ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Safety of rheumatic disease medication use during pregnancy and lactation

Safety of rheumatic disease medication use during pregnancy and lactation
Literature review current through: Jan 2024.
This topic last updated: May 01, 2023.

INTRODUCTION — Inflammatory disorders such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), and inflammatory bowel disease (IBD) often occur in women of childbearing age; thus, the treatment of such disorders in women during pregnancy is an important aspect of their management.

When considering treatment options, a major tenet of treating women during pregnancy is to keep disease under good control while minimizing or avoiding medications that may increase maternal or fetal risk. The relative benefits and risks to the mother and fetus of using a particular medication to maintain disease control or to treat active disease during pregnancy depend upon the specific clinical context and may be influenced by gestational age and other factors. Importantly, untreated disease carries its own risks to both the mother and the developing fetus.

The risks in pregnancy and during lactation of antiinflammatory, immunosuppressive, and biologic medications used for the treatment of rheumatic diseases will be reviewed here. Additional information regarding each medication is provided in the drug information topic for each agent. Approaches to the therapy of individual conditions are presented separately within the appropriate topic reviews on the treatment of given disorders, and for some conditions, in separate topic reviews. (See "Rheumatoid arthritis and pregnancy" and "Pregnancy in women with systemic lupus erythematosus".)

Strategies for the preservation of fertility in patients undergoing cytotoxic therapy; cryopreservation of sperm, oocytes, and embryos; and the effects of antiinflammatory and immunosuppressive medications on male fertility are discussed in detail separately. (See "Fertility and reproductive hormone preservation: Overview of care prior to gonadotoxic therapy or surgery" and "Effects of antiinflammatory and immunosuppressive drugs on gonadal function and teratogenicity in men with rheumatic diseases".)

RISK CATEGORIES AND TREATMENT CONTEXT — The US Food and Drug Administration (FDA) has changed the format for labeling medications for safety of use during pregnancy and lactation. The previous letter categories inaccurately implied increased risk as one went from category A to B to C to D to X; in actuality, these categories reflected the quantity and type of data available. The system that began implementation in mid-2015 provides a discussion of the available data regarding a particular drug's safety in pregnancy and lactation, as well as information regarding the risk of untreated disease [1].

There is limited information regarding the toxicities and teratogenic potential of many antiinflammatory and immunosuppressive medications in pregnant women because there are ethical issues involved in testing such drugs in the setting of pregnancy. For some medications, the available evidence is limited to or primarily from animal research.

For practical purposes, we divide the medications used to treat rheumatic diseases into four categories of risk for use during pregnancy. We recognize that some drugs may fall into more than one category depending upon various factors. These categories are:

Minimal fetal or maternal risk

Selective use allowed during pregnancy

Moderate to high risk of fetal harm

Limited information

Information related to the toxicity and teratogenicity of rheumatic disease medications in pregnant women must be interpreted in light of the background risk of adverse pregnancy outcomes in all women. Major congenital anomalies are defined as events of medical, surgical, or cosmetic significance. It is estimated that the prevalence of major congenital anomalies is 2 to 4 percent among liveborn infants. This prevalence does not vary among ethnic groups (table 1). Evaluation of lactation risk often involves calculation of the relative infant dose (RID), which is the dose administered to the infant through breast milk in mg/kg/day divided by the maternal dose in mg/kg/day. Estimates of less than 10 percent of the maternal dose are generally considered safe.

MINIMAL FETAL OR MATERNAL RISK — The following medications are viewed as relatively safe in pregnancy:

Hydroxychloroquine (HCQ) (see 'Hydroxychloroquine' below)

Sulfasalazine (SSZ) (see 'Sulfasalazine' below)

Low-dose aspirin (see 'Aspirin (low dose)' below)

Azathioprine (AZA) and 6-mercaptopurine (6-MP) (see 'Azathioprine and 6-mercaptopurine' below)

Colchicine (see 'Colchicine' below)

Hydroxychloroquine

Pregnancy – HCQ can be continued during pregnancy. For most patients with rheumatic disease, the benefits of HCQ during pregnancy outweigh the potential risks.

Lactation – HCQ use is compatible with breastfeeding.

HCQ is an antimalarial agent that is effective in the treatment of several rheumatic diseases. Specific details regarding the use and monitoring of this medication are presented separately. (See "Antimalarial drugs in the treatment of rheumatic disease" and "Pregnancy in women with systemic lupus erythematosus" and "Rheumatoid arthritis and pregnancy".)

HCQ is stored in tissues, particularly the liver, and has a half-life of approximately eight weeks. Thus, fetal exposure cannot be avoided by discontinuing the medication at the time the pregnancy is discovered.

HCQ crosses the placenta, but most studies have not described fetal toxicity with HCQ doses used for the treatment of systemic lupus erythematosus (SLE) or other rheumatic diseases [2-4]. However, one large population-based cohort study of rheumatic disease patients used claims data and compared 2045 HCQ-exposed pregnancies and 21,679 matched pregnancies not exposed to HCQ. HCQ use during the first trimester was associated with a small increased risk of major congenital malformations (adjusted relative risk 1.26, 95% CI 1.04-1.54) [5]. The absolute risk for exposed infants was slightly higher than in unexposed infants of autoimmune rheumatic disease patients (5.48 versus 4.32 percent). However, no particular pattern of malformations was identified. Moreover, this study did not account for variables not included in the data set, such as use of alcohol, tobacco, folic acid, other drugs, and over-the-counter medications, as well as unidentified confounders [6]. In a subsequent cohort study of pregnant individuals enrolled in the MotherToBaby/Organization of Teratology Information Specialists Autoimmune Diseases in Pregnancy Study who were receiving HCQ, 279 persons exposed to HCQ were compared with 279 unexposed pregnant persons. There was no increased risk of structural congenital anomalies among infants exposed to HCQ in utero [7].

Several studies have suggested that continuation of HCQ may improve pregnancy outcome in women with SLE; these studies have not raised any safety concerns. As an example, in a retrospective series of 257 pregnancies from the Johns Hopkins Lupus Cohort, the pregnancies were divided into three groups: mothers with no HCQ exposure during pregnancy (n = 163), those in whom HCQ was used continuously (n = 56), and those in whom HCQ was stopped during the first trimester or within three months of conception (n = 38) [8]. The following observations were made:

There were no differences among the three groups in miscarriages, stillbirths, pregnancy losses, and congenital abnormalities.

SLE flares were more common, and the mean prednisone doses were higher, among the women who stopped taking HCQ during pregnancy. However, serious complications of SLE were not more common in that group.

There are inconclusive data on whether HCQ reduces the risk of preeclampsia in SLE pregnancies. One retrospective cohort study of 119 SLE pregnancies that used propensity score matching demonstrated the safety of HCQ treatment but no reduction in preeclampsia [9].

Another retrospective cohort study showed a trend toward less preeclampsia in SLE patients treated with HCQ during pregnancy [10].

The safety of HCQ in SLE pregnancy was further corroborated by two other studies [3,11]:

In a series of 36 pregnancies in women with SLE exposed to HCQ during pregnancy, the percentage of women with successful pregnancies (ie, lack of teratogenicity and good obstetric outcome) was not different from that of 53 control women [3].

During a mean of 26 months' follow-up of the children resulting from 133 HCQ-exposed pregnancies, there was no difference in the rate of growth, and there was no evidence of visual or developmental abnormalities [11].

HCQ is transferred to human breast milk: breastfed infants are exposed to approximately 2 percent of the maternal dose, which is well within levels considered to be safe [12]. The American Academy of Pediatrics (AAP) considers use of HCQ compatible with breastfeeding.

Sulfasalazine

Pregnancy – For women with active inflammatory disease who require treatment during pregnancy, SSZ is an acceptable therapy with relatively low risk. Concomitant folate supplementation is advised because this drug is a dihydropteroate synthase inhibitor that can lead to folate deficiency. SSZ does not appear to increase the risk of adverse fetal outcome if the mother is taking a multivitamin with at least 0.4 mg of folic acid.

Lactation – SSZ is transferred in low concentration into breast milk and is considered compatible with breastfeeding in healthy, full-term infants. However, women taking SSZ should avoid breastfeeding premature infants or those with hyperbilirubinemia or glucose-6-phosphate dehydrogenase (G6PD) deficiency.

SSZ possesses both antiinflammatory properties mediated by its 5-aminosalicylic acid moiety and antibacterial characteristics associated with its sulfapyridine moiety. Specific details regarding the use and monitoring of this medication are presented separately. (See "Sulfasalazine and 5-aminosalicylates in the treatment of inflammatory bowel disease" and "Sulfasalazine: Pharmacology, administration, and adverse effects in the treatment of rheumatoid arthritis".)

SSZ has been used since its development in the 1940s as an agent for the treatment of inflammatory bowel disease (IBD) and rheumatoid arthritis (RA). Most of the information regarding the use of this medication during pregnancy is gleaned from its effects on patients with IBD.

SSZ and its metabolite sulfapyridine cross the placenta [13]. However, neither SSZ nor sulfapyridine causes significant displacement of bilirubin from albumin [14]. Thus, SSZ use by pregnant women does not appear to increase the risk of kernicterus.

Larger case series of patients have shown no increased incidence of adverse pregnancy outcomes in pregnancies in which the mother is taking SSZ [15,16]. One study reported 247 IBD patients who had a total of 240 births [15]. There was no increased risk of fetal anomalies in offspring of mothers treated with SSZ alone or with the combination of glucocorticoids and SSZ. In addition, none of the 107 mothers who received SSZ during pregnancy had babies with jaundice.

The amount of SSZ found in the breast milk and in the serum of nursing babies is low [17,18]. Sulfapyridine concentrations in the serum of nursing babies is 40 percent that of the level of the metabolite in mothers. Diarrhea in a nursing child whose mother was taking SSZ has been reported [19]; thus, in infants having diarrhea, consider discontinuation of SSZ in the lactating mother.

Aspirin (low dose)

Pregnancy – Salicylate use in low doses (eg, aspirin, 81 to 160 mg a day) is considered compatible with pregnancy.

Lactation – Salicylates in low doses (eg, aspirin, 81 mg daily as an antiplatelet agent) are compatible with nursing [20,21].

High-dose aspirin should generally be avoided during pregnancy and lactation, as discussed elsewhere. (See 'Aspirin (moderate to high dose)' below.)

