 72 families. Gastroenterology 1996; 111:604.
  23. Polito JM 2nd, Childs B, Mellits ED, et al. Crohn's disease: influence of age at diagnosis on site and clinical type of disease. Gastroenterology 1996; 111:580.
  24. Peeters M, Nevens H, Baert F, et al. Familial aggregation in Crohn's disease: increased age-adjusted risk and concordance in clinical characteristics. Gastroenterology 1996; 111:597.
  25. Henckaerts L, Van Steen K, Verstreken I, et al. Genetic risk profiling and prediction of disease course in Crohn's disease patients. Clin Gastroenterol Hepatol 2009; 7:972.
  26. Annese V, Andreoli A, Astegiano M, et al. Clinical features in familial cases of Crohn's disease and ulcerative colitis in Italy: a GISC study. Italian Study Group for the Disease of Colon and Rectum. Am J Gastroenterol 2001; 96:2939.
  27. Grandbastien B, Peeters M, Franchimont D, et al. Anticipation in familial Crohn's disease. Gut 1998; 42:170.
  28. Abhyankar A, Ham M, Moss AC. Meta-analysis: the impact of disease activity at conception on disease activity during pregnancy in patients with inflammatory bowel disease. Aliment Pharmacol Ther 2013; 38:460.
  29. Pedersen N, Bortoli A, Duricova D, et al. The course of inflammatory bowel disease during pregnancy and postpartum: a prospective European ECCO-EpiCom Study of 209 pregnant women. Aliment Pharmacol Ther 2013; 38:501.
  30. Nørgård B, Hundborg HH, Jacobsen BA, et al. Disease activity in pregnant women with Crohn's disease and birth outcomes: a regional Danish cohort study. Am J Gastroenterol 2007; 102:1947.
  31. Shaffer JL, Kershaw A, Berrisford MH. Sulphasalazine-induced infertility reversed on transfer to 5-aminosalicylic acid. Lancet 1984; 1:1240.
  32. Wu FC, Aitken RJ, Ferguson A. Inflammatory bowel disease and male infertility: effects of sulfasalazine and 5-aminosalicylic acid on sperm-fertilizing capacity and reactive oxygen species generation. Fertil Steril 1989; 52:842.
  33. Friedman S, Larsen MD, Magnussen B, et al. Paternal use of azathioprine/6-mercaptopurine or methotrexate within 3 months before conception and long-term health outcomes in the offspring-A nationwide cohort study. Reprod Toxicol 2017; 73:196.
  34. Saxena AK, Dhungel S, Bhattacharya S, et al. Effect of chronic low dose of methotrexate on cellular proliferation during spermatogenesis in rats. Arch Androl 2004; 50:33.
  35. Winter RW, Larsen MD, Magnussen B, et al. Birth outcomes after preconception paternal exposure to methotrexate: A nationwide cohort study. Reprod Toxicol 2017; 74:219.
  36. Akbari M, Shah S, Velayos FS, et al. Systematic review and meta-analysis on the effects of thiopurines on birth outcomes from female and male patients with inflammatory bowel disease. Inflamm Bowel Dis 2013; 19:15.
  37. Meserve J, Luo J, Zhu W, et al. Paternal Exposure to Immunosuppressive and/or Biologic Agents and Birth Outcomes in Patients With Immune-Mediated Inflammatory Diseases. Gastroenterology 2021; 161:107.
  38. Xu L, Han S, Liu Y, et al. The influence of immunosuppressants on the fertility of males who undergo renal transplantation and on the immune function of their offspring. Transpl Immunol 2009; 22:28.
  39. Bengtson MB, Aamodt G, Mahadevan U, Vatn MH. Inadequate Gestational Weight Gain, the Hidden Link Between Maternal IBD and Adverse Pregnancy Outcomes: Results from the Norwegian Mother and Child Cohort Study. Inflamm Bowel Dis 2017; 23:1225.
