IMR Press / CEOG / Volume 51 / Issue 2 / DOI: 10.31083/j.ceog5102054
Open Access Review
What Obstetricians Need to Know about Crohn’s Disease—A Narrative Review
Show Less
1 Department of Obstetrics and Gynecology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
2 Department of Obstetrics, Gynecology and Neonatology, “Dr. Alfred Rusescu” National Institute for Maternal and Child Health, 127715 Bucharest, Romania
3 First Department of Surgery, Bucharest Oncological Institute Prof. Dr. Alexandru Trestioreanu, Carol Davila University of Medicine and Pharmacy, 022328 Bucharest, Romania
4 Agrippa Ionescu Hospital Gastroenterology Department, Carol Davila University of Medicine and Pharmacy, 011356 Bucharest, Romania
5 Department of Obstetrics and Gynecology, Filantropia Clinical Hospital, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
*Correspondence: anca.simionescu@umfcd.ro (Anca Angela Simionescu)
Clin. Exp. Obstet. Gynecol. 2024, 51(2), 54; https://doi.org/10.31083/j.ceog5102054
Submitted: 31 July 2023 | Revised: 27 November 2023 | Accepted: 6 December 2023 | Published: 23 February 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Objective: Crohn’s disease (CD) is an immune-mediated inflammatory bowel disease (IBD), which comprises an idiopathic aberrant systemic and local inflammatory response. This response is a result of unknown interactions between the luminal content and the intestinal wall. This article is a review of the current state of knowledge providing information to help obstetricians to manage patients with CD, and to understand the particularities of these patients, with emphasis during pregnancy and postpartum, including recommendation for the birthing methods. It is important to explain the usefulness of the pursue of treatment during pregnancy, taking into consideration the drugs allowed during pregnancy, and addressing the challenges that CD may pose in addition to the physiological adaptations of pregnancy. Mechanism: As both an obstetrician and a gastroenterologist, this topic can be approach from two distinct perspectives. Firstly, how CD influences fertility and pregnancy, and secondly, an exploration on how hormonal changes and immune system tolerance during pregnancy probably influences CD. Findings in Brief: Data shows that pregnancy outcomes are influenced by the clinical course of CD at the time of conception. Latent disease prior to conception is associated with uneventful pregnancies and favorable neonatal outcomes, comparable to general population. Conversely, an active disease during pregnancy and ileal localization can be associated with prematurity, stillbirth, and small-for-gestational age (SGA) infants. A high risk of preeclampsia was reported in pregnancy with severe CD and oral or systemic corticosteroids administration. Optimal management approach involves a multidisciplinary team consisting of an obstetrician, gastroenterologist, and surgeon. Thiopurines and biologic agents are considered safe during pregnancy and breastfeeding. In infants with CD, alteration in the composition of the maternal microbiome may contribute to the systemic inflammation and to influence the transmission of an altered microbiota to the infants. This suggests that modulating the early microbiome can be an effective strategy to reduce cases of CD. Conclusions: Healthcare practitioners and patients must be aware that CD patients can have a successful pregnancy and a healthy infant. A multidisciplinary team can provide supportive care and help address significant information to adapt the treatment plan, and to monitor pregnancy.

Keywords
inflammatory bowel disease
Crohn's disease
pregnancy
perianal fistulae
1. Introduction

Crohn’s disease (CD) is an immune-mediated inflammatory bowel disease (IBD) characterized by an idiopathic aberrant systemic and local inflammatory response, due to and including insufficiently understood interactions between the luminal content and the intestinal wall. CD is a chronic, acquired immune digestive disease, with the onset generally between 15 and 35 years old, coinciding with the reproductive period [1], being thus understood all the concerns regarding the unwanted effects of pregnancy on the underlying disease, but also of CD on the fetus. This condition is characterized by periods of flare/inflammatory activity and quiescent periods.

CD can affect any segment of the gastrointestinal tract, predominantly the terminal ileum and adjacent colon, characterized by a segmental, asymmetric distribution of immune determined granulomatous inflammation [2]. Common clinical manifestation includes abdominal pain and tenderness, fatigue, fever, weight loss, malnutrition, chronic diarrhea, as well as perineal and perianal lesions such as hemorrhoids, skin tags, fissures, abscesses, ulcers, fistulae, lymphoedema, strictures. Extradigestive symptoms are represented by anemia, specific granulomatous skin lesions, and spondyloarthritis [3, 4, 5]. It is characterized by periods of remission alternating with relapses, with surgical intervention sometimes necessary [6]. A population-based review has demonstrated that, for a majority of patients, CD is a progressive and destructive disease, particularly when located in the ileum and ileocolonic regions. Risk factors for progression include onset at age <40 years, perianal disease, and need for steroid treatment [7].

In the active phase of CD, pregnancy poses a higher risk of miscarriage, preterm delivery, poor maternal weight gain and small-for-gestational age (SGA) babies, risk of need for Cesarean section (CS) [8, 9, 10, 11, 12, 13]. A high risk of preeclampsia has been reported in pregnancies with severe CD and the administration of oral or systemic corticosteroids administration [14]. In the past, due to the necessity for control of CD activity and mitigating pregnancy complications, concerns about the risk of surgery during pregnancy and to the medications prescribed for IBD, it was believed that pregnancy was contraindicated for women suffering from CD [15].

The study on pregnant women with uncontrolled CD is very limited, given such cases are not very common [16]. Moreover, small series focusing on pregnancy with ileostomy have reported uneventful pregnancies, with very rare stoma complications, including bleeding, retraction, stenosis, laceration, prolapse, hernia, or intestinal obstructions [17].

This article is a review on the current knowledge about women with CD who wish to conceive or who are already pregnant. It is also important to mention that many of the studies discussed in the literature were not exclusively focused on CD, but on IBD in general. Medical management, including all the therapies for CD during pregnancy and breastfeeding, will not be discussed here because these aspects are well covered by recent guidelines [18, 19, 20].

2. Pathogenesis of CD and Interaction with the Immunity of Pregnancy

These multifactorial pathologies arise from the interplay between an altered response of the immune system and an abnormal intestinal microflora, culminating in chronic inflammation of the digestive tract. Medical experts are still trying to discover the exact etiopathogenesis of CD, but so far this has not been successful. However, it was found that genetic susceptibility plays an extremely important role, and up to 200 genetic loci associated with an increased risk of developing these pathologies have been identified [21, 22]. Studying these loci highlighted the role of innate immunity and bacterial implication in IBD etiology through numerous genes that are involved in innate immune cell function, adaptive immunity, or epithelial barrier function [22].