Aspirin has been used as an analgesic and antiinflammatory agent in the management of rheumatic disease. Low-dose aspirin (81 mg a day) is used as part of the management of antiphospholipid syndrome and for prevention of preeclampsia as well. Aspirin, an acetylated salicylate (acetylsalicylic acid), is classified among the nonsteroidal antiinflammatory drugs (NSAIDs). The safety of moderate- to high-dose aspirin is described together with the non-salicylate NSAIDs. (See 'NSAIDs' below.)

Data regarding the safety of aspirin during pregnancy have limitations, as the outcomes associated with different dose ranges are often not presented.

In a 2002 meta-analysis of 22 studies of aspirin exposure during pregnancy, the overall risk of congenital anomalies was not statistically significantly elevated (odds ratio [OR] 1.33, 95% CI 0.94-1.89), suggesting that aspirin did not expose an increased teratogenic risk. This same meta-analysis did report a significantly increased risk of the rare finding of gastroschisis (OR 2.37, 95% CI 1.44-3.88) [22]. However, information on aspirin dose, trimester of exposure, and duration of therapy were not available.

An older observational study of Australian women attending an antenatal clinic reported that 6.6 percent of women were taking salicylates (dose not reported) [23]. Among the women who were constant takers, defined as daily use (dose unknown), there was a higher incidence of anemia and postpartum hemorrhage, prolonged gestation, and perinatal mortality. However, salicylate dose, indications for aspirin use, and confounders were not discussed.

In another study of 50,282 pregnancies, 9736 had intermediate exposure to salicylates, and 5128 had heavy exposure during the first four months of gestation (defined as >8 days during a month period, dose not specified) [24]. No increase in congenital anomalies were reported.

Low-dose aspirin has been used to prevent preeclampsia, providing further evidence of its relative safety. Individual patient data from over 32,000 women and their babies showed that low-dose aspirin had no significant effect on the risk of bleeding events in either the women or their babies [25]. Low doses of aspirin have also been used during pregnancy in selected patients with antiphospholipid syndrome; it has also been used in patients undergoing in vitro fertilization/intracytoplasmic sperm injection [26]. (See "Antiphospholipid syndrome: Obstetric implications and management in pregnancy" and "Preeclampsia: Prevention".)

Azathioprine and 6-mercaptopurine

Pregnancy – When immunosuppression is necessary (eg, in renal transplant recipients, recently active lupus, or IBD), AZA or 6-MP can be used during pregnancy. While these medications do not appear to increase the risk of teratogenicity, higher rates of other pregnancy complications (low birth weight, prematurity, and jaundice) have been reported. AZA, the medication more commonly used in rheumatologic disorders, may be safer than 6-MP, and there is a general consensus that AZA may be safer than many other immunosuppressive agents during pregnancy [27].

Lactation – AZA is considered compatible with breastfeeding. Available evidence suggests that excretion of AZA in breast milk is very low in most women [28,29].

AZA is a purine metabolism antagonist and is metabolized to 6-MP in vivo. Thus, these two medications should theoretically have the same effects during pregnancy. Specific details regarding the use and monitoring of these medications are presented separately. (See "Pharmacology and side effects of azathioprine when used in rheumatic diseases" and "Overview of azathioprine and mercaptopurine use in inflammatory bowel disease".)

Radioactive labeling studies in humans have shown that 64 to 93 percent of AZA administered to mothers appears in fetal blood as inactive metabolites [30]. This is because the placenta metabolizes AZA to thiouric acid, which is inactive, rather than 6-MP [30]. After birth, AZA is metabolized to 6-MP.

Data on the use of AZA in pregnant women come primarily from studies of patients who have undergone organ transplantation and patients who have Crohn disease [27,31-33].

Among transplant recipients, one report evaluated 146 renal allograft recipients who received AZA and prednisone (90 percent), AZA alone (2 percent), or glucocorticoids alone (8 percent) [34]. Low birth weights, prematurity, jaundice, respiratory distress syndrome, and aspiration all occurred significantly more frequently among the offspring of mothers in the two AZA groups. In addition, there were isolated cases of immunoglobulin deficiency, chromosomal abnormalities, and malformations, but the frequency of these events was not higher than that expected in a healthy obstetrical population [27].

In Crohn disease, a nationwide cohort study in Denmark included 900 children born to women with that disease between 1996 and 2004 [31]. Twenty mothers in that study were exposed to AZA or 6-MP during pregnancy. The following findings were observed:

Women exposed to AZA or 6-MP alone or in combination with other Crohn disease medications had higher risks of preterm births (25 percent) compared with patients who received the combination of glucocorticoids and SSZ (13 percent), SSZ alone (4 percent), or no medications for Crohn disease during pregnancy (7 percent). Compared with the reference group of patients who received no medications, the relative risk (RR) of preterm birth was 4.2 in the AZA or 6-MP group (95% CI 1.4-12.5). It is possible that patients using these medications were sicker, which may have also affected the risk of preterm birth.

Congenital abnormalities occurred in 15 percent of the AZA or 6-MP group, compared with 4, 6, and 6 percent in women treated with glucocorticoid and SSZ, with SSZ alone, and with no medications, respectively. Compared with the reference group of patients who received no medications, the AZA or 6-MP group had nearly three times the risk of congenital abnormalities (RR 2.9, 95% CI 0.9-8.9).

This study was limited by the small number of patients treated with AZA or 6-MP and by the fact that the investigators were not able to adjust their analysis for disease severity. In addition, results were not separately analyzed for each individual drug but were combined.

Other data on the use of AZA or 6-MP by women who are pregnant are more encouraging with regard to the risk of adverse birth outcomes.

A retrospective cohort study of 155 IBD patients reported no statistical difference in fertility, congenital anomalies, infections, and maternal malignancies among the group treated with 6-MP [32].

A retrospective study of 101 pregnancies in women with IBD on AZA also revealed no association with poor pregnancy outcomes [33].

Breastfeeding during AZA treatment of the mother has been discouraged in the past, including in a recommendation of the World Health Organization (WHO) that was formulated in 1988 [35,36]. While the manufacturers' inserts for AZA and 6-MP say that they are excreted in breast milk and should not be used in women who are breastfeeding, data suggest that excretion in breast milk is very low in most women [28,29]. One study of four women being treated with AZA found no detectable 6-MP in their breast milk [29]. The theoretical absolute infant dose was less than 0.09 percent of the mother's weight-adjusted dose, well within safety limits. Another study, involving 10 nursing women who provided a total of 31 breast milk samples, reported the following [28]:

Twenty-nine of the 31 samples contained no detectable 6-MP.

Two samples from one woman contained low concentrations of 6-MP (1.2 and 7.6 ng/mL, compared with a plasma concentration of 50 ng/mL that is normally considered therapeutic).

The AZA metabolites 6-MP and 6-thioguanine were not detectable in neonatal blood, and there were no clinical or hematologic signs of immunosuppression in any of the 10 neonates. Two smaller studies of women following renal transplantation reported similar findings [37,38]. Recommendations from professional organizations such as the British Society of Rheumatologists and the European Alliance of Associations for Rheumatology (EULAR; formerly known as European League Against Rheumatism) consider AZA compatible with nursing [39,40].

Colchicine

PregnancyColchicine is compatible with pregnancy [41].

LactationColchicine can be continued in nursing mothers [42-44].

Colchicine is used to treat familial Mediterranean fever (FMF), autoinflammatory conditions, and crystal-induced arthritis. While colchicine blocks mitotic cells in metaphase, it does not appear to cause human fetal anomalies [45]. A meta-analysis that included 554 pregnancies in women with FMF who continued colchicine during pregnancy found no congenital anomalies [41].

The use and side effects of colchicine are discussed in the drug information topic on this agent and in the treatment sections for the various disorders in which it is employed. Colchicine has drug interactions and altered metabolism in certain patient populations, which are also discussed separately.

While colchicine is transmitted into breast milk, the levels of drug found in the breast milk of women who are lactating and taking this medication are considered safe [42]. Case reports of breastfed infants whose mothers are taking this medication have not reported any safety concerns [43], and the AAP considers colchicine to be compatible with breastfeeding [44].

SELECTIVE USE ALLOWED DURING PREGNANCY

NSAIDs — The relative safety and the nature of the risks during pregnancy and lactation of nonsteroidal antiinflammatory drugs (NSAIDs) are generally dependent upon the timing of use during pregnancy, the dose, and the specific class of medication. These drugs are widely used for their analgesic, antiinflammatory, and antiplatelet properties. The use of nonselective NSAIDs (eg, ibuprofen) during pregnancy and lactation are discussed in the sections that follow. The use of high-dose aspirin or cyclooxygenase 2 (COX-2) inhibitors is generally avoided. (See 'Aspirin (moderate to high dose)' below and 'COX-2 inhibitors' below.)

Specific details regarding the use and monitoring of these medications are presented separately. (See "NSAIDs: Therapeutic use and variability of response in adults" and "Nonselective NSAIDs: Overview of adverse effects" and "Overview of COX-2 selective NSAIDs".)

Fertility — Women who are having difficulty with conceiving may wish to avoid use of NSAIDs if disease control would not be compromised. Case reports and some small series have shown transient infertility in some women treated with NSAIDs, which appears to be mediated by inhibition of the rupture of the luteinized follicle [46-48].

Pregnancy up to 20 weeks — NSAID use during the first 20 weeks of pregnancy is appropriate when needed. Risk from NSAID use during the first trimester appears low overall. There does not appear to be an increased risk of spontaneous abortion.

Major risks of NSAID use during early pregnancy have not been clearly identified. Among three studies involving greater than 200,000 pregnancies overall and over 11,000 NSAID-exposed pregnancies (including a 2002 meta-analysis and two subsequent population-based studies), there was no significant increase compared with unexposed infants in the frequency of musculoskeletal or cardiac malformations, nor was there an effect upon infant survival [22,49,50].

However, some studies have suggested small risks of certain adverse birth outcomes. In a nationwide, population-based cohort study representing 1.8 million pregnancies in South Korea, exposure to NSAIDs during the first trimester was associated with a slight increased risk for neonatal outcomes of major congenital malformations (propensity score adjusted relative risk [RR] 1.14, 95% CI 1.10-11.18), low birth weight (adjusted RR 1.29, 95% CI 1.25-1.33), and oligohydramnios (adjusted RR 1.09, 95% CI 1.01-1.19) [51]. However, no particular pattern of congenital anomaly was identified, nor could the authors rule out confounding by indication. In another population-based study, there was a borderline association of ibuprofen use during the first trimester with structural heart defects detected during the first 18 months of life (odds ratio [OR] 1.2, 95% CI 1.0-1.6) [50].