  40. Mullin GE. Micronutrients and inflammatory bowel disease. Nutr Clin Pract 2012; 27:136.
  41. Cornish J, Tan E, Teare J, et al. A meta-analysis on the influence of inflammatory bowel disease on pregnancy. Gut 2007; 56:830.
  42. Mahadevan U, Sandborn WJ, Li DK, et al. Pregnancy outcomes in women with inflammatory bowel disease: a large community-based study from Northern California. Gastroenterology 2007; 133:1106.
  43. Nguyen GC, Boudreau H, Harris ML, Maxwell CV. Outcomes of obstetric hospitalizations among women with inflammatory bowel disease in the United States. Clin Gastroenterol Hepatol 2009; 7:329.
  44. Kane S, Kisiel J, Shih L, Hanauer S. HLA disparity determines disease activity through pregnancy in women with inflammatory bowel disease. Am J Gastroenterol 2004; 99:1523.
  45. Nielsen OH, Andreasson B, Bondesen S, Jarnum S. Pregnancy in ulcerative colitis. Scand J Gastroenterol 1983; 18:735.
  46. Hanan IM, Kirsner JB. Inflammatory bowel disease in the pregnant woman. Clin Perinatol 1985; 12:669.
  47. Rogers RG, Katz VL. Course of Crohn's disease during pregnancy and its effect on pregnancy outcome: a retrospective review. Am J Perinatol 1995; 12:262.
  48. Korelitz BI. Pregnancy. In: Seminars in Colon and Rectal Surgery, Peppercorn MA (Ed), WB Saunders, 1993. p.48.
  49. Castiglione F, Pignata S, Morace F, et al. Effect of pregnancy on the clinical course of a cohort of women with inflammatory bowel disease. Ital J Gastroenterol 1996; 28:199.
  50. Riis L, Vind I, Politi P, et al. Does pregnancy change the disease course? A study in a European cohort of patients with inflammatory bowel disease. Am J Gastroenterol 2006; 101:1539.
  51. Bröms G, Granath F, Linder M, et al. Complications from inflammatory bowel disease during pregnancy and delivery. Clin Gastroenterol Hepatol 2012; 10:1246.
  52. Bortoli A, Pedersen N, Duricova D, et al. Pregnancy outcome in inflammatory bowel disease: prospective European case-control ECCO-EpiCom study, 2003-2006. Aliment Pharmacol Ther 2011; 34:724.
  53. Bröms G, Granath F, Linder M, et al. Birth outcomes in women with inflammatory bowel disease: effects of disease activity and drug exposure. Inflamm Bowel Dis 2014; 20:1091.
  54. Getahun D, Fassett MJ, Longstreth GF, et al. Association between maternal inflammatory bowel disease and adverse perinatal outcomes. J Perinatol 2014; 34:435.
  55. Nørgård B, Fonager K, Sørensen HT, Olsen J. Birth outcomes of women with ulcerative colitis: a nationwide Danish cohort study. Am J Gastroenterol 2000; 95:3165.
  56. Stephansson O, Larsson H, Pedersen L, et al. Crohn's disease is a risk factor for preterm birth. Clin Gastroenterol Hepatol 2010; 8:509.
  57. Reddy D, Murphy SJ, Kane SV, et al. Relapses of inflammatory bowel disease during pregnancy: in-hospital management and birth outcomes. Am J Gastroenterol 2008; 103:1203.
  58. Baiocco PJ, Korelitz BI. The influence of inflammatory bowel disease and its treatment on pregnancy and fetal outcome. J Clin Gastroenterol 1984; 6:211.
  59. Khosla R, Willoughby CP, Jewell DP. Crohn's disease and pregnancy. Gut 1984; 25:52.
  60. Mayberry JF, Weterman IT. European survey of fertility and pregnancy in women with Crohn's disease: a case control study by European collaborative group. Gut 1986; 27:821.
  61. Fedorkow DM, Persaud D, Nimrod CA. Inflammatory bowel disease: a controlled study of late pregnancy outcome. Am J Obstet Gynecol 1989; 160:998.