From a structural point of view, the contact area is represented by the following components: bacteria, mucous layer, antimicrobial peptides, and barrier cells. Inflammatory pathologies involve alterations in the protective mucus layer [23]. Through these openings that form in the mucous layer, bacteria can more readily reach the epithelial layer, resulting in inflammatory responses that lead to the release of cytokines, such as interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). These cytokines exacerbate apoptotic processes, resulting in weakening of the epithelial layer and an exaggerated, inadequate migration of bacteria [24]. Finally, after the weakening of the barrier function of the intestinal epithelium, there is an exposure of the mucosa to bacteria, which attracts immune cells and leads to a self-perpetuating pro-inflammatory status. Throughout disease progression, the process of inflammation is induced by type 1 T helper (Th1) cytokines, CD4+ T cells, macrophages and Th3 lymphocytes activation in the intestinal inflammatory tissue. CD4+ T cells is represented by Forkhead box P3 (Foxp3): Foxp3+CD4+ Treg cells, Th3, and Th17. Th17 cells have protective role against extracellular bacteria and may also play a role in inflammation. Th17 cells activation results in the formation of intestinal granulomas. Some antigens produced by bacteria can induce CD4+ T cells to differentiate into targeted cytotoxic T lymphocytes (CTLs). These cells release interleukin (IL)-17 to stimulate Th17 cells to produce transforming growth factor and interferon-α (IFN-α), which can cause persistent inflammation and fibrosis [25]. Intestinal immunity is controlled by regulatory T (Treg) cells that are considered to be responsible for the pathogenesis of IBD. This involvement in IBD is by a decreased number of peripheral Treg cells and abnormal differentiation [26].

In pregnancy, there are immune shifts to facilitate alloimmune fetal tolerance, the maternal immune response changes from the inflammatory Th1 cytokine pattern to the Th2 pattern. The maintenance of pregnancy and feto-maternal tolerance is associated with Th2 and Th17/Th2 cells, and CD4+CD25+Foxp3+ Treg cells. Th1-type and Th17-type cytokines that promote fetal semi-allograft rejection may compromise pregnancy, whereas Th2-type cytokines may improve pregnancy outcomes. Treg cells play an essential role in the induction of paternal antigen tolerance early in pregnancy, by suppressing immune responses caused by paternal and fetal antigen recognition. Late in pregnancy, the occurrence of preeclampsia may be explained by the disturbance of paternal antigen-specific tolerance, marked by a decrease in clonal populations of decidual effector Treg cells [27]. The immunology of pregnancy and feto-maternal immune tolerance is related to CD4+ T cell cytokines [28, 29]. T cell responses in immune and autoimmune disease are influenced by the state of pregnancy.

During pregnancy, in the first-trimester, there is a marked and rapid increase in the levels of estrogen and progesterone. The maximum levels of estrogens are reached in the third-trimester to ensure the necessary support for the development of the placenta and the vascularization of the uterus, facilitating normal fetal development. A study shows that low levels of 17β-estradiol decrease the paracellular permeability of vascular endothelial cells, while increased levels of estrogen increase permeability, due to the biphasic modulation of the tight junction molecule occluding [30]. The additional study shows that gut epithelial cells in male rats exhibit less resistant to injury than in female rats, and that application of estrogen to male gut cells results in an obvious decrease in the inflammatory susceptibility [31]. Taking all of these into account, it can be concluded that estrogens play a protective role in digestive inflammatory pathologies, such as CD and thus pregnancy does not represent a risk factor through its hormonal load; on the contrary, it displays a ‘potential’ protective factor for women with this pathology [32]. Another interesting fact is the role of human chorionic gonadotropin (hCG) in autoimmune diseases such as rheumatoid arthritis (RA), Sjogren syndrome, and IBD. It was proven that hCG mediates early expression of regulatory T cells, reduces the level of inflammatory cytokines such as IL-17, and increases the IL-10 levels [33]. Thus, administering hCG has been demonstrated to prevent the occurrence of autoimmune diabetes in mice by downregulating the Th1 response [34]. A decrease in the level of inflammatory markers such as IL-6 and TNF-α, and the recruitment of neutrophils were also demonstrated in patients that were administered hCG peptides [33, 35]. The recently reported subset of T follicular helper cells (Tfh) is implicated in the pathogenesis of various autoimmune disorders, including CD, which is associated with humoral immune responses [36].

Given the immune mechanisms described earlier, Th1- and Th17-type autoimmune disorders during pregnancy are associated with a lower risk of progression and a reduced rate of exacerbation when there is a rise in Th2-type cytokines in pregnancy (e.g., CD, RA, Graves disease, and multiple sclerosis). Conversely, Th2-type autoimmune disorders could worsen in pregnancy with an increase in Th2-type cytokines (e.g., psoriasis, lupus nephritis, systemic lupus erythematosus (SLE)). Furthermore, in the postpartum period, inflammatory and autoimmune disorders may worsen due to the loss of the immunosuppressive state of pregnancy and the decrease in estrogen levels. This decrease can lead to a reduction in Th2-type cytokines along with the decrease of Th1 and Th17-type.

3. Menstrual Cycle, Sexual Dysfunction and Fertility in CD

Autoimmune diseases have been associated with a high frequency of flares and exacerbation of autoimmune disease symptomatology due to the changes in immune hormones modulated by the chronic disease, which modifies Th1/Th2 immunity [37]. In IBD populational studies, menstrual period is associated with dysmenorrhea, especially one year before the diagnosis of CD [38, 39].

The reason for sexual dysfunction among CD patients is considered to be multifactorial, including psychological factors related to CD manifestations, such as an altered perception of body image, followed by disease-specific factors, surgical interventions, osteotomy, medical therapies, pelvic floor disorders and hypogonadism, and social and cultural traits of personality [40]. Due to these reasons along with the higher incidence of depressive symptoms in these patients, a significantly lower rate of sexual contact is reported among them compared to a control group [41]. A study shows that in general this is not a matter of structural impotence, reinforced by an objectified defect, but rather a greater difficulty in achieving orgasm, deep and increased dyspareunia [42]. Considering this significant impact on sexual life, not only will the quality of life be affected, but also the conception rate. The causes for increased sexual dysfunction should be discussed together with the patient, and efforts should be made to find a solution through treatment, surgery, and psychotherapy.