NSAID use is not clearly associated with an increased rate of spontaneous abortion [52,53]. Several observation studies have suggested that NSAID use may be associated with an increased risk of spontaneous abortion [54-57]; however, these studies were limited by a lack of adjustment for the disorder for which NSAIDs were taken, and the effects of other comorbidities and medications, as well as dosing regimens, were not fully analyzed. As examples:

A study of 1097 women showed that the risk of spontaneous abortion was increased in those women who used NSAIDs during early pregnancy compared with unexposed controls and with women who used acetaminophen [57]. After adjusting for multiple confounders, including maternal age, previous spontaneous abortion, and smoking, the effect remained, but only for women who used NSAIDs around conception (adjusted hazard ratio [HR] 1.9, 95% CI 1.3-2.7). Results for unexposed controls were similar to those for acetaminophen users. NSAID use for more than 14 days was associated with a higher risk of miscarriage compared with use for ≤14 days, and a lower body mass index (BMI <25) was associated with elevated risk, while a higher BMI was not. These findings are consistent with the hypothesis that impaired embryo implantation due to inhibition of prostaglandin production may increase risk of miscarriage.

A large, nested, case-control study using data from a Canadian registry suggested that exposure to (non-salicylate) nonselective and COX-2 selective NSAIDs in the first 20 weeks of pregnancy may be associated with an increased risk of miscarriage (OR 2.4, 95% CI 2.1-2.8) [54]. These findings should be interpreted with great caution. The control group was not matched for maternal age, which is an important risk factor for spontaneous abortion, and smoking status and use of over-the-counter NSAIDs were not known.

A large cohort study that linked data from medication dispensing records to information on obstetric outcomes for 65,457 pregnancies, including 6508 pregnant women with spontaneous abortions and 4495 pregnant women exposed to NSAIDs, found no significant increase in the risk of spontaneous abortion following NSAID exposure after controlling for maternal age, diabetes, thyroid disease, hypercoagulation disorders, smoking status, inflammatory conditions, history of recurrent miscarriage, in vitro fertilization, and other factors [53]. This was true both for nonselective NSAIDs (HR 1.10, 95% CI 0.99-1.22) and COX-2 selective NSAIDs (HR 1.43, 95% CI 0.79-2.59). The results for the subset of patients exposed to COX-2 selective NSAIDs were imprecise, given the low number of exposed patients (71) and events (11), and further study of the safety of these medications is needed. An increased risk observed specifically with indomethacin was likely to be related to its therapeutic use for preterm labor.

Pregnancy after 20 weeks — We generally avoid NSAIDs after 20 weeks gestation because of a small increased risk of oligohydramnios. In selected patients who have no alternative therapy, these medications may be used from 20 to 30 weeks with the caveat that amniotic fluid levels should be monitored by ultrasonography. From 30 weeks onward, we suggest avoiding the use of NSAIDs altogether because of the risk of premature closure of the ductus arteriosus. (See 'Third-trimester use of NSAIDs' below.)

In 2020, the US Food and Drug Administration (FDA) issued a warning that use of NSAIDs around 20 weeks gestation or later in pregnancy may rarely cause fetal kidney dysfunction leading to oligohydramnios and, in some cases, neonatal kidney impairment [58]. These outcomes could be seen within 48 hours of NSAID use but typically occurred after days to weeks of treatment. Oligohydramnios was often, but not always, reversible with discontinuation of the NSAIDs, but persistent oligohydramnios could potentially cause further complications. The FDA suggested that, if NSAID treatment was necessary between 20 to 30 weeks of pregnancy, it be at the lowest effective dose and shortest duration possible, monitoring by ultrasound be considered for those treated for more than 48 hours, and the drug be discontinued if oligohydramnios occurs. (See "Oligohydramnios: Etiology, diagnosis, and management in singleton gestations".)

Lactation — There is limited information on the safety of most NSAIDs in breastfeeding women. Ibuprofen is the preferred NSAID in breastfeeding individuals but only because it is secreted into breast milk in very small amounts, and there is the most information on its safety.

Ibuprofen levels were undetectable in the breast milk of individuals taking 1600 mg of ibuprofen a day [59].

Glucocorticoids

Pregnancy – We use the lowest effective glucocorticoid dose during pregnancy. Whether stress-dose glucocorticoids are indicated in women who have been on glucocorticoids during pregnancy and undergo cesarean delivery is controversial. However, a 2016 review suggests that the recommendation for stress-dose glucocorticoids in this setting is based upon limited data [60]. Moreover, the 2017 American College of Rheumatology guideline for the perioperative management of antirheumatic medication for patients undergoing joint replacement does not recommend stress-dose glucocorticoids during joint replacement [61].

Lactation – Glucocorticoids are considered compatible with breastfeeding. They are excreted in breastmilk in low concentration. We advise discarding breast milk for the first four hours following ingestion of a dose of prednisone ≥20 mg, as the peak concentration in breast milk is achieved two hours after maternal ingestion.

Glucocorticoids have a variety of side effects when given systemically or by inhalation. Specific details regarding the use and monitoring of these drugs, including the use of inhaled agents during pregnancy and prenatal use to promote fetal lung maturity in women at risk of premature birth, are presented separately. (See "Pharmacologic use of glucocorticoids" and "Major adverse effects of systemic glucocorticoids" and "Major side effects of inhaled glucocorticoids" and "Management of asthma during pregnancy", section on 'Inhaled glucocorticoids' and "Antenatal corticosteroid therapy for reduction of neonatal respiratory morbidity and mortality from preterm delivery".)

The most commonly used short-acting glucocorticoids are prednisone, prednisolone, and methylprednisolone. The most common long-acting agents are dexamethasone and betamethasone. Prednisone and prednisolone cross the placenta but appear in only small amounts in cord blood [62]. By contrast, dexamethasone and betamethasone reach higher concentrations in the fetus because they are metabolized less efficiently by the placenta.

Early evidence suggested that glucocorticoid exposure increases the risk of cleft palate formation in fetuses exposed in the first trimester. As an example, a cohort study and meta-analysis from year 2000 of epidemiologic studies of women who used glucocorticoids during pregnancy did report an increased risk of bearing a child with cleft palate (OR 3.4, 95% CI 1.97-5.69) [63]. Additionally, the administration of glucocorticoids to pregnant rodents can result in cleft palates in the offspring [64].

However, in contrast to these observations, no increase in orofacial clefts was identified among 51,973 infants exposed to glucocorticoids during the first trimester of pregnancy compared with unexposed infants in a 2011 nationwide cohort study of patients in Denmark involving 832,636 live births, among whom a total of 1232 isolated orofacial clefts were identified [65]. Orofacial clefts (cleft lip and/or palate) occur in approximately 1.7 in 1000 live births in the general population [66].

Glucocorticoid therapy during pregnancy may increase the risk of premature rupture of the membranes (PROM) and intrauterine growth restriction [67,68] and, in the mother, may increase the risk of pregnancy-induced hypertension, gestational diabetes, osteoporosis, and infection [27]. To avoid these risks, we recommend using the lowest dose of glucocorticoid possible during pregnancy to control disease activity. Use of high-dose glucocorticoids should be limited to women who have organ-threatening disease when the benefits are likely to outweigh the risks. A detailed discussion with the pregnant woman regarding the potential risks and benefits of high-dose glucocorticoids during pregnancy should take place.

The approach to addressing the potential need for stress dose glucocorticoids during labor and delivery is similar to that for patients undergoing surgery, which is discussed in detail separately (see "The management of the surgical patient taking glucocorticoids"). Significant suppression of the hypothalamic-pituitary adrenal axis in the newborn is infrequent and typically transient [69].

Glucocorticoids are excreted into the breast milk [70], but their use during lactation is deemed compatible by the American Academy of Pediatrics (AAP) and the British Society of Rheumatologists. We advise discarding breast milk for the first four hours following ingestion of a dose of prednisone ≥20 mg, as the peak concentration in breast milk is achieved two hours after ingestion of this medication by the lactating mother.

Tumor necrosis factor inhibitors

Pregnancy – Tumor necrosis factor (TNF)-alpha blockers may be used in women who require these medications for the maintenance or establishment of control of active inflammatory disease during pregnancy. Some expert guidance suggests these medications should be discontinued in the late second or early third trimester [71]. However, use of these drugs can be extended, if necessary, to a later gestational age if benefits outweigh potential risks for an individual patient. Certolizumab is compatible for use throughout pregnancy. Only very limited information is available regarding golimumab.

Infants exposed to TNF-alpha blockade in utero should avoid live vaccines (table 2) during the first six months of life but otherwise can follow a standard vaccination schedule for inactive vaccines. Among the live vaccines that should be avoided during this period are rotavirus and Bacillus Calmette-Guérin (BCG). Rotavirus vaccine is the live vaccine generally recommended for administration before six months of age in the United States, and BCG is administered prior to six months in some other countries, although not the United States [39,40,72].

Lactation – TNF blockers are considered compatible with breastfeeding. The large size of these molecules results in little of the drug being transported during lactation.

Observational studies and case reports of women exposed to TNF inhibitors during pregnancy suggest that their pregnancy outcomes, including rates of preterm birth, spontaneous miscarriage, and congenital anomalies, are similar to those in women with rheumatoid arthritis (RA) or inflammatory bowel disease (IBD) who have not received these medications [27,73-83]. These data are limited by the small number of studies exploring this issue but are supported by animal studies that have not raised any significant concerns [84,85]. Specific details regarding the use and monitoring of these medications are presented separately. (See "Overview of biologic agents in the rheumatic diseases", section on 'TNF inhibition' and "Tumor necrosis factor-alpha inhibitors: An overview of adverse effects".)

More data are required to better assess the safety of these medications in pregnancy. Observations regarding safety during pregnancy have been made about the following TNF inhibitors to varying degrees:

Etanercept (a soluble fusion protein)

Infliximab (a chimeric monoclonal antibody)

Adalimumab (a humanized monoclonal antibody)

Certolizumab pegol (CZP; a pegylated human monoclonal antibody Fab' fragment)

Golimumab (a human monoclonal antibody)

A prospective multicenter cohort study that included 495 TNF-alpha inhibitor-exposed infants suggested that the risk of major congenital anomalies was increased in exposed (5 percent) versus non-exposed infants (1.5 percent; OR 2.2, 95% CI 1.0-4.8) [86]. However, this study was limited by the lack of a non-exposed population with rheumatologic disease or IBD, and no particular pattern of malformations were identified, making it difficult to attribute causality to the TNF inhibitors.