  62. Kane S, Lemieux N. The role of breastfeeding in postpartum disease activity in women with inflammatory bowel disease. Am J Gastroenterol 2005; 100:102.
  63. Shmidt E, Dubinsky MC. Inflammatory Bowel Disease and Pregnancy. Am J Gastroenterol 2022; 117:60.
  64. Julsgaard M, Hvas CL, Gearry RB, et al. Fecal Calprotectin Is Not Affected by Pregnancy: Clinical Implications for the Management of Pregnant Patients with Inflammatory Bowel Disease. Inflamm Bowel Dis 2017; 23:1240.
  65. Kim ES, Tarassishin L, Eisele C, et al. Longitudinal Changes in Fecal Calprotectin Levels Among Pregnant Women With and Without Inflammatory Bowel Disease and Their Babies. Gastroenterology 2021; 160:1118.
  66. Tandon P, Lee EY, Maxwell C, et al. Fecal Calprotectin May Predict Adverse Pregnancy-Related Outcomes in Patients with Inflammatory Bowel Disease. Dig Dis Sci 2021; 66:1639.
  67. Kammerlander H, Nielsen J, Kjeldsen J, et al. Fecal Calprotectin During Pregnancy in Women With Moderate-Severe Inflammatory Bowel Disease. Inflamm Bowel Dis 2018; 24:839.
  68. Cappell MS, Colon VJ, Sidhom OA. A study at 10 medical centers of the safety and efficacy of 48 flexible sigmoidoscopies and 8 colonoscopies during pregnancy with follow-up of fetal outcome and with comparison to control groups. Dig Dis Sci 1996; 41:2353.
  69. Homan WP, Thorbjarnarson B. Crohn disease and pregnancy. Arch Surg 1976; 111:545.
  70. Kanal E, Barkovich AJ, Bell C, et al. ACR guidance document for safe MR practices: 2007. AJR Am J Roentgenol 2007; 188:1447.
  71. Czeizel AE, Rockenbauer M. A population based case-control teratologic study of oral metronidazole treatment during pregnancy. Br J Obstet Gynaecol 1998; 105:322.
  72. Caro-Patón T, Carvajal A, Martin de Diego I, et al. Is metronidazole teratogenic? A meta-analysis. Br J Clin Pharmacol 1997; 44:179.
  73. Beard CM, Noller KL, O'Fallon WM, et al. Cancer after exposure to metronidazole. Mayo Clin Proc 1988; 63:147.
  74. Falagas ME, Walker AM, Jick H, et al. Late incidence of cancer after metronidazole use: a matched metronidazole user/nonuser study. Clin Infect Dis 1998; 26:384.
  75. Koss CA, Baras DC, Lane SD, et al. Investigation of metronidazole use during pregnancy and adverse birth outcomes. Antimicrob Agents Chemother 2012; 56:4800.
  76. American Academy of Pediatrics Committee on Drugs. Transfer of drugs and other chemicals into human milk. Pediatrics 2001; 108:776.
  77. Loebstein R, Addis A, Ho E, et al. Pregnancy outcome following gestational exposure to fluoroquinolones: a multicenter prospective controlled study. Antimicrob Agents Chemother 1998; 42:1336.
  78. Czeizel AE, Rockenbauer M, Sørensen HT, Olsen J. Augmentin treatment during pregnancy and the prevalence of congenital abnormalities: a population-based case-control teratologic study. Eur J Obstet Gynecol Reprod Biol 2001; 97:188.
  79. Jodlowski TZ, Oehler R, Kam LW, Melnychuk I. Emerging therapies in the treatment of Clostridium difficile-associated disease. Ann Pharmacother 2006; 40:2164.
  80. Reid G, Kumar H, Khan AI, et al. The case in favour of probiotics before, during and after pregnancy: insights from the first 1,500 days. Benef Microbes 2016; 7:353.
  81. Mogadam M, Dobbins WO 3rd, Korelitz BI, Ahmed SW. Pregnancy in inflammatory bowel disease: effect of sulfasalazine and corticosteroids on fetal outcome. Gastroenterology 1981; 80:72.