Women with CD, especially those who underwent surgical treatment, have shown lower fertility rates compared to the control group, potentially due to inflammation and scarring of the fallopian tubes [10, 43, 44]. In cases of women with CD in remission, who have never undergone surgical treatment, fertility rates are age-dependent and almost equal the general population. Optimizing disease management may help conception [43]. However, there are a multitude of variables that can affect fertility, which include the status of the disease, prior surgical interventions, and the medical treatment approached. In terms of the type of intervention, there are no studies that demonstrate a better fertility rate for the laparoscopic approach than the classic one, except in the case of ileal pouch-anal anastomosis [45, 46, 47].

The studies about assisted reproduction techniques (ART) in CD show a similar success rate of in vitro fertilization (IVF) procedures among patients with CD and those without this pathology [48, 49]. IBD patients over 30, who have undergone surgical interventions and have been unsuccessful in conceiving for 6 months, should be recommended a fertility evaluation and consider ART [50, 51, 52]. It is important to note that there is a study that shows that medications for inflammatory digestive pathologies do not impact the effectiveness of ART or egg freezing, nor do the hormones used for increasing the success of ART do not lead to a destabilization of the underlying disease [19].

Nonetheless, a period of remission of 3 to 6 months until conception is recommended to reduce the risk of relapse or flare-up during pregnancy and the postpartum period [53]. For this purpose, oral contraceptives, hormonal or non-hormonal intrauterine devices, or contraceptive implants can be recommended. It should also be considered that oral contraceptives are not effective in the case of active small bowel inflammation or extensive small bowel resection [54]. Caution must be taken in patients with personal or family history of deep venous thrombosis or pulmonary embolism.

Table 1 (Ref. [1, 19]) summarize the main idea about fertility and contraception in women with CD.

Table 1.Fertility and contraception in women with CD.
Fertility Active disease decrease fertility.
Decreased fertility in patients with ileal pouch anal anastomosis.
ART are recommended if, after 6 months of unprotected sexual contact, the couple does not get pregnant.
Contraception Long-acting, non-estrogen-containing contraception in patients with active disease due to the risk of venous thromboembolism [19].
Absorption of oral contraceptives is almost normal in patients with mild CD or short ileal resections.
In cases of extensive small bowel resection or active disease, non-oral contraceptives are recommended [1].

CD, Crohn’s disease; ART, assisted reproduction techniques.

4. Pregnancy and CD: Planning Pregnancy and Monitoring

The greatest priority during pregnancy is to control the underlying disease as best as possible, so that no symptoms or relapse appear. This is generally dictated by the status of the disease at the time of conception. A disease in remission at that time not only facilitates conception but also helps in maintaining control during all trimesters of pregnancy, especially in cases of perineal pain or discharge. Table 2 (Ref. [18, 19, 55]) present the main recommendations about pregnancy planning and treatment during pregnancy, including education and nutritional status in case of CD during pregnancy. The follow-up of pregnancy in cases of CD in remission is identical as in high-risk pregnancies, with gestational weight and blood pressure careful monitoring.

Table 2.The main recommendations for CD before planning pregnancy, pregnancy monitoring and treatment during pregnancy [18, 19, 55].
Screening before conceiving Papanicolaou smear.
Vaccination with all necessary non-live vaccines prior to pregnancy: diphtheria, tetanus, pertussis (dTpa), and influenza.
Live vaccination is contraindicated if the patient is receiving immunosuppressive treatment.
Alcohol, smoking, and illicit drugs cessation.
Education Heredity of IBD.
Mode of delivery.
Safety of ART when needed.
Adequate gestational weight gain.
Disease flare and advice when is important to make an appointment to the doctor.
Safety of breastfeeding.
Disease management Three months of corticosteroid-free remission.
Confirm remission based on clinically, biochemical, intestinal ultrasonography, magnetic resonance imaging, and radiological assessment.
Recognition of ileostomy complications if present.
Medication during pregnancy Low risk medication:
Mesalazine/sulphasalazine (but these are not currently recommended for CD management, only in ulcerative colitis)
Corticosteroids (only if needed for a flare, as remission free corticosteroid should be achieved at least 3 months before conception)
Azathioprine (may be stopped only if prescribed in association with infliximab and already in long term remission, together with assessing infliximab through levels)
Anti-TNF-α (infliximab, adalimumab and biosimilars)—are considered safe during pregnancy, labor, breastfeeding; may consider continuing to the end of pregnancy, while early discontinuation may increase the risk of relapse
Limited data about medication:
Vedolizumab, ustekinumab (should be probably continued, as antiTNF-α)
Ciclosporin (only used for induction of remission, in active flare, not suited for conception time)
Contraindicated:
Metotrexate
Tofacitinib
Thalidomide
Filgotinib
Ozanimond
Nutritional status 2–5 mg daily dose of folate for cases with fiber-restricted diet, extensive ileal disease or those taking sulfasalazine for three months prior to conception, or in the first 12 weeks of pregnancy.
Normal range for iron, vitamin B12, and folate before conception.

CD, Crohn’s disease; IBD, inflammatory bowel disease; TNF-α, tumor necrosis factor factor-α.

The pregnancy follow-up in case of CD in remission is identical as in high-risk pregnancies, with careful monitoring of gestational weight and blood pressure. Clinical examination of the perineum and checking for clinical symptoms may help confirm or exclude CD relapse. Fig. 1 shows two perianal fistulas diagnosed at 30 weeks of pregnancy in a patient with CD.

Fig. 1.

Perianal fistula in a primipara primigravida with CD at 30 weeks of gestation after surgical treatment: Fistulectomy associated with slow total sphincterotomy, using loose setons (Silvestri). CD, Crohn’s disease.

More than half of CD patients with perianal lesions have multiple lesions [56]. Fig. 2 shows typical lesions at colonoscopy in a patient with CD.

Fig. 2.

Colonoscopy aspect of CD: a narrow colonic stricture with ulcerated mucosa. CD, Crohn’s disease.