CZP lacks the antibody Fc- region that is responsible for placental transfer of immunoglobulins. In humans, only minimal, if any, placental transfer occurs [87]. Updated results from the manufacturer's pharmacovigilance database, including 1137 prospectively documented maternal CZP exposures, identified 538 known outcomes (of 301 women with rheumatic diseases, 195 with Crohn disease, and 43 others) through March 6, 2017 [83]. Eighty-one percent of pregnancies had exposures in the first trimester, the major period for organogenesis. There was no increase in teratogenicity or fetal death in CZP-exposed pregnancies compared with the general population. Among the outcomes were 459 live births (85 percent), 47 miscarriages (9 percent), 27 elective abortions (5 percent), and 5 stillbirths (1 percent). There were 8 major congenital malformations (1.7 percent) among the 459 live births; this level is comparable to the background rate of congenital anomalies.

There are only very limited data available regarding the safety of golimumab during pregnancy. In one study of TNF inhibitors that included three pregnancies with exposure to golimumab, none of the congenital anomalies occurred in the golimumab group [86]. Animal studies also suggest that risk of golimumab during pregnancy is low [88].

Serious infections do not appear to be increased in offspring exposed to TNF inhibitors during pregnancy [89,90]. In one study of 380 children of RA mothers who were exposed to TNF inhibitors during pregnancy, the frequency of serious infections in the year following birth was similar to that in offspring of RA patients not exposed to TNF inhibitors and to non-RA offspring [89].

TNF inhibitors are large molecules and, as such, little of these drugs are transferred to breast milk. The concentration of etanercept in breast milk has been measured in a single patient who was lactating (but not nursing her newborn) and was approximately 4 percent of the maternal serum concentration, well below medication levels that are considered incompatible with nursing [91]. Similarly, in a patient who was receiving infliximab for the treatment of Crohn disease, levels of the medication were undetectable in the patient's breast milk [92]. Given that little drug is transferred into breast milk and the drug is likely digested in the infant's gut and therefore not absorbed by the infant, current guidelines consider these medications compatible with lactation [39,40].

Intravenous immune globulin

Pregnancy – Intravenous immune globulin (IVIG; sometimes abbreviated by industry and regulatory agencies as "IGIV") is considered compatible with pregnancy [93]. It is used for a range of indications, including in primary or secondary antibody immunodeficiencies, dermatomyositis, and antiphospholipid syndrome. (See "Overview of intravenous immune globulin (IVIG) therapy" and "Causes of cholestasis in neonates and young infants", section on 'Gestational alloimmune liver disease (neonatal hemochromatosis)'.)

LactationIVIG is compatible with breastfeeding.

IVIG is a modulator of inflammatory processes hypothesized to operate through anti-idiotype mechanisms. Specific details regarding the use and monitoring of this medication are presented separately. (See "Overview of intravenous immune globulin (IVIG) therapy".)

Immune globulin given by the subcutaneous route has been used safely in pregnancy. (See "Subcutaneous and intramuscular immune globulin therapy".)

There is little information regarding the teratogenicity of IVIG in animals. One report showed that IVIG was tolerated well in a mouse model of antiphospholipid syndrome in the setting of pregnancy [94].

In humans, IVIG appears to cross the placenta in significant amounts after 30 to 32 weeks of gestation, even after modification that alters the Fc binding sites [95]. There have been no reports of fetal malformations in humans. However, hemolytic disease of the newborn and (prior to 1994) transmission of hepatitis C have been reported in selected cases [96,97]. IVIG crosses the placenta beginning in the second trimester, but the highest transport is in the third trimester.

Available data suggest that IVIG is compatible with nursing, although there has been one case report of a transient rash in a breastfeeding infant of a mother receiving IVIG [98].

Cyclosporine

Pregnancy – When the administration of cyclosporine A (CSA) is necessary during pregnancy, the minimum dose should be used, and the maternal blood pressure and renal function should be monitored closely.

Lactation – Most evidence suggests that CSA can be taken by lactating women.

CSA is a calcineurin inhibitor. Specific details regarding the use and monitoring of this medication are presented separately. (See "Pharmacology of cyclosporine and tacrolimus" and "Cyclosporine and tacrolimus nephrotoxicity".)

There is little or no transplacental passage of CSA in rodents [99]. Studies in pregnant rats have generally shown no effect of CSA on organogenesis, although some renal proximal tubular cell damage can occur [100,101]. There are conflicting reports on the transfer of CSA across the human placenta; some reports have found little or no transfer, while others have found CSA levels in the placenta that were equivalent to those in maternal blood [102-104].

Other studies have reported the passage of cyclosporine to the fetus late in the third trimester, at birth, and with breastfeeding [105-114]. The cord blood to maternal plasma ratios ranged from 0.35 to 0.63.

Teratogenicity data in humans are derived primarily from organ transplant recipients. The risk of teratogenicity among the offspring of women treated with CSA appears to be low, but premature labor and infants who are small for gestational age have been reported [27,115]. Such findings may be related to the underlying medical condition rather than the drug and could also be related to both the medical condition and the drug. The following studies indicate the range of findings:

A 2001 meta-analysis of 15 studies of women who received CSA during pregnancy reported major malformations in 4.1 percent of offspring, a rate similar to that of the general population [116].

A second study compared the pregnancy outcomes of 115 renal transplant recipients (154 total pregnancies) treated with CSA with those of patients whose transplant immunosuppression regimen consisted only of azathioprine (AZA) and prednisone [34]. The overall complication rate among newborns in the CSA group was slightly lower, and no congenital malformations were seen. However, CSA was associated with lower birth weights and a higher incidence of maternal diabetes, hypertension, and renal allograft rejection. These may be related to the patients' medical conditions rather than to CSA itself.

CSA is excreted in breast milk, and therapeutic levels have been reported in some breastfed infants [84,114,117-119]. While the AAP has expressed concern regarding the potential toxicity of this agent in nursing infants [44], other professional organizations have suggested that CSA is compatible with nursing. Taken together, the evidence suggests that CSA can be taken by lactating women [39,40,120].

Tacrolimus

PregnancyTacrolimus use is compatible with pregnancy.

LactationTacrolimus is compatible with breastfeeding.

Tacrolimus is a calcineurin inhibitor. Specific details regarding the use and monitoring of this medication are presented separately. (See "Pharmacology of cyclosporine and tacrolimus" and "Cyclosporine and tacrolimus nephrotoxicity".)

Tacrolimus has been used as an alternative to more aggressive immunosuppressive agents for patients requiring therapy during pregnancy, although there are relatively few data concerning the effect of tacrolimus on pregnancy, and the long-term immunomodulatory effects of tacrolimus on the offspring of women who receive this medication are not known [121]. Among 100 pregnancies in 84 women treated with tacrolimus, 68 pregnancies progressed to live births. Sixty percent of these live births were premature [122]. There were four babies with malformations, but there was no consistent pattern of anatomic abnormality. Three other studies in which infants were exposed to tacrolimus in utero, representing a total of 66 pregnancies, showed no increased risk of fetal anomalies [123-125].

Two case reports found that the maximal dose infants received from nursing mothers was 0.02 to 0.5 percent of the mother's weight-adjusted dose, supporting this medication's use in breastfeeding women [126,127].

MODERATE TO HIGH RISK OF FETAL HARM — Some medications that are used to treat rheumatic diseases and have known potential for causing teratogenic effects may be used in pregnancy if the mother's health is directly threatened by an inflammatory illness. Such medications can be used with apparent safety but are always employed with reluctance because of their possible adverse impact on the pregnancy.

Cyclophosphamide

Pregnancy – We do not use cyclophosphamide (CYC) during pregnancy, except in life-threatening medical conditions in which no alternative therapy is available. The risk of teratogenicity from CYC is highest when the medication is used during the first trimester.

Lactation – CYC is excreted in breast milk and is contraindicated during breastfeeding.

CYC is an alkylating agent. Specific details regarding the use and monitoring of this medication are presented separately. (See "General toxicity of cyclophosphamide in rheumatic diseases".)

The administration of CYC to pregnant animals produces congenital anomalies such as exophthalmos, cleft palate, skeletal abnormalities, fetal resorption, and growth retardation. The risk of teratogenicity with CYC exposure in humans is high, and the medication should be avoided in all circumstances except the development of disease complications that pose grave health threats to the mother.

Most of the data in humans related to CYC exposure during pregnancy come from isolated case reports. Normal offspring have been born to mothers who received CYC during pregnancy [128]. The syndrome of "cyclophosphamide embryopathy" is discussed in more detail separately. (See "General toxicity of cyclophosphamide in rheumatic diseases", section on 'Teratogenicity'.)

The likelihood of embryotoxicity resulting from CYC administration during pregnancy varies according to the stage of pregnancy at which exposure to the drug occurs:

The risk of teratogenicity from CYC is highest when the medication is used during the first trimester [27].

Better outcomes are possible if exposure occurs during the second or third trimester. At least 36 women have been treated with CYC as part of chemotherapeutic regimens for breast cancer during the late second or third trimester without adverse pregnancy or neonatal events being described [129,130].

Limited data suggest that the pregnancies of patients with systemic lupus erythematosus (SLE) fare poorly when severe disease flares occur during pregnancy, regardless of when CYC is used [131].

In organ- or life-threatening disease, administration of CYC to pregnant women is an option after a detailed discussion of the potential risks and benefits of such treatment.

CYC is excreted in breast milk; it should be avoided during lactation [132].

Methotrexate

Pregnancy – We strongly advise against using methotrexate (MTX) during pregnancy. We also strongly advise contraceptive use in patients taking MTX. Women on MTX should discontinue this medication and should allow one to three full menstrual cycles to pass before attempting to conceive.

Lactation – While MTX is excreted into breast milk in low concentrations, it is considered contraindicated during breastfeeding [133].

MTX is a folate antagonist. Specific details regarding the use and monitoring of this medication are presented separately. (See "Major side effects of low-dose methotrexate" and "Hepatotoxicity associated with chronic low-dose methotrexate for nonmalignant disease" and "Use of methotrexate in the treatment of rheumatoid arthritis".)