  82. Järnerot G, Into-Malmberg MB, Esbjörner E. Placental transfer of sulphasalazine and sulphapyridine and some of its metabolites. Scand J Gastroenterol 1981; 16:693.
  83. Järnerot G, Andersen S, Esbjörner E, et al. Albumin reserve for binding of bilirubin in maternal and cord serum under treatment with sulphasalazine. Scand J Gastroenterol 1981; 16:1049.
  84. Habal FM, Hui G, Greenberg GR. Oral 5-aminosalicylic acid for inflammatory bowel disease in pregnancy: safety and clinical course. Gastroenterology 1993; 105:1057.
  85. Bell CM, Habal FM. Safety of topical 5-aminosalicylic acid in pregnancy. Am J Gastroenterol 1997; 92:2201.
  86. Trallori G, d'Albasio G, Bardazzi G, et al. 5-Aminosalicylic acid in pregnancy: clinical report. Ital J Gastroenterol 1994; 26:75.
  87. Diav-Citrin O, Park YH, Veerasuntharam G, et al. The safety of mesalamine in human pregnancy: a prospective controlled cohort study. Gastroenterology 1998; 114:23.
  88. US Food and Drug Administration. Asacol (mesalamine) delayed release tablets -- Detailed View: Safety Labeling Changes Approved By FDA Center for Drug Evaluation and Research (CDER), May 2010. Available at: http://www.fda.gov/Safety/MedWatch/SafetyInformation/ucm215476.htm (Accessed on December 14, 2011).
  89. Berlin CM Jr, Yaffe SJ. Disposition of salicylazosulfapyridine (Azulfidine) and metabolites in human breast milk. Dev Pharmacol Ther 1980; 1:31.
  90. Nelis GF. Diarrhoea due to 5-aminosalicylic acid in breast milk. Lancet 1989; 1:383.
  91. Park-Wyllie L, Mazzotta P, Pastuszak A, et al. Birth defects after maternal exposure to corticosteroids: prospective cohort study and meta-analysis of epidemiological studies. Teratology 2000; 62:385.
  92. Hviid A, Mølgaard-Nielsen D. Corticosteroid use during pregnancy and risk of orofacial clefts. CMAJ 2011; 183:796.
  93. Reinisch JM, Simon NG, Karow WG, Gandelman R. Prenatal exposure to prednisone in humans and animals retards intrauterine growth. Science 1978; 202:436.
  94. Yang K. Placental 11 beta-hydroxysteroid dehydrogenase: barrier to maternal glucocorticoids. Rev Reprod 1997; 2:129.
  95. Effect of corticosteroids for fetal maturation on perinatal outcomes. NIH Consensus Development Panel on the Effect of Corticosteroids for Fetal Maturation on Perinatal Outcomes. JAMA 1995; 273:413.
  96. Cowchock FS, Reece EA, Balaban D, et al. Repeated fetal losses associated with antiphospholipid antibodies: a collaborative randomized trial comparing prednisone with low-dose heparin treatment. Am J Obstet Gynecol 1992; 166:1318.
  97. Gur C, Diav-Citrin O, Shechtman S, et al. Pregnancy outcome after first trimester exposure to corticosteroids: a prospective controlled study. Reprod Toxicol 2004; 18:93.
  98. Greenberger PA, Odeh YK, Frederiksen MC, Atkinson AJ Jr. Pharmacokinetics of prednisolone transfer to breast milk. Clin Pharmacol Ther 1993; 53:324.
  99. Alstead EM, Ritchie JK, Lennard-Jones JE, et al. Safety of azathioprine in pregnancy in inflammatory bowel disease. Gastroenterology 1990; 99:443.
  100. Francella A, Dyan A, Bodian C, et al. The safety of 6-mercaptopurine for childbearing patients with inflammatory bowel disease: a retrospective cohort study. Gastroenterology 2003; 124:9.