Complete blood count for hemoglobin level, C-reactive protein (CRP) and fecal calprotectin (FC) for disease activity, B12 level, liver enzymes, and albumin are performed in every trimester of pregnancy. A careful evaluation of fetal growth is required during every visit because patients suffering from CD are prone to malnutrition, which can be difficult to correct during the early stages of pregnancy when placental development takes place rapidly, and in the last trimester when the nutrient requirement of the fetus increases. Serum albumin levels are an important marker of nutritional status and of CD activity [57, 58, 59, 60], but these levels may decrease during pregnancy due to physiological hemodilution [61]. Additionally, the guidelines recommend that women with CD be closely investigated regarding vitamin D level, folic acid level, micronutrient status, micronutrient deficiencies, and ferritin levels. Specific guidelines about clinical nutrition counselling and recommendations in CD have been updated [62]. A study conducted by the Epidemiology Committee (EpiCom) of the European Crohn’s and Colitis Organization (ECCO) demonstrated that there is no difference in the evolution of the disease in pregnant versus non-pregnant women with CD [53]. There are studies that reinforce this idea, demonstrating that patients with active disease at the time of conception are more likely to have an active disease during pregnancy, than those who conceive when in remission [63, 64, 65]. It was also demonstrated that pregnancies that occur during the period of active disease are more subject to obstetric complications, such as preterm birth, SGA infants, and spontaneous abortion. For this reason, the ECCO guidelines recommend achieving remission before getting pregnant [19, 66].

Additionally, recommendations for the 9 months of pregnancy are provided, from which the women with IBD must not deviate for a favorable evolution of the pregnancy. In their study, Oron et al. [67] concluded that patients with IBD who gained less than 12 kg during pregnancy frequently had complications, such as premature birth, SGA infants, and the need for neonatal intensive care. A study from 2017 also showed that women with CD had inadequate gestational weight gain (34.3%) more often than pregnant women without IBD (19.4%) [68].

Furthermore, meta-analysis has demonstrated an increased risk of gestational diabetes in the case of CD-associated pregnancies [69]. The theory that this occurs more often due to corticosteroid treatment has been proposed, but this connection is still questionable. Many authors recommend for early screening of gestational diabetes in patients undergoing corticotherapy treated during pregnancy [70]. There is no clear consensus regarding the association between steroids exposure and gestational diabetes [69, 70]. Patients with active IBD are more prone to severe forms of preeclampsia [63], as well as venous thromboembolism complication than in the general population. This is more elevated, especially antepartum and in the 12-week postpartum period, compared with women without IBD [14]. As such, there are several recommendations for thromboprophylaxis in CD in pregnancy. The British Royal College of Gynecologists and Obstetricians recommend venous thromboembolism prophylaxis for pregnant women with IBD who had one or more episodes of active disease during the third-trimester of pregnancy [71]. The Canadian Association of Gastroenterology recommends that women with IBD who have undergone a CS, at least be given anticoagulants during pregnancy and, if there is a history of venous thromboembolism, up to 6 weeks after birth [72].

Complex and multiple perianal fistulas in pregnancy can be extremely debilitating for women, requiring medical and surgical treatment. Perianal fistulas may be treated in order to avoid perianal abscess and premature delivery. During pregnancy, medical interventions such as antibiotherapy are employed, while in postpartum period, immunomodulators and biologic treatment may be used.

Joint management with specialist surgeon is required if perianal disease becomes active in pregnancy. In case of perianal fistulas, the methods of surgical treatment are as follows: fistulotomy with partial sphincterotomy, use of fibrin adhesives, anal fistula plug, endorectal advancement flap, and intersphincteric fistula tract ligation procedure. All these procedures have very low efficiency in patients with CD. If the patient has active proctitis and pelvic abscess, it is recommended to only drain the abscesses and place the free seton, given the potential risks of iatrogenic injuries and delayed wound healing [73].

The use of setons keeps the fistulas open, allowing fluid to escape and prevent abscess formation. The setons delay the healing and closure of the fistula. Its use during treatment with infliximab can prevent an abscess from forming. Currently, uncut setons are used because the ‘cutting’ setons, which, when sequentially tightened at regular intervals, caused damage to the sphincter and anal incontinence [74].

5. Types of Delivery

The most common perineal manifestations are perineal fistulae and abscesses, as well as anal fistulas of rectovaginal type [75, 76, 77, 78, 79]. A higher incidence of CS was founded in women with CD when compared to women without CD, but for obstetrical factors [80]. Recent data and current guidelines suggest that CS is recommended for patients with active perineal disease or rectovaginal fistulae [81, 82]. While there is no CS recommendation for patients with CD without perineal disease and without relapse, obstetrical factors must be considered [18, 19, 20, 83]. Episiotomy should be avoided if possible [84]. Cheng et al. [84] have reported on a case-control series involving 11 patients with vaginal delivery compared with 50 patients CS, showing no differences for perianal disease; the rate of perianal flare was found to be similar in pregnant and non-pregnant CD control cases. Approximately two-thirds of CS delivery cases had a symptomatic perianal flare within 2 years after delivery, compared to 45% for vaginal delivery [84]. It should also be considered that for these women, the choice regarding the type of birth has implications both for the current pregnancy and for subsequent ones. It is also important to mention that none of the birth methods are ideal and free of possible complications. In these cases, vaginal birth can be associated with perineal trauma, sphincter injury, or perineal fistulae [85], while cesarean birth should be avoided as much as possible due to the risk of adhesion formation in patients who often need subsequent surgical treatment for the underlying pathology. In approximately 50% of cases with CD after several years of evolution, the perineal region is also involved, prompting questions regarding the indicated mode of birth.

The risk of 3rd and 4th degree ruptures is not higher in patients with IBD after vaginal birth [86].

6. The Role of Microbiota

It has been recently hypothesized that the gut microbiota is a key factor in the pathogenesis of CD [87]. Newborns whose mothers have CD develop this pathology more often than the rest of the population, suggesting that the microbiota also plays an important role in this pathology. A pregnant woman with CD has an altered microbiota, transmitting this altered microbiota to the newborn, which will be the ideal environment for the baby to develop the same pathology. Besides this direct transmission, there are also indirect factors, such as maternal diet, feeding behavior, or antibiotic exposure that increase the risk of developing IBD [88, 89, 90, 91, 92]. Also, due to the mother’s pathology, girls are more often born by CS and are exclusively breastfed less often than the general population, and these two factors contribute to the development of an altered microbiota [93, 94]. A study from 2020 showed that babies born from mothers with IBD have a higher abundance of proinflammatory bacteria like Proteobacteria and a lower level than the control group of Bifidobacteria, which are generally part of the microbiome of the eutrophic newborn [95]. There are also studies that suggest a female predominance for familial CD and IBD, a female sex-specific epigenetic inheritance pattern and higher transmission rate from mothers with CD compared to fathers [96, 97].

Considering all of the above, it would appear that in the future, modulating the early microbiome could be an effective strategy to reduce cases of CD. Breastfeeding is crucial and should be encouraged.