Animal studies have shown that MTX is embryotoxic in early pregnancy and causes skeletal abnormalities and cleft palate later in pregnancy [134].

In humans, MTX is teratogenic and an abortifacient. MTX exposure during pregnancy can induce multiple congenital deformities such as cleft palate, hydrocephalus, anencephaly, meningoencephalocele, congenital stenosis of tubular long bones, abnormal facial features (low-set ears, micrognathia), and delayed ossification [135,136].

Most of the data on adverse events related to MTX use in pregnancy are from patients undergoing cancer therapy [137-140]. The rate of congenital malformations has been estimated to be 9 to 17 percent [27,139]. In a 2006 review of the published literature, among 63 pregnancies in which the fetus was exposed to MTX during the first trimester, 19 were terminated electively [27]. Among the remaining 44 pregnancies, the following outcomes were observed:

Healthy infants – 29 (66 percent)

Miscarriages – 11 (25 percent)

Congenital anomalies – 4 (9 percent)

In women with exposure very early during pregnancy, the risk of congenital anomalies may be lower than 9 percent.

MTX is widely distributed in maternal tissues and can persist in the liver for up to four months after exposure. However, in one study, 136 women exposed to MTX preconception (within three months) did not have a higher risk of congenital anomalies than expected compared with the general population, while the 188 women exposed postconception did have an increased rate, as expected [141]. Nevertheless, while somewhat reassuring for certain unplanned pregnancies, we continue to recommend that MTX be discontinued one to three months prior to conception. Folic acid supplementation should be continued during this time and throughout the pregnancy. (See "Use of methotrexate in the treatment of rheumatoid arthritis".)

MTX is excreted in low concentrations in breast milk; data suggest that a breastfeeding infant would receive less than 1 percent of the mother's dose [142]. Despite this, expert guidelines suggest avoiding MTX in breastfeeding mothers [39,40,72].

Mycophenolate mofetil

Pregnancy – We avoid the use of this medication during pregnancy and use mycophenolate mofetil (MMF) only in patients who employ reliable means of contraception. MMF use can decrease the effectiveness of oral contraceptives. This medication should be discontinued six weeks prior to conception.

Lactation – There are no data on the transmission of MMF into breast milk; therefore, this medication should be avoided in lactating women.

MMF is an inhibitor of purine biosynthesis. Specific details regarding the use and monitoring of this medication are presented separately. (See "Mycophenolate: Overview of use and adverse effects in the treatment of rheumatic diseases".)

A 2006 review of data from the manufacturer identified 119 human pregnancies with maternal or fetal exposure to MMF [27]. The outcomes, known for only 76 of the pregnancies, included the following:

Healthy live births – 22 (29 percent)

Pregnancy terminations – 13 (17 percent)

Miscarriages – 20 (26 percent)

Congenital anomalies – 10 (13 percent)

Similar findings have been noted in kidney transplant recipients [143]. Problems reported with MMF include increases both in first-trimester pregnancy loss and in congenital malformations, such as cleft lip and palate, anomalies of the distal limbs, heart, esophagus, and kidneys, and others [144-147]. A possibly characteristic phenotype including cleft lip and palate, microtia, and atresia of external auditory canals has been reported in six cases [148]. This medication should be discontinued six weeks prior to pregnancy. (See "Sexual and reproductive health after kidney transplantation", section on 'Management of immunosuppression'.)

Leflunomide

Pregnancy – We strongly advise against using leflunomide (LEF) during pregnancy. We also strongly advise contraceptive use in patients taking LEF. In women treated with LEF within 24 months of planning pregnancy, demonstration of undetectable blood levels or using a cholestyramine wash-out with subsequent demonstration of undetectable levels is needed. (See "Pharmacology, dosing, and adverse effects of leflunomide in the treatment of rheumatoid arthritis", section on 'Treatment to accelerate drug elimination'.)

Lactation – LEF is contraindicated during lactation.

Limited data are available on the outcomes of pregnancies occurring in women taking LEF, an antimetabolite that inhibits dihydroorotate dehydrogenase, the enzyme that catalyzes the rate-limiting step in pyrimidine biosynthesis [139,149-154]. Although the half-life of LEF is only approximately 15 days, its major metabolite (teriflunomide) undergoes extensive enterohepatic circulation and remains detectable in serum for up to two years. Thus, discontinuation of LEF immediately before conception is insufficient for drug elimination. However, elimination of the drug and its metabolites can be accelerated through the administration of cholestyramine (8 g orally three times daily for 11 days) and can be confirmed by measurement of drug levels less than 0.02 mg/L on two tests performed two weeks apart. Additional cholestyramine treatment is indicated if plasma levels remain greater than 0.02 mg/L [27].

Specific details regarding the use and monitoring of this medication are presented separately. (See "Pharmacology, dosing, and adverse effects of leflunomide in the treatment of rheumatoid arthritis".)

Concerns regarding its safety during pregnancy derive both from animal studies, which have demonstrated embryotoxicity and marked teratogenicity [149,155], and from the persistence of drug metabolites for many months following discontinuation of therapy. Several case reports have also raised concerns, although good pregnancy outcomes have been observed as well [151-153].

The largest reported series of women exposed to LEF during conception and during the first few weeks of pregnancy compared 64 women with rheumatoid arthritis (RA) who became pregnant while taking LEF with 108 pregnant women with RA not on LEF and 78 healthy pregnant women [153]. LEF was discontinued in all exposed patients, with the last dose at a mean of 3.1 weeks postconception (range 0 to 8.6 weeks). Most women (95 percent) were treated with cholestyramine, resulting in undetectable levels at a mean of 10.7 weeks postconception. There were no significant differences in the rate of major structural defects in the exposed patients compared with either control group (5.4, 4.2, and 4.2 percent, respectively). No specific pattern of major or minor anomalies was seen, nor was there an increase in rates of pregnancy loss.

Third-trimester use of NSAIDs — Nonsteroidal antiinflammatory drugs (NSAIDs) should not be used during the third trimester because of the risk of premature closure of the ductus arteriosus. The inhibition of prostaglandin synthesis by NSAIDs or high-dose aspirin therapy in the third trimester has the potential for causing premature closure of the ductus arteriosus; indomethacin and ibuprofen appear to have strong ductal effects [156,157].

Aspirin (moderate to high dose) — We avoid the use of moderate- to high-dose salicylates in pregnant women and during lactation, although there is much less evidence available regarding the safety of higher-dose aspirin in such patients, compared with the available information regarding other NSAIDs and low-dose aspirin regimens. In particular, high-dose aspirin should be avoided during the third trimester because of the risk of premature closure of the ductus arteriosus.

Salicylates can cross the placenta and enter fetal circulation. Moderate or high doses of aspirin have been reported to be associated with fetal mortality, intrauterine growth retardation, salicylate intoxication, abnormal bleeding, and neonatal metabolic acidosis [27,158,159]. As an example, there has been a case report of metabolic acidosis in an infant whose mother was taking 2.4 g of aspirin a day [160]. High-dose aspirin near delivery may also increase the risk of fetal or neonatal bleeding or bruising, although data are inconsistent [20,156,161,162].

Reported maternal adverse effects include anemia, abnormal bleeding, and prolongation of gestation and of labor.

The safety of low-dose aspirin is described separately. (See 'Aspirin (low dose)' above.)

LIMITED INFORMATION

Overview of drugs with limited information — There is only limited information on which to base conclusions regarding the safety of the following medications in human pregnancy. However, for the biologic agents composed of an immunoglobulin (Ig) G1 component, placental transfer would be negligible during the first trimester. Thus, continuing these medications through conception is reasonable. There are insufficient data to make recommendations regarding the use during pregnancy of the small molecules.

Drugs for which there are limited data include:

Biologics with limited data:

Abatacept

Anakinra

Belimumab

Ixekizumab

Rituximab

Secukinumab

Tocilizumab

Ustekinumab

Small molecules with limited data:

Tofacitinib

There is generally insufficient evidence related to the risks associated with the use of certain biologic agents and small molecules in pregnancy. Drugs for which the risk is unknown include:

Biologic agents with unknown risk:

Canakinumab

Sarilumab

Guselkumab

Risankizumab

Small molecules with unknown risk:

Apremilast

Baricitinib

Upadacitinib

Biologics with limited data

Abatacept

PregnancyAbatacept can be used up through conception.

LactationAbatacept is a large molecule, and it is unlikely that significant amounts of this medication will be transferred to breast milk. While there are limited data on its compatibility with breastfeeding, given its large size, very little of this drug is likely to be transferred into breast milk.

Abatacept (cytotoxic T-lymphocyte-associated protein 4-Ig [CTLA4-Ig]) is an inhibitor of T-cell costimulation. Specific details regarding the use and monitoring of this medication are presented separately. (See "Treatment of rheumatoid arthritis in adults resistant to initial conventional synthetic (nonbiologic) DMARD therapy", section on 'Methotrexate plus abatacept' and "Treatment of rheumatoid arthritis in adults resistant to initial biologic DMARD therapy", section on 'Abatacept'.)

Abatacept interferes with T-cell activation by binding to accessory molecules (CD80 and CD86) and by blocking interaction with CD28 (CTLA4) [163]. Abatacept is not teratogenic in animals, but there are no adequate studies of its effects on human pregnancy to allow an assessment of its risk.

Anakinra

PregnancyAnakinra can be used through conception.

LactationAnakinra is a large molecule, and it is unlikely that significant amounts of this medication will be transferred to breast milk. While there are limited data on its compatibility with breastfeeding, given its large size, very little of this drug is likely to be transferred into breast milk.

Anakinra is a recombinant human interleukin 1 receptor antagonist (IL-1RA). Specific details regarding the use and monitoring of this medication are presented separately. (See "Overview of biologic agents in the rheumatic diseases", section on 'IL-1 inhibition'.)

Studies on rats and rabbits using anakinra at doses up to 100-fold higher than those employed in humans did not indicate problems relating either to infertility or fetal harm [164]. There have been no adequate studies of these issues in humans, although several case reports of women with adult-onset Still's disease treated through pregnancy resulted in healthy neonates [165,166]. A systematic review reported that IL-1 inhibition during 88 pregnancies was not associated with an increased risk of perinatal outcomes [167].