  101. van Dieren JM, Kuipers EJ, Samsom JN, et al. Revisiting the immunomodulators tacrolimus, methotrexate, and mycophenolate mofetil: their mechanisms of action and role in the treatment of IBD. Inflamm Bowel Dis 2006; 12:311.
  102. Baumgart DC, Pintoffl JP, Sturm A, et al. Tacrolimus is safe and effective in patients with severe steroid-refractory or steroid-dependent inflammatory bowel disease--a long-term follow-up. Am J Gastroenterol 2006; 101:1048.
  103. Ierardi E, Principi M, Francavilla R, et al. Oral tacrolimus long-term therapy in patients with Crohn's disease and steroid resistance. Aliment Pharmacol Ther 2001; 15:371.
  104. Ng SC, Arebi N, Kamm MA. Medium-term results of oral tacrolimus treatment in refractory inflammatory bowel disease. Inflamm Bowel Dis 2007; 13:129.
  105. Benson A, Barrett T, Sparberg M, Buchman AL. Efficacy and safety of tacrolimus in refractory ulcerative colitis and Crohn's disease: a single-center experience. Inflamm Bowel Dis 2008; 14:7.
  106. Nørgård B, Pedersen L, Christensen LA, Sørensen HT. Therapeutic drug use in women with Crohn's disease and birth outcomes: a Danish nationwide cohort study. Am J Gastroenterol 2007; 102:1406.
  107. Coelho J, Beaugerie L, Colombel JF, et al. Pregnancy outcome in patients with inflammatory bowel disease treated with thiopurines: cohort from the CESAME Study. Gut 2011; 60:198.
  108. Christensen LA, Dahlerup JF, Nielsen MJ, et al. Azathioprine treatment during lactation. Aliment Pharmacol Ther 2008; 28:1209.
  109. Sau A, Clarke S, Bass J, et al. Azathioprine and breastfeeding: is it safe? BJOG 2007; 114:498.
  110. Bar Oz B, Hackman R, Einarson T, Koren G. Pregnancy outcome after cyclosporine therapy during pregnancy: a meta-analysis. Transplantation 2001; 71:1051.
  111. Petri M. Immunosuppressive drug use in pregnancy. Autoimmunity 2003; 36:51.
  112. Treacy G. Using an analogous monoclonal antibody to evaluate the reproductive and chronic toxicity potential for a humanized anti-TNFalpha monoclonal antibody. Hum Exp Toxicol 2000; 19:226.
  113. Østensen M, Khamashta M, Lockshin M, et al. Anti-inflammatory and immunosuppressive drugs and reproduction. Arthritis Res Ther 2006; 8:209.
  114. Chakravarty EF, Sanchez-Yamamoto D, Bush TM. The use of disease modifying antirheumatic drugs in women with rheumatoid arthritis of childbearing age: a survey of practice patterns and pregnancy outcomes. J Rheumatol 2003; 30:241.
  115. Johns DG, Rutherford LD, Leighton PC, Vogel CL. Secretion of methotrexate into human milk. Am J Obstet Gynecol 1972; 112:978.
  116. O'Donnell S, O'Morain C. Review article: use of antitumour necrosis factor therapy in inflammatory bowel disease during pregnancy and conception. Aliment Pharmacol Ther 2008; 27:885.
  117. Mahadevan U, Cucchiara S, Hyams JS, et al. The London Position Statement of the World Congress of Gastroenterology on Biological Therapy for IBD with the European Crohn's and Colitis Organisation: pregnancy and pediatrics. Am J Gastroenterol 2011; 106:214.
  118. Chaparro M, Verreth A, Lobaton T, et al. Long-Term Safety of In Utero Exposure to Anti-TNFα Drugs for the Treatment of Inflammatory Bowel Disease: Results from the Multicenter European TEDDY Study. Am J Gastroenterol 2018; 113:396.
  119. Matro R, Martin CF, Wolf D, et al. Exposure Concentrations of Infants Breastfed by Women Receiving Biologic Therapies for Inflammatory Bowel Diseases and Effects of Breastfeeding on Infections and Development. Gastroenterology 2018; 155:696.