7. Postpartum Management

It is extremely important that the baby is closely monitored even after birth, through a follow-up with clinical, biochemical, and psychological assessment. Within the first 12 months postpartum, 30% of patients have a flare, most frequently for those who reduced IBD therapy during or immediately following pregnancy, or for those with active disease in the third-trimester [98, 99]. During breastfeeding, all IBD medications taken during pregnancy should be continued. The levels of these medications in breastmilk are very low, so they are safe during lactation, with negligible effect on the infant [18]. Live vaccines (Bacille Calmette-Guérin (BCG), rotavirus, measles, mumps, and rubella) are postponed for infants <12 month exposed to biologic therapy during the third-trimester of pregnancy, until at least after 6 months because of the risk of transplacental transfer [100].

8. Conclusions

Healthcare practitioners and patients must be aware that CD patients can have a successful pregnancy and a healthy infant. During pre-conception, prenatal, and postpartum periods, a multidisciplinary team can provide supportive care and help address significant information to adapt the treatment plan, and balance benefits to outweigh risks in preventing flares. Biologic therapies are demonstrated to be safe during pregnancy, labor, and breastfeeding and it must be continued throughout. Vaginal birth and breastfeeding must be encouraged, since CS is recommended to patients with active perineal disease. Attention must be paid to live vaccination after biologic therapies in the third-trimester of pregnancy, live vaccines are contraindicated in infants at least 6 months postpartum due to the risk of placental transfer. Lastly, live vaccination is contraindicated in mothers receiving immunosuppressive treatment.

Author Contributions

AAS and BMD designed the research study. AAS, BMD, AMM and MC performed the research. AAS, BMD, AMM and MC analyzed the data. BMD and AAS wrote the manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics Approval and Consent to Participate

Written informed consent has been obtained from the patients from Figs. 1,2 to publish these pictures.

Acknowledgment

Not applicable.

Funding

This research received no external funding.