Anakinra (recombinant IL-1RA) is a nonglycosylated form of the human cytokine IL-1RA. The native form is present in breast milk and is thought to contribute to the known antiinflammatory properties of milk. Thus, there appears to be no risk to a nursing infant from administration of anakinra.

Belimumab — Belimumab is a human monoclonal antibody that inhibits the soluble form of a B-cell survival factor (known as BlyS or BAFF). Specific details regarding the use and monitoring of this medication are presented separately. (See "Overview of the management and prognosis of systemic lupus erythematosus in adults", section on 'Approach to drug therapy'.)

Pregnancy – Across 18 clinical trials, birth defects were more common among belimumab-exposed pregnancies compared with placebo-exposed pregnancies (5.6 versus 0 percent) [168]. However, no consistent pattern of birth defects was found suggesting that there is not an increased rate of congenital malformations with early in utero exposure to belimumab. The American College of Rheumatology 2020 Reproductive Health Guidelines suggest that this medication can be continued up until conception [169].

Lactation – No data are available to assess safety of belimumab in breastfeeding mothers.

Ixekizumab — Ixekizumab is a monoclonal antibody that selectively targets IL-17A. It is used for the treatment of plaque psoriasis.

Pregnancy – One study of 99 pregnancies in which mothers were exposed to ixekizumab showed no safety signals [170].

LactationIxekizumab is a large molecule and will have low transfer into breast milk.

Rituximab

PregnancyRituximab can be used through conception. In life-threatening conditions, this medication can be used during pregnancy.

Lactation – Transmission of rituximab in breast milk is low [171,172]. Women receiving this medication can breastfeed their infants.

Rituximab is a chimeric monoclonal antibody that leads to peripheral B-cell depletion through targeting the CD20 antigen present on B lymphocytes, the precursors of plasma cells. Specific details regarding the use and monitoring of rituximab are presented separately. (See "Rituximab: Principles of use and adverse effects in rheumatoid arthritis".)

While the manufacturer recommends discontinuing this medication for one year prior to conception, little if any rituximab is likely to be transferred through the placenta before 12 weeks of gestation. Therefore, if medically indicated, this medication can be used through conception. In life-threatening conditions, it can be used throughout pregnancy after a detailed discussion of the potential risks and benefits of such therapy

Rituximab has been detected in high concentrations in umbilical cord blood. Case reports in humans provide information about the use of rituximab in the setting of pregnancy. The range of data is summarized by the following points:

Rituximab caused complete but transient B cell depletion in the child of a mother with Burkitt lymphoma who received rituximab and CHOP (cyclophosphamide, doxorubicin, vincristine, prednisone) chemotherapy during pregnancy [173]. The infant demonstrated B-cell count recovery, a normal immunophenotype, and normal responses to vaccinations.

A patient under treatment for Hodgkin lymphoma inadvertently received rituximab during the first trimester of pregnancy [174]. The course of pregnancy remained normal, and a healthy, full-term baby was delivered.

A patient with non-Hodgkin lymphoma was treated in the second trimester with rituximab [175]. A healthy baby was delivered at 35 weeks. Peripheral B-cell counts measured when the infant was four weeks old were normal.

Fetal and neonatal outcomes have been reported for a series of pregnancies with rituximab exposure (for malignancy or autoimmune illness), either preconception or during pregnancy [176]. Of 153 pregnancies with known outcomes, 90 resulted in live births. Twenty-two infants were born prematurely, with one neonatal death at six weeks. Eleven neonates had hematologic abnormalities; none had corresponding infections. Four neonatal infections were reported (fever, bronchiolitis, cytomegalovirus hepatitis, and chorioamnionitis). Two congenital malformations were identified: clubfoot in one twin and cardiac malformation in a singleton birth, both in association with maternal exposure prior to conception. Of 21 pregnancies in which rituximab exposure occurred in pregnant women, only 2 patients received medication during the first trimester, and 6 out of 21 patients were exposed to multiagent chemotherapy. Seven neonates had cytopenias, but no congenital anomalies were reported.

Secukinumab

Pregnancy – Data from the Novartis global safety database that includes 292 pregnancies did not show any safety signals [177].

LactationSecukinumab is a large molecule, and it is unlikely that significant amounts of this medication will be transferred to breast milk. While there are limited data on its compatibility with breastfeeding, given its large size, very little of this drug is likely to be transferred into breast milk.

Secukinumab is a human IgG1K monoclonal antibody that binds to IL-17A and is used to treat psoriasis, ankylosing spondylitis, and psoriatic arthritis.

Tocilizumab

PregnancyTocilizumab can be used through conception.

LactationTocilizumab is a large molecule and it is unlikely that significant amounts of this medication will be transferred to breast milk. While there are limited data on its compatibility with breastfeeding, given its large size, very little of this drug is likely to be transferred into breast milk.

Tocilizumab is an IL-6 receptor inhibitor. Specific details regarding the use and monitoring of this medication are presented separately. (See "Treatment of rheumatoid arthritis in adults resistant to initial conventional synthetic (nonbiologic) DMARD therapy", section on 'Methotrexate plus IL-6 inhibitor/IL-6 inhibitor monotherapy' and "Treatment of rheumatoid arthritis in adults resistant to initial biologic DMARD therapy", section on 'Tocilizumab'.)

Based upon animal data, tocilizumab may cause fetal harm, but there are no adequate studies of its effects on human pregnancy to allow an assessment of its risk [178].

Pregnancy outcomes have been reported for 32 pregnancies, which were associated with 13 elective abortions, 7 spontaneous abortions (5 also exposed to methotrexate [MTX]), 1 newborn infant death due to respiratory distress after fetomaternal hemorrhage due to placenta previa, and 10 healthy newborns [179]. No teratogenic effects were observed.

In another case series of 50 pregnancies exposed to tocilizumab with known outcomes, 36 resulted in live births. While there were 5 low-birthweight infants born and 1 case of neonatal asphyxia, there were no reports of congenital anomalies [180].

In an additional 16 cases of maternal exposure during pregnancies, there were 4 spontaneous abortions, 1 of which was in a case with fetal hydrops of unclear etiology, and 1 pregnancy termination. Of the 11 live births, there were no congenital anomalies [181].

Ustekinumab

Pregnancy – Very limited data have not revealed adverse pregnancy outcomes after ustekinumab exposure; this medication can be used through conception [182].

LactationUstekinumab is a large molecule and it is unlikely that significant amounts of this medication will be transferred to breast milk. While there are limited data on its compatibility with breastfeeding, given its large size, very little of this drug is likely to be transferred into breast milk.

Ustekinumab is a monoclonal antibody that is directed against IL-12 and IL-23 and is predominantly used to treat psoriasis and inflammatory bowel disease (IBD).

Two case series, one of three pregnancies, the other of one pregnancy, have not shown increased risk of adverse pregnancy outcome [183,184]. Registry data have not shown an increase in the rate of spontaneous abortions or congenital malformations.

Small molecules with limited data

Tofacitinib

Pregnancy – We avoid tofacitinib during pregnancy as there is insufficient data to conclude its safety; based upon European Alliance of Associations for Rheumatology (EULAR) recommendations, discontinue this medication two months prior to conception.

Lactation – We avoid tofacitinib during lactation in women who are breastfeeding. There are insufficient data to address the issue of safety for breastfed infants, but given that it is a small molecule, it is likely to be transferred into breast milk at high concentrations.

Tofacitinib is an oral Janus kinase inhibitor used in the management of rheumatoid arthritis (RA) and other conditions. There are insufficient data regarding effects of this agent during pregnancy and lactation. In animals, the medication is teratogenic. While there is limited information regarding safety in human pregnancy, one report described outcomes in 47 pregnancies following tofacitinib exposure [185]. Thirteen of these women were also receiving MTX, and, in one, the exposure was blinded. There were seven spontaneous miscarriages, eight medical terminations, and one congenital anomaly (malformation of the pulmonic valve). In 25 births, there were no anomalies; six were lost to follow-up.

COX-2 inhibitors — There are insufficient data on the safety of cyclooxygenase 2 (COX-2) inhibitors during pregnancy; therefore, these drugs should be avoided.

OTHER RESOURCES — The LactMed database, produced by the National Library of Medicine in the United States, is a free, authoritative reference for lactation compatibility for prescription and over-the-counter drugs. This resource provides the most current available data on drug levels in human milk and infant serum, potential adverse effects on breastfeeding infants and lactation, and recommendations for alternative drugs. A free app is also available.

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: Side effects of anti-inflammatory and anti-rheumatic drugs" and "Society guideline links: Breastfeeding and infant nutrition".)

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: Rheumatoid arthritis and pregnancy (Beyond the Basics)" and "Patient education: Disease-modifying antirheumatic drugs (DMARDs) in rheumatoid arthritis (Beyond the Basics)" and "Patient education: Inflammatory bowel disease and pregnancy (Beyond the Basics)" and "Patient education: Systemic lupus erythematosus and pregnancy (Beyond the Basics)")

SUMMARY AND RECOMMENDATIONS

Hydroxychloroquine (HCQ), sulfasalazine (SSZ), azathioprine (AZA), and 6-mercaptopurine (6-MP) are considered compatible with pregnancy. (See 'Minimal fetal or maternal risk' above.)

Low-dose glucocorticoid (eg, 5 to 15 mg of prednisone per day) is compatible with pregnancy, although higher doses and use later in pregnancy increases the risk of gestational diabetes and pregnancy-induced hypertension. (See 'Glucocorticoids' above.)

In women with a history of delayed conception, discontinuing nonsteroidal antiinflammatory drugs (NSAIDs) may be reasonable. Risk of NSAID use during the first trimester cannot be excluded but appears low. NSAIDs should not be used beyond week 20 of gestation. Use of NSAIDs after the start of the third trimester carries the additional risk of premature closure of the ductus arteriosus. (See 'NSAIDs' above.)

Tumor necrosis factor (TNF)-alpha inhibitors, cyclosporine, tacrolimus, and intravenous immune globulin (IVIG) are compatible with use during pregnancy. Some experts discontinue TNF-alpha blockers during the third trimester, although use of these drugs can be extended, if necessary, to a later gestational age if benefits outweigh potential risks for an individual patient. Certolizumab is compatible with use throughout pregnancy. (See 'Minimal fetal or maternal risk' above and 'Selective use allowed during pregnancy' above.)

Infants exposed to TNF inhibitors in utero should avoid live vaccines during the first six months of life. (See 'Tumor necrosis factor inhibitors' above.)