  120. Nielsen OH, Gubatan JM, Juhl CB, et al. Biologics for Inflammatory Bowel Disease and Their Safety in Pregnancy: A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol 2022; 20:74.
  121. Meyer A, Neumann A, Drouin J, et al. Benefits and Risks Associated With Continuation of Anti-Tumor Necrosis Factor After 24 Weeks of Pregnancy in Women With Inflammatory Bowel Disease : A Nationwide Emulation Trial. Ann Intern Med 2022; 175:1374.
  122. Nguyen GC, Seow CH, Maxwell C, et al. The Toronto Consensus Statements for the Management of Inflammatory Bowel Disease in Pregnancy. Gastroenterology 2016; 150:734.
  123. Luu M, Benzenine E, Doret M, et al. Continuous Anti-TNFα Use Throughout Pregnancy: Possible Complications For the Mother But Not for the Fetus. A Retrospective Cohort on the French National Health Insurance Database (EVASION). Am J Gastroenterol 2018; 113:1669.
  124. Nielsen OH, Loftus EV Jr, Jess T. Safety of TNF-α inhibitors during IBD pregnancy: a systematic review. BMC Med 2013; 11:174.
  125. Schnitzler F, Fidder H, Ferrante M, et al. Outcome of pregnancy in women with inflammatory bowel disease treated with antitumor necrosis factor therapy. Inflamm Bowel Dis 2011; 17:1846.
  126. Seirafi M, de Vroey B, Amiot A, et al. Factors associated with pregnancy outcome in anti-TNF treated women with inflammatory bowel disease. Aliment Pharmacol Ther 2014; 40:363.
  127. O'Byrne LJ, Alqatari SG, Maher GM, et al. Fetal and maternal outcomes after maternal biologic use during conception and pregnancy: A systematic review and meta-analysis. BJOG 2022; 129:1236.
  128. Mahadevan U, Long MD, Kane SV, et al. Pregnancy and Neonatal Outcomes After Fetal Exposure to Biologics and Thiopurines Among Women With Inflammatory Bowel Disease. Gastroenterology 2021; 160:1131.
  129. Katz JA, Antoni C, Keenan GF, et al. Outcome of pregnancy in women receiving infliximab for the treatment of Crohn's disease and rheumatoid arthritis. Am J Gastroenterol 2004; 99:2385.
  130. Lichtenstein GR, Feagan BG, Cohen RD, et al. Serious infections and mortality in association with therapies for Crohn's disease: TREAT registry. Clin Gastroenterol Hepatol 2006; 4:621.
  131. Lichtenstein GR, Feagan BG, Mahadevan U, et al. Pregnancy Outcomes Reported During the 13-Year TREAT Registry: A Descriptive Report. Am J Gastroenterol 2018; 113:1678.
  132. Ben-Horin S, Yavzori M, Kopylov U, et al. Detection of infliximab in breast milk of nursing mothers with inflammatory bowel disease. J Crohns Colitis 2011; 5:555.
  133. Mahadevan U, Wolf DC, Dubinsky M, et al. Placental transfer of anti-tumor necrosis factor agents in pregnant patients with inflammatory bowel disease. Clin Gastroenterol Hepatol 2013; 11:286.
  134. Zelinkova Z, van der Ent C, Bruin KF, et al. Effects of discontinuing anti-tumor necrosis factor therapy during pregnancy on the course of inflammatory bowel disease and neonatal exposure. Clin Gastroenterol Hepatol 2013; 11:318.
  135. Cheent K, Nolan J, Shariq S, et al. Case Report: Fatal case of disseminated BCG infection in an infant born to a mother taking infliximab for Crohn's disease. J Crohns Colitis 2010; 4:603.
  136. Beaulieu DB, Ananthakrishnan AN, Martin C, et al. Use of Biologic Therapy by Pregnant Women With Inflammatory Bowel Disease Does Not Affect Infant Response to Vaccines. Clin Gastroenterol Hepatol 2018; 16:99.