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Ng SC, Shi HY, Hamidi N, Underwood FE, Tang W, Benchimol EI, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. 2017; 390: 2769–2778.
[2]
Strober W, Fuss I, Mannon P. The fundamental basis of inflammatory bowel disease. The Journal of Clinical Investigation. 2007; 117: 514–521.
[3]
Sairenji T, Collins KL, Evans DV. An Update on Inflammatory Bowel Disease. Primary Care. 2017; 44: 673–692.
[4]
Cozzi G, Scagnellato L, Lorenzin M, Savarino E, Zingone F, Ometto F, et al. Spondyloarthritis with inflammatory bowel disease: the latest on biologic and targeted therapies. Nature Reviews. Rheumatology. 2023; 19: 503–518.
[5]
Crohn’s & Colitis Australia. Final report of the first audit of the organisation and provision of IBD services in Australia. 2016. Available at: https://crohnsandcolitis.org.au/wp-content/uploads/2022/02/Final_Web_Audit_17-Updated-3-Feb-2.pdf (Accessed: 4 November 2023).
[6]
Lamb CA, Kennedy NA, Raine T, Hendy PA, Smith PJ, Limdi JK, et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut. 2019; 68: s1–s106.
[7]
Tarrant KM, Barclay ML, Frampton CMA, Gearry RB. Perianal disease predicts changes in Crohn’s disease phenotype-results of a population-based study of inflammatory bowel disease phenotype. The American Journal of Gastroenterology. 2008; 103: 3082–3093.
[8]
Fonager K, Sørensen HT, Olsen J, Dahlerup JF, Rasmussen SN. Pregnancy outcome for women with Crohn’s disease: a follow-up study based on linkage between national registries. The American Journal of Gastroenterology. 1998; 93: 2426–2430.
[9]
Stephansson O, Larsson H, Pedersen L, Kieler H, Granath F, Ludvigsson JF, et al. Crohn’s disease is a risk factor for preterm birth. Clinical Gastroenterology and Hepatology. 2010; 8: 509–515.
[10]
Cornish J, Tan E, Teare J, Teoh TG, Rai R, Clark SK, et al. A meta-analysis on the influence of inflammatory bowel disease on pregnancy. Gut. 2007; 56: 830–837.
[11]
Stephansson O, Larsson H, Pedersen L, Kieler H, Granath F, Ludvigsson JF, et al. Congenital abnormalities and other birth outcomes in children born to women with ulcerative colitis in Denmark and Sweden. Inflammatory Bowel Diseases. 2011; 17: 795–801.
[12]
Mahadevan U, Sandborn WJ, Li DK, Hakimian S, Kane S, Corley DA. Pregnancy outcomes in women with inflammatory bowel disease: a large community-based study from Northern California. Gastroenterology. 2007; 133: 1106–1112.
[13]
Molnár T, Farkas K, Nagy F, Lakatos PL, Miheller P, Nyári T, et al. Pregnancy outcome in patients with inflammatory bowel disease according to the activity of the disease and the medical treatment: a case-control study. Scandinavian Journal of Gastroenterology. 2010; 45: 1302–1306.
[14]
Boyd HA, Basit S, Harpsøe MC, Wohlfahrt J, Jess T. Inflammatory Bowel Disease and Risk of Adverse Pregnancy Outcomes. PLoS ONE. 2015; 10: e0129567.
[15]
Imrie AH. Pregnancy and Crohn’s disease. British Medical Journal. 1970; 2: 299.
[16]
Germain A, Chateau T, Beyer-Berjot L, Zerbib P, Lakkis Z, Amiot A, et al. Surgery for Crohn’s disease during pregnancy: A nationwide survey. United European Gastroenterology Journal. 2020; 8: 736–740.
[17]
Whiteley I, Gullick J. The embodied experience of pregnancy with an ileostomy. Journal of Clinical Nursing. 2018; 27: 3931–3944.
[18]
Torres J, Chaparro M, Julsgaard M, Katsanos K, Zelinkova Z, Agrawal M, et al. European Crohn’s and Colitis Guidelines on Sexuality, Fertility, Pregnancy, and Lactation. Journal of Crohn’s & Colitis. 2023; 17: 1–27.
[19]
Mahadevan U, Robinson C, Bernasko N, Boland B, Chambers C, Dubinsky M, 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: 1508–1524.
[20]
Nguyen GC, Seow CH, Maxwell C, Huang V, Leung Y, Jones J, et al. The Toronto Consensus Statements for the Management of Inflammatory Bowel Disease in Pregnancy. Gastroenterology. 2016; 150: 734–757.e1.
[21]
Cleynen I, Boucher G, Jostins L, Schumm LP, Zeissig S, Ahmad T, et al. Inherited determinants of Crohn’s disease and ulcerative colitis phenotypes: a genetic association study. Lancet. 2016; 387: 156–167.
[22]
Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012; 491: 119–124.
[23]
Cornick S, Tawiah A, Chadee K. Roles and regulation of the mucus barrier in the gut. Tissue Barriers. 2015; 3: e982426.
[24]
Shen L, Su L, Turner JR. Mechanisms and functional implications of intestinal barrier defects. Digestive Diseases. 2009; 27: 443–449.
[25]
Li N, Shi RH. Updated review on immune factors in pathogenesis of Crohn’s disease. World Journal of Gastroenterology. 2018; 24: 15–22.
[26]
Yamada A, Arakaki R, Saito M, Tsunematsu T, Kudo Y, Ishimaru N. Role of regulatory T cell in the pathogenesis of inflammatory bowel disease. World Journal of Gastroenterology. 2016; 22: 2195–2205.
[27]
Tsuda S, Nakashima A, Shima T, Saito S. New Paradigm in the Role of Regulatory T Cells During Pregnancy. Frontiers in Immunology. 2019; 10: 573.
[28]
Simionescu AA, Danciu BM, Stanescu AMA. State-of-the-Art Review of Pregnancy-Related Psoriasis. Medicina. 2021; 57: 804.
[29]
Piccinni MP, Lombardelli L, Logiodice F, Kullolli O, Parronchi P, Romagnani S. How pregnancy can affect autoimmune diseases progression? Clinical and Molecular Allergy. 2016; 14: 11.
[30]
Ye L, Martin TA, Parr C, Harrison GM, Mansel RE, Jiang WG. Biphasic effects of 17-beta-estradiol on expression of occludin and transendothelial resistance and paracellular permeability in human vascular endothelial cells. Journal of Cellular Physiology. 2003; 196: 362–369.
[31]
Homma H, Hoy E, Xu DZ, Lu Q, Feinman R, Deitch EA. The female intestine is more resistant than the male intestine to gut injury and inflammation when subjected to conditions associated with shock states. American Journal of Physiology. Gastrointestinal and Liver Physiology. 2005; 288: G466–G472.
[32]
Kane S. IBD: Activity of IBD during pregnancy. Nature Reviews. Gastroenterology & Hepatology. 2013; 10: 571–572.
[33]
Koldehoff M, Katzorke T, Wisbrun NC, Propping D, Wohlers S, Bielfeld P, et al. Modulating impact of human chorionic gonadotropin hormone on the maturation and function of hematopoietic cells. Journal of Leukocyte Biology. 2011; 90: 1017–1026.
[34]
Khil LY, Jun HS, Kwon H, Yoo JK, Kim S, Notkins AL, et al. Human chorionic gonadotropin is an immune modulator and can prevent autoimmune diabetes in NOD mice. Diabetologia. 2007; 50: 2147–2155.
[35]
van den Berg HR, Khan NA, van der Zee M, Bonthuis F, IJzermans JN, Dik WA, et al. Synthetic oligopeptides related to the [beta]-subunit of human chorionic gonadotropin attenuate inflammation and liver damage after (trauma) hemorrhagic shock and resuscitation. Shock. 2009; 31: 285–291.
[36]