Abatacept, anakinra, belimumab, ixekizumab, rituximab, secukinumab, and tocilizumab may be used up until conception. While there are limited or no data on other biologics, one can consider continuing these medications through conception. Tofacitinib and the other small molecules should be avoided in pregnant and lactating women. (See 'Limited information' above.)

In pregnant women, we limit the use of high-dose glucocorticoids, cyclophosphamide (CYC), and rituximab only to women with life-threatening rheumatologic autoimmune disease in whom the benefits of these agents is felt by the patient and clinician to outweigh the risks. (See 'Glucocorticoids' above and 'Cyclophosphamide' above and 'Rituximab' above.)

Leflunomide (LEF), methotrexate (MTX), and mycophenolate mofetil (MMF) should be avoided in pregnant women with autoimmune disease, and counseling should be offered in the setting of inadvertent exposure. (See 'Methotrexate' above and 'Leflunomide' above and 'Mycophenolate mofetil' above.)

  1. https://www.fda.gov/drugs/developmentapprovalprocess/developmentresources/labeling/ucm093307.htm (Accessed on October 27, 2017).
  2. Parke A, West B. Hydroxychloroquine in pregnant patients with systemic lupus erythematosus. J Rheumatol 1996; 23:1715.
  3. Buchanan NM, Toubi E, Khamashta MA, et al. Hydroxychloroquine and lupus pregnancy: review of a series of 36 cases. Ann Rheum Dis 1996; 55:486.
  4. Cooper WO, Cheetham TC, Li DK, et al. Brief report: Risk of adverse fetal outcomes associated with immunosuppressive medications for chronic immune-mediated diseases in pregnancy. Arthritis Rheumatol 2014; 66:444.
  5. Huybrechts KF, Bateman BT, Zhu Y, et al. Hydroxychloroquine early in pregnancy and risk of birth defects. Am J Obstet Gynecol 2021; 224:290.e1.
  6. Bermas BL, Chambers C. Hydroxychloroquine early in pregnancy and risk of birth defects: don't throw out the baby with the bathwater. Am J Obstet Gynecol 2021; 224:548.
  7. Chambers CD, Johnson DL, Xu R, et al. Birth Outcomes in Women Who Have Taken Hydroxycholoroquine During Pregnancy: A Prospective Cohort Study. Arthritis Rheumatol 2022; 74:711.
  8. Clowse ME, Magder L, Witter F, Petri M. Hydroxychloroquine in lupus pregnancy. Arthritis Rheum 2006; 54:3640.
  9. Liu Y, Zhang Y, Wei Y, Yang H. Effect of hydroxychloroquine on preeclampsia in lupus pregnancies: a propensity score-matched analysis and meta-analysis. Arch Gynecol Obstet 2021; 303:435.
  10. Do SC, Rizk NM, Druzin ML, Simard JF. Does Hydroxychloroquine Protect against Preeclampsia and Preterm Delivery in Systemic Lupus Erythematosus Pregnancies? Am J Perinatol 2020; 37:873.
  11. Costedoat-Chalumeau N, Amoura Z, Duhaut P, et al. Safety of hydroxychloroquine in pregnant patients with connective tissue diseases: a study of one hundred thirty-three cases compared with a control group. Arthritis Rheum 2003; 48:3207.
  12. Canadian rheumatology association. Canadian Consensus Conference on hydroxychloroquine. J Rheumatol 2000; 27:2919.
  13. Järnerot G, Into-Malmberg MB, Esbjörner E. Placental transfer of sulphasalazine and sulphapyridine and some of its metabolites. Scand J Gastroenterol 1981; 16:693.
  14. 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.
  15. 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.
  16. Levy N, Roisman I, Teodor I. Ulcerative colitis in pregnancy in Israel. Dis Colon Rectum 1981; 24:351.
  17. Esbjörner E, Järnerot G, Wranne L. Sulphasalazine and sulphapyridine serum levels in children to mothers treated with sulphasalazine during pregnancy and lactation. Acta Paediatr Scand 1987; 76:137.
  18. Järnerot G, Into-Malmberg MB. Sulphasalazine treatment during breast feeding. Scand J Gastroenterol 1979; 14:869.
  19. Branski D, Kerem E, Gross-Kieselstein E, et al. Bloody diarrhea--a possible complication of sulfasalazine transferred through human breast milk. J Pediatr Gastroenterol Nutr 1986; 5:316.
  20. Drug Facts and Comparisons. In: Facts and Comparison, 50th, Wolters Kluwer, St. Louis 1996.
  21. Datta P, Rewers-Felkins K, Kallem RR, et al. Transfer of Low Dose Aspirin Into Human Milk. J Hum Lact 2017; 33:296.
  22. Kozer E, Nikfar S, Costei A, et al. Aspirin consumption during the first trimester of pregnancy and congenital anomalies: a meta-analysis. Am J Obstet Gynecol 2002; 187:1623.
  23. Collins E, Turner G. Maternal effects of regular salicylate ingestion in pregnancy. Lancet 1975; 2:335.
  24. Slone D, Siskind V, Heinonen OP, et al. Aspirin and congenital malformations. Lancet 1976; 1:1373.
  25. Askie LM, Duley L, Henderson-Smart DJ, et al. Antiplatelet agents for prevention of pre-eclampsia: a meta-analysis of individual patient data. Lancet 2007; 369:1791.
  26. Wang L, Huang X, Li X, et al. Efficacy evaluation of low-dose aspirin in IVF/ICSI patients evidence from 13 RCTs: A systematic review and meta-analysis. Medicine (Baltimore) 2017; 96:e7720.
  27. Østensen M, Khamashta M, Lockshin M, et al. Anti-inflammatory and immunosuppressive drugs and reproduction. Arthritis Res Ther 2006; 8:209.
  28. Sau A, Clarke S, Bass J, et al. Azathioprine and breastfeeding: is it safe? BJOG 2007; 114:498.
  29. Moretti ME, Verjee Z, Ito S, Koren G. Breast-feeding during maternal use of azathioprine. Ann Pharmacother 2006; 40:2269.
  30. Saarikoski S, Seppälä M. Immunosuppression during pregnancy: transmission of azathioprine and its metabolites from the mother to the fetus. Am J Obstet Gynecol 1973; 115:1100.
  31. 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.
  32. 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.
  33. Moskovitz DN, Bodian C, Chapman ML, et al. The effect on the fetus of medications used to treat pregnant inflammatory bowel-disease patients. Am J Gastroenterol 2004; 99:656.
  34. Armenti VT, Ahlswede KM, Ahlswede BA, et al. National transplantation Pregnancy Registry--outcomes of 154 pregnancies in cyclosporine-treated female kidney transplant recipients. Transplantation 1994; 57:502.
  35. WHO Working Group on Drugs and Human Lactation. Drugs and Human Lactation. Elsevier; Amsterdam, 1988.
  36. EBPG Expert Group on Renal Transplantation. European best practice guidelines for renal transplantation. Section IV: Long-term management of the transplant recipient. IV.10. Pregnancy in renal transplant recipients. Nephrol Dial Transplant 2002; 17 Suppl 4:50.
  37. Coulam CB, Moyer TP, Jiang NS, Zincke H. Breast-feeding after renal transplantation. Transplant Proc 1982; 14:605.
  38. Grekas DM, Vasiliou SS, Lazarides AN. Immunosuppressive therapy and breast-feeding after renal transplantation. Nephron 1984; 37:68.
  39. Flint J, Panchal S, Hurrell A, et al. BSR and BHPR guideline on prescribing drugs in pregnancy and breastfeeding-Part I: standard and biologic disease modifying anti-rheumatic drugs and corticosteroids. Rheumatology (Oxford) 2016; 55:1693.
  40. Götestam Skorpen C, Hoeltzenbein M, Tincani A, et al. The EULAR points to consider for use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Ann Rheum Dis 2016; 75:795.
  41. Indraratna PL, Virk S, Gurram D, Day RO. Use of colchicine in pregnancy: a systematic review and meta-analysis. Rheumatology (Oxford) 2018; 57:382.
  42. Guillonneau M, Aigrain EJ, Galliot M, et al. Colchicine is excreted at high concentrations in human breast milk. Eur J Obstet Gynecol Reprod Biol 1995; 61:177.
  43. Ben-Chetrit E, Scherrmann JM, Levy M. Colchicine in breast milk of patients with familial Mediterranean fever. Arthritis Rheum 1996; 39:1213.
  44. American Academy of Pediatrics Committee on Drugs. Transfer of drugs and other chemicals into human milk. Pediatrics 2001; 108:776.
  45. Cohen MM, Levy M, Eliakim M. A cytogenic evaluation of long-term colchicine therapy in the treatment of Familial Mediterranean fever (FMF). Am J Med Sci 1977; 274:147.
  46. Mendonça LL, Khamashta MA, Nelson-Piercy C, et al. Non-steroidal anti-inflammatory drugs as a possible cause for reversible infertility. Rheumatology (Oxford) 2000; 39:880.
  47. Uhler ML, Hsu JW, Fisher SG, Zinaman MJ. The effect of nonsteroidal anti-inflammatory drugs on ovulation: a prospective, randomized clinical trial. Fertil Steril 2001; 76:957.
  48. Pall M, Fridén BE, Brännström M. Induction of delayed follicular rupture in the human by the selective COX-2 inhibitor rofecoxib: a randomized double-blind study. Hum Reprod 2001; 16:1323.
  49. Daniel S, Matok I, Gorodischer R, et al. Major malformations following exposure to nonsteroidal antiinflammatory drugs during the first trimester of pregnancy. J Rheumatol 2012; 39:2163.
  50. Nezvalová-Henriksen K, Spigset O, Nordeng H. Effects of ibuprofen, diclofenac, naproxen, and piroxicam on the course of pregnancy and pregnancy outcome: a prospective cohort study. BJOG 2013; 120:948.
  51. Choi EY, Jeong HE, Noh Y, et al. Neonatal and maternal adverse outcomes and exposure to nonsteroidal anti-inflammatory drugs during early pregnancy in South Korea: A nationwide cohort study. PLoS Med 2023; 20:e1004183.