  137. Julsgaard M, Christensen LA, Gibson PR, et al. Concentrations of Adalimumab and Infliximab in Mothers and Newborns, and Effects on Infection. Gastroenterology 2016; 151:110.
  138. Chambers CD, Johnson DL, Xu R, et al. Birth outcomes in women who have taken adalimumab in pregnancy: A prospective cohort study. PLoS One 2019; 14:e0223603.
  139. Ben-Horin S, Yavzori M, Katz L, et al. Adalimumab level in breast milk of a nursing mother. Clin Gastroenterol Hepatol 2010; 8:475.
  140. Moens A, van der Woude CJ, Julsgaard M, et al. Pregnancy outcomes in inflammatory bowel disease patients treated with vedolizumab, anti-TNF or conventional therapy: results of the European CONCEIVE study. Aliment Pharmacol Ther 2020; 51:129.
  141. Mahadevan U, Dubinsky M, Vermeire S, et al. Vedolizumab exposure in pregnancy: Outcomes from clinical studies in ulcerative colitis and Crohn's disease. Abstract. Am J Gastroenterol 2015; 80:S1.
  142. Lahat A, Shitrit AB, Naftali T, et al. Vedolizumab Levels in Breast Milk of Nursing Mothers With Inflammatory Bowel Disease. J Crohns Colitis 2018; 12:120.
  143. Levy RA, de Jesús GR, de Jesús NR, Klumb EM. Critical review of the current recommendations for the treatment of systemic inflammatory rheumatic diseases during pregnancy and lactation. Autoimmun Rev 2016; 15:955.
  144. Risankizumab. United States Prescribing Information. Revised 09/2022. US Food & Drug Administration. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/761105s016lbl.pdf (Accessed on November 04, 2022).
  145. Nazareth M, Cristiano L, Kooijmans M. Natalizumab use during pregnancy. Am J Gastroenterol 2008; 104.
  146. Upadacitinib. United States Prescribing Information. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/211675s010lbl.pdf (Accessed on November 16, 2022).
  147. Lewis JH, Weingold AB. The use of gastrointestinal drugs during pregnancy and lactation. Am J Gastroenterol 1985; 80:912.
  148. Briggs GG, Freeman RK, Yaffe SJ. Drugs in Pregnancy and Lactation, 8th ed, Lippincott Williams & Wilkins, Philadelphia, PA 2008.
  149. Boulton R, Hamilton M, Lewis A, et al. Fulminant ulcerative colitis in pregnancy. Am J Gastroenterol 1994; 89:931.
  150. Watson WJ, Gaines TE. Third-trimester colectomy for severe ulcerative colitis. A case report. J Reprod Med 1987; 32:869.
  151. Anderson JB, Turner GM, Williamson RC. Fulminant ulcerative colitis in late pregnancy and the puerperium. J R Soc Med 1987; 80:492.
  152. Dozois EJ, Wolff BG, Tremaine WJ, et al. Maternal and fetal outcome after colectomy for fulminant ulcerative colitis during pregnancy: case series and literature review. Dis Colon Rectum 2006; 49:64.
  153. Dubinsky M, Abraham B, Mahadevan U. Management of the pregnant IBD patient. Inflamm Bowel Dis 2008; 14:1736.
  154. Foulon A, Dupas JL, Sabbagh C, et al. Defining the Most Appropriate Delivery Mode in Women with Inflammatory Bowel Disease: A Systematic Review. Inflamm Bowel Dis 2017; 23:712.
  155. Van Horn C, Barrett P. Pregnancy, delivery, and postpartum experiences of fifty-four women with ostomies. J Wound Ostomy Continence Nurs 1997; 24:151.
  156. Tirath A, Kane SV. Disease Flares in the Postpartum Period: Vigilance Is Key. Inflamm Bowel Dis 2022; 28:484.
  157. Bennett A, Mamunes A, Kim M, et al. The Importance of Monitoring the Postpartum Period in Moderate to Severe Crohn's Disease. Inflamm Bowel Dis 2022; 28:409.
Topic 4083 Version 52.0

References