Mesquita D, Jr, Cruvinel WM, Resende LS, Mesquita FV, Silva NP, Câmara NOS, et al. Follicular helper T cell in immunity and autoimmunity. Brazilian Journal of Medical and Biological Research. 2016; 49: e5209.
[37]
Oertelt-Prigione S. Immunology and the menstrual cycle. Autoimmunity Reviews. 2012; 11: A486–A492.
[38]
Saha S, Zhao YQ, Shah SA, Esposti SD, Lidofsky S, Salih S, et al. Menstrual cycle changes in women with inflammatory bowel disease: a study from the ocean state Crohn’s and colitis area registry. Inflammatory Bowel Diseases. 2014; 20: 534–540.
[39]
Kane SV, Sable K, Hanauer SB. The menstrual cycle and its effect on inflammatory bowel disease and irritable bowel syndrome: a prevalence study. The American Journal of Gastroenterology. 1998; 93: 1867–1872.
[40]
Perez de Arce E, Quera R, Ribeiro Barros J, Yukie Sassaki L. Sexual Dysfunction in Inflammatory Bowel Disease: What the Specialist Should Know and Ask. International Journal of General Medicine. 2021; 14: 2003–2015.
[41]
Roseira J, Magro F, Fernandes S, Simões C, Portela F, Vieira AI, et al. Sexual Quality of Life in Inflammatory Bowel Disease: A Multicenter, National-Level Study. Inflammatory Bowel Diseases. 2020; 26: 746–755.
[42]
Nøhr EA, Nielsen J, Nørgård BM, Friedman S. Sexual Health in Women with Inflammatory Bowel Disease in the Danish National Birth Cohort. Journal of Crohn’s & Colitis. 2020; 14: 1082–1089.
[43]
Ban L, Tata LJ, Humes DJ, Fiaschi L, Card T. Decreased fertility rates in 9639 women diagnosed with inflammatory bowel disease: a United Kingdom population-based cohort study. Alimentary Pharmacology & Therapeutics. 2015; 42: 855–866.
[44]
Waljee A, Waljee J, Morris AM, Higgins PDR. Threefold increased risk of infertility: a meta-analysis of infertility after ileal pouch anal anastomosis in ulcerative colitis. Gut. 2006; 55: 1575–1580.
[45]
Lee S, Crowe M, Seow CH, Kotze PG, Kaplan GG, Metcalfe A, et al. The impact of surgical therapies for inflammatory bowel disease on female fertility. The Cochrane Database of Systematic Reviews. 2019; 7: CD012711.
[46]
Bartels SAL, DʼHoore A, Cuesta MA, Bensdorp AJ, Lucas C, Bemelman WA. Significantly increased pregnancy rates after laparoscopic restorative proctocolectomy: a cross-sectional study. Annals of Surgery. 2012; 256: 1045–1048.
[47]
Beyer-Berjot L, Maggiori L, Birnbaum D, Lefevre JH, Berdah S, Panis Y. A total laparoscopic approach reduces the infertility rate after ileal pouch-anal anastomosis: a 2-center study. Annals of Surgery. 2013; 258: 275–282.
[48]
Laube R, Tran Y, Paramsothy S, Leong RW. Assisted Reproductive Technology in Crohn’s Disease and Ulcerative Colitis: A Systematic Review and Meta-Analysis. The American Journal of Gastroenterology. 2021; 116: 2334–2344.
[49]
Hernandez-Nieto C, Sekhon L, Lee J, Gounko D, Copperman A, Sandler B. Infertile patients with inflammatory bowel disease have comparable in vitro fertilization clinical outcomes to the general infertile population. Gynecological Endocrinology. 2020; 36: 554–557.
[50]
Fréour T, Miossec C, Bach-Ngohou K, Dejoie T, Flamant M, Maillard O, et al. Ovarian reserve in young women of reproductive age with Crohn’s disease. Inflammatory Bowel Diseases. 2012; 18: 1515–1522.
[51]
Şenateş E, Çolak Y, Erdem ED, Yeşil A, Coşkunpınar E, Şahin Ö, et al. Serum anti-Müllerian hormone levels are lower in reproductive-age women with Crohn’s disease compared to healthy control women. Journal of Crohn’s & Colitis. 2013; 7: e29–e34.
[52]
Practice Committee of American Society for Reproductive Medicine. Diagnostic evaluation of the infertile female: a committee opinion. Fertility and Sterility. 2012; 98: 302–307.
[53]
Pedersen N, Bortoli A, Duricova D, D Inca R, Panelli MR, Gisbert JP, et al. The course of inflammatory bowel disease during pregnancy and postpartum: a prospective European ECCO-EpiCom Study of 209 pregnant women. Alimentary Pharmacology & Therapeutics. 2013; 38: 501–512.
[54]
Centers for Disease Control and Prevention (CDC). U S. Medical Eligibility Criteria for Contraceptive Use, 2010. MMWR. Recommendations and Reports: Morbidity and Mortality Weekly Report. Recommendations and Reports. 2010; 59: 1–86.
[55]
Dorney E, Black KI. Preconception care. Australian Journal of General Practice. 2018; 47: 424–429.
[56]
Geltzeiler CB, Wieghard N, Tsikitis VL. Recent developments in the surgical management of perianal fistula for Crohn’s disease. Annals of Gastroenterology. 2014; 27: 320–330.
[57]
Qin G, Tu J, Liu L, Luo L, Wu J, Tao L, et al. Serum Albumin and C-Reactive Protein/Albumin Ratio Are Useful Biomarkers of Crohn’s Disease Activity. Medical Science Monitor. 2016; 22: 4393–4400.
[58]
Khan N, Patel D, Shah Y, Trivedi C, Yang YX. Albumin as a prognostic marker for ulcerative colitis. World Journal of Gastroenterology. 2017; 23: 8008–8016.
[59]
Yang DH, Yang SK, Park SH, Lee HS, Boo SJ, Park JH, et al. Usefulness of C-reactive protein as a disease activity marker in Crohn’s disease according to the location of disease. Gut and Liver. 2015; 9: 80–86.
[60]
Kammerlander H, Nielsen J, Kjeldsen J, Knudsen T, Gradel KO, Friedman S, et al. Fecal Calprotectin During Pregnancy in Women With Moderate-Severe Inflammatory Bowel Disease. Inflammatory Bowel Diseases. 2018; 24: 839–848.
[61]
Abbassi-Ghanavati M, Greer LG, Cunningham FG. Pregnancy and laboratory studies: a reference table for clinicians. Obstetrics and Gynecology. 2009; 114: 1326–1331.
[62]
Bischoff SC, Bager P, Escher J, Forbes A, Hébuterne X, Hvas CL, et al. ESPEN guideline on Clinical Nutrition in inflammatory bowel disease. Clinical Nutrition. 2023; 42: 352–379.
[63]
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. Alimentary Pharmacology & Therapeutics. 2013; 38: 460–466.
[64]
de Lima-Karagiannis A, Zelinkova-Detkova Z, van der Woude CJ. The Effects of Active IBD During Pregnancy in the Era of Novel IBD Therapies. The American Journal of Gastroenterology. 2016; 111: 1305–1312.
[65]
Rottenstreich A, Fridman Lev S, Rotem R, Mishael T, Grisaru Granovsky S, Koslowsky B, et al. Disease flare at prior pregnancy and disease activity at conception are important determinants of disease relapse at subsequent pregnancy in women with inflammatory bowel diseases. Archives of Gynecology and Obstetrics. 2020; 301: 1449–1454.
[66]
Kim MA, Kim YH, Chun J, Lee HS, Park SJ, Cheon JH, et al. The Influence of Disease Activity on Pregnancy Outcomes in Women With Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Journal of Crohn’s & Colitis. 2021; 15: 719–732.
[67]
Oron G, Yogev Y, Shcolnick S, Hod M, Fraser G, Wiznitzer A, et al. Inflammatory bowel disease: risk factors for adverse pregnancy outcome and the impact of maternal weight gain. The Journal of Maternal-fetal & Neonatal Medicine. 2012; 25: 2256–2260.
[68]
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. Inflammatory Bowel Diseases. 2017; 23: 1225–1233.