  52. Edwards DR, Aldridge T, Baird DD, et al. Periconceptional over-the-counter nonsteroidal anti-inflammatory drug exposure and risk for spontaneous abortion. Obstet Gynecol 2012; 120:113.
  53. Daniel S, Koren G, Lunenfeld E, et al. Fetal exposure to nonsteroidal anti-inflammatory drugs and spontaneous abortions. CMAJ 2014; 186:E177.
  54. Nakhai-Pour HR, Broy P, Sheehy O, Bérard A. Use of nonaspirin nonsteroidal anti-inflammatory drugs during pregnancy and the risk of spontaneous abortion. CMAJ 2011; 183:1713.
  55. Nielsen GL, Sørensen HT, Larsen H, Pedersen L. Risk of adverse birth outcome and miscarriage in pregnant users of non-steroidal anti-inflammatory drugs: population based observational study and case-control study. BMJ 2001; 322:266.
  56. Li DK, Liu L, Odouli R. Exposure to non-steroidal anti-inflammatory drugs during pregnancy and risk of miscarriage: population based cohort study. BMJ 2003; 327:368.
  57. Li DK, Ferber JR, Odouli R, Quesenberry C. Use of nonsteroidal antiinflammatory drugs during pregnancy and the risk of miscarriage. Am J Obstet Gynecol 2018; 219:275.e1.
  58. US FDA Drug Safety Communication. FDA recommends avoiding use of NSAIDs in pregnancy at 20 weeks or later because they can result in low amniotic fluid. https://www.fda.gov/media/142967/download (Accessed on November 19, 2020).
  59. Townsend RJ, Benedetti TJ, Erickson SH, et al. Excretion of ibuprofen into breast milk. Am J Obstet Gynecol 1984; 149:184.
  60. MacKenzie CR, Goodman SM. Stress Dose Steroids: Myths and Perioperative Medicine. Curr Rheumatol Rep 2016; 18:47.
  61. Goodman SM, Springer B, Guyatt G, et al. 2017 American College of Rheumatology/American Association of Hip and Knee Surgeons Guideline for the Perioperative Management of Antirheumatic Medication in Patients With Rheumatic Diseases Undergoing Elective Total Hip or Total Knee Arthroplasty. Arthritis Care Res (Hoboken) 2017; 69:1111.
  62. Beitins IZ, Bayard F, Ances IG, et al. The transplacental passage of prednisone and prednisolone in pregnancy near term. J Pediatr 1972; 81:936.
  63. 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.
  64. PINSKY L, DIGEORGE AM. CLEFT PALATE IN THE MOUSE: A TERATOGENIC INDEX OF GLUCOCORTICOID POTENCY. Science 1965; 147:402.
  65. Hviid A, Mølgaard-Nielsen D. Corticosteroid use during pregnancy and risk of orofacial clefts. CMAJ 2011; 183:796.
  66. Mossey PA, Little J, Munger RG, et al. Cleft lip and palate. Lancet 2009; 374:1773.
  67. Guller S, Kong L, Wozniak R, Lockwood CJ. Reduction of extracellular matrix protein expression in human amnion epithelial cells by glucocorticoids: a potential role in preterm rupture of the fetal membranes. J Clin Endocrinol Metab 1995; 80:2244.
  68. Lockwood CJ, Radunovic N, Nastic D, et al. Corticotropin-releasing hormone and related pituitary-adrenal axis hormones in fetal and maternal blood during the second half of pregnancy. J Perinat Med 1996; 24:243.
  69. LaRochelle GE Jr, LaRochelle AG, Ratner RE, Borenstein DG. Recovery of the hypothalamic-pituitary-adrenal (HPA) axis in patients with rheumatic diseases receiving low-dose prednisone. Am J Med 1993; 95:258.
  70. Ost L, Wettrell G, Björkhem I, Rane A. Prednisolone excretion in human milk. J Pediatr 1985; 106:1008.
  71. Heller MM, Wu JJ, Murase JE. Fatal case of disseminated BCG infection after vaccination of an infant with in utero exposure to infliximab. J Am Acad Dermatol 2011; 65:870.
  72. Sheibani S, Cohen R, Kane S, et al. The effect of maternal peripartum anti-TNFa use on infant immune response. Dig Dis Sci 2016.
  73. Roux CH, Brocq O, Breuil V, et al. Pregnancy in rheumatology patients exposed to anti-tumour necrosis factor (TNF)-alpha therapy. Rheumatology (Oxford) 2007; 46:695.
  74. Vinet E, Pineau C, Gordon C, et al. Biologic therapy and pregnancy outcomes in women with rheumatic diseases. Arthritis Rheum 2009; 61:587.
  75. Ali YM, Kuriya B, Orozco C, et al. Can tumor necrosis factor inhibitors be safely used in pregnancy? J Rheumatol 2010; 37:9.
  76. Chambers C, Johnson DL, Jones KL, the OTIS Collaborative Research Group. Pregnancy outcome in women exposed to anti-TNF-alpha medications: the OTIS Rheumatoid Arthritis in Pregnancy Study. Arthritis Rheum 2004; 50:S479.
  77. Hyrich KL, Symmons DP, Watson KD, et al. Pregnancy outcome in women who were exposed to anti-tumor necrosis factor agents: results from a national population register. Arthritis Rheum 2006; 54:2701.
  78. 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.
  79. Ventura SJ, Mosher WD, Curtin SC, et al. Trends in pregnancies and pregnancy rates by outcome: estimates for the United States, 1976-96. Vital Health Stat 21 2000; :1.
  80. Coburn LA, Wise PE, Schwartz DA. The successful use of adalimumab to treat active Crohn's disease of an ileoanal pouch during pregnancy. Dig Dis Sci 2006; 51:2045.
  81. Strangfeld A. Pregnancy outcome after exposure to biologics: results from the German biologics register RABBIT. Arthritis Rheum 2007; 56:S311.
  82. Clowse ME, Wolf DC, Förger F, et al. Pregnancy Outcomes in Subjects Exposed to Certolizumab Pegol. J Rheumatol 2015; 42:2270.
  83. Clowse MEB, Scheuerle AE, Chambers C, et al. Pregnancy Outcomes After Exposure to Certolizumab Pegol: Updated Results From a Pharmacovigilance Safety Database. Arthritis Rheumatol 2018; 70:1399.
  84. 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.
  85. Reprotox Agent Detail - Adalimumab www.reprotox.org/Members/AgentDetail.aspx?a=4262 (Accessed on March 25, 2010).
  86. Weber-Schoendorfer C, Oppermann M, Wacker E, et al. Pregnancy outcome after TNF-α inhibitor therapy during the first trimester: a prospective multicentre cohort study. Br J Clin Pharmacol 2015; 80:727.
  87. 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.
  88. Martin PL, Oneda S, Treacy G. Effects of an anti-TNF-alpha monoclonal antibody, administered throughout pregnancy and lactation, on the development of the macaque immune system. Am J Reprod Immunol 2007; 58:138.
  89. Vinet É, De Moura C, Pineau CA, et al. Serious Infections in Rheumatoid Arthritis Offspring Exposed to Tumor Necrosis Factor Inhibitors: A Cohort Study. Arthritis Rheumatol 2018; 70:1565.
  90. Chambers CD, Johnson DL, Luo Y, et al. Serious or Opportunistic Infections in Infants Born to Pregnant Women with Rheumatoid Arthritis and Treated with a Biologic Medication [abstract]. Arthritis Rheumatol. 2017; 69 (suppl 10). http://acrabstracts.org/abstract/serious-or-opportunistic-infections-in-infants-born-to-pregnant-women-with-rheumatoid-arthritis-and-treated-with-a-biologic-medication/. (Accessed on November 13, 2017).
  91. Ostensen M, Eigenmann GO. Etanercept in breast milk. J Rheumatol 2004; 31:1017.
  92. Stengel JZ, Arnold HL. Is infliximab safe to use while breastfeeding? World J Gastroenterol 2008; 14:3085.
  93. Tanaka H, Haba R, Itoh S, et al. Prenatal high-dose immunoglobulin treatment for neonatal hemochromatosis: a case report and review of the literature. J Obstet Gynaecol Res 2011; 37:1891.
  94. Bakimer R, Guilburd B, Zurgil N, Shoenfeld Y. The effect of intravenous gamma-globulin on the induction of experimental antiphospholipid syndrome. Clin Immunol Immunopathol 1993; 69:97.
  95. Höckel M, Kaufmann R. Placental transfer of class G immunoglobulins treated with beta-propiolactone (beta-PL) for intravenous application--a case report. J Perinat Med 1986; 14:205.
  96. Potter M, Stockley R, Storry J, Slade R. ABO alloimmunisation after intravenous immunoglobulin infusion. Lancet 1988; 1:932.
  97. Reznikoff-Etievant MF, de Lachaux Y, Marpeau L, Couroucé A. Intravenous immunoglobulins and hepatitis C transmission in healthy pregnant women. Lancet 1992; 340:986.
  98. Achiron A, Kishner I, Dolev M, et al. Effect of intravenous immunoglobulin treatment on pregnancy and postpartum-related relapses in multiple sclerosis. J Neurol 2004; 251:1133.
  99. Bäckman L, Brandt I, Appelkvist EL, Dallner G. Tissue and subcellular localizations of 3H-cyclosporine A in mice. Pharmacol Toxicol 1988; 62:110.
  100. Schmid BP. Monitoring of organ formation in rat embryos after in vitro exposure to azathioprine, mercaptopurine, methotrexate or cyclosporin A. Toxicology 1984; 31:9.
  101. Mason RJ, Thomson AW, Whiting PH, et al. Cyclosporine-induced fetotoxicity in the rat. Transplantation 1985; 39:9.
  102. Nandakumaran M, Eldeen AS. Transfer of cyclosporine in the perfused human placenta. Dev Pharmacol Ther 1990; 15:101.
  103. Venkataramanan R, Koneru B, Wang CC, et al. Cyclosporine and its metabolites in mother and baby. Transplantation 1988; 46:468.
  104. Di Paolo S, Monno R, Stallone G, et al. Placental imbalance of vasoactive factors does not affect pregnancy outcome in patients treated with Cyclosporine A after transplantation. Am J Kidney Dis 2002; 39:776.
  105. Lewis GJ, Lamont CA, Lee HA, Slapak M. Successful pregnancy in a renal transplant recipient taking cyclosporin A. Br Med J (Clin Res Ed) 1983; 286:603.
  106. Flechner SM, Katz AR, Rogers AJ, et al. The presence of cyclosporine in body tissues and fluids during pregnancy. Am J Kidney