[69]
Tandon P, Govardhanam V, Leung K, Maxwell C, Huang V. Systematic review with meta-analysis: risk of adverse pregnancy-related outcomes in inflammatory bowel disease. Alimentary Pharmacology & Therapeutics. 2020; 51: 320–333.
[70]
Leung YPY, Kaplan GG, Coward S, Tanyingoh D, Kaplan BJ, Johnston DW, et al. Intrapartum corticosteroid use significantly increases the risk of gestational diabetes in women with inflammatory bowel disease. Journal of Crohn’s & Colitis. 2015; 9: 223–230.
[71]
Nelson-Piercy C, MacCallum P, Mackillop L. Reducing the Risk of Venous Thromboembolism during Pregnancy and the Puerperium. 2015. Available at: https://www.rcog.org.uk/globalassets/documents/guidelines/gtg-37a.pdf (Accessed: 26 December 2022).
[72]
Nguyen GC, Bernstein CN, Bitton A, Chan AK, Griffiths AM, Leontiadis GI, et al. Consensus statements on the risk, prevention, and treatment of venous thromboembolism in inflammatory bowel disease: Canadian Association of Gastroenterology. Gastroenterology. 2014; 146: 835–848.e6.
[73]
Lewis RT, Bleier JIS. Surgical treatment of anorectal crohn disease. Clinics in Colon and Rectal Surgery. 2013; 26: 90–99.
[74]
de Zoeten EF, Pasternak BA, Mattei P, Kramer RE, Kader HA. Diagnosis and treatment of perianal Crohn disease: NASPGHAN clinical report and consensus statement. Journal of Pediatric Gastroenterology and Nutrition. 2013; 57: 401–412.
[75]
American Gastroenterological Association Clinical Practice Committee. American Gastroenterological Association medical position statement: perianal Crohn’s disease. Gastroenterology. 2003; 125: 1503–1507.
[76]
Schwartz DA, Loftus EV, Jr, Tremaine WJ, Panaccione R, Harmsen WS, Zinsmeister AR, et al. The natural history of fistulizing Crohn’s disease in Olmsted County, Minnesota. Gastroenterology. 2002; 122: 875–880.
[77]
Rankin GB, Watts HD, Melnyk CS, Kelley ML, Jr. National Cooperative Crohn’s Disease Study: extraintestinal manifestations and perianal complications. Gastroenterology. 1979; 77: 914–920.
[78]
Platell C, Mackay J, Collopy B, Fink R, Ryan P, Woods R. Anal pathology in patients with Crohn’s disease. The Australian and New Zealand Journal of Surgery. 1996; 66: 5–9.
[79]
Safar B, Sands D. Perianal Crohn’s disease. Clinics in Colon and Rectal Surgery. 2007; 20: 282–293.
[80]
Burke KE, Haviland MJ, Hacker MR, Shainker SA, Cheifetz AS. Indications for Mode of Delivery in Pregnant Women with Inflammatory Bowel Disease. Inflammatory Bowel Diseases. 2017; 23: 721–726.
[81]
Remzi FH, Gorgun E, Bast J, Schroeder T, Hammel J, Philipson E, et al. Vaginal delivery after ileal pouch-anal anastomosis: a word of caution. Diseases of the Colon and Rectum. 2005; 48: 1691–1699.
[82]
Brondfield MN, Mahadevan U. Inflammatory bowel disease in pregnancy and breastfeeding. Nature Reviews. Gastroenterology & Hepatology. 2023; 20: 504–523.
[83]
Selinger C, Carey N, Cassere S, Nelson-Piercy C, Fraser A, Hall V, et al. Standards for the provision of antenatal care for patients with inflammatory bowel disease: guidance endorsed by the British Society of Gastroenterology and the British Maternal and Fetal Medicine Society. Frontline Gastroenterology. 2020; 12: 182–187.
[84]
Cheng AG, Oxford EC, Sauk J, Nguyen DD, Yajnik V, Friedman S, et al. Impact of mode of delivery on outcomes in patients with perianal Crohn’s disease. Inflammatory Bowel Diseases. 2014; 20: 1391–1398.
[85]
Brandt LJ, Estabrook SG, Reinus JF. Results of a survey to evaluate whether vaginal delivery and episiotomy lead to perineal involvement in women with Crohn’s disease. The American Journal of Gastroenterology. 1995; 90: 1918–1922.
[86]
Lever G, Chipeta H, Glanville T, Selinger C. Perineal Outcomes After Delivery in 179 Mothers with Inflammatory Bowel Disease Compared to 31 258 Controls: A Single-Centre Cohort Study. Journal of Crohn’s & Colitis. 2022; 16: 511–514.
[87]
Ni J, Wu GD, Albenberg L, Tomov VT. Gut microbiota and IBD: causation or correlation? Nature Reviews. Gastroenterology & Hepatology. 2017; 14: 573–584.
[88]
Benchimol EI, Kaplan GG, Otley AR, Nguyen GC, Underwood FE, Guttmann A, et al. Rural and Urban Residence During Early Life is Associated with Risk of Inflammatory Bowel Disease: A Population-Based Inception and Birth Cohort Study. The American Journal of Gastroenterology. 2017; 112: 1412–1422.
[89]
Kronman MP, Zaoutis TE, Haynes K, Feng R, Coffin SE. Antibiotic exposure and IBD development among children: a population-based cohort study. Pediatrics. 2012; 130: e794–e803.
[90]
Xu L, Lochhead P, Ko Y, Claggett B, Leong RW, Ananthakrishnan AN. Systematic review with meta-analysis: breastfeeding and the risk of Crohn’s disease and ulcerative colitis. Alimentary Pharmacology & Therapeutics. 2017; 46: 780–789.
[91]
De Vroey B, Colombel JF. IBD in 2010: optimizing treatment and minimizing adverse events. Nature Reviews. Gastroenterology & Hepatology. 2011; 8: 74–76.
[92]
Lundgren SN, Madan JC, Emond JA, Morrison HG, Christensen BC, Karagas MR, et al. Maternal diet during pregnancy is related with the infant stool microbiome in a delivery mode-dependent manner. Microbiome. 2018; 6: 109.
[93]
Malmborg P, Bahmanyar S, Grahnquist L, Hildebrand H, Montgomery S. Cesarean section and the risk of pediatric Crohn’s disease. Inflammatory Bowel Diseases. 2012; 18: 703–708.
[94]
Sevelsted A, Stokholm J, Bønnelykke K, Bisgaard H. Cesarean section and chronic immune disorders. Pediatrics. 2015; 135: e92–e98.
[95]
Torres J, Hu J, Seki A, Eisele C, Nair N, Huang R, et al. Infants born to mothers with IBD present with altered gut microbiome that transfers abnormalities of the adaptive immune system to germ-free mice. Gut. 2020; 69: 42–51.
[96]
Zelinkova Z, Stokkers PC, van der Linde K, Kuipers EJ, Peppelenbosch MP, van der Woude CPJ. Maternal imprinting and female predominance in familial Crohn’s disease. Journal of Crohn’s & Colitis. 2012; 6: 771–776.
[97]
Moller FT, Andersen V, Wohlfahrt J, Jess T. Familial risk of inflammatory bowel disease: a population-based cohort study 1977-2011. The American Journal of Gastroenterology. 2015; 110: 564–571.
[98]
Bennett A, Mamunes A, Kim M, Duley C, Garrett A, Annis K, et al. The Importance of Monitoring the Postpartum Period in Moderate to Severe Crohn’s Disease. Inflammatory Bowel Diseases. 2022; 28: 409–414.
[99]
Yu A, Friedman S, Ananthakrishnan AN. Incidence and Predictors of Flares in the Postpartum Year Among Women With Inflammatory Bowel Disease. Inflammatory Bowel Diseases. 2020; 26: 1926–1932.
[100]
Goulden B, Chua N, Parker E, Giles I. A systematic review of live vaccine outcomes in infants exposed to biologic disease modifying anti-rheumatic drugs in utero. Rheumatology. 2022; 61: 3902–3906.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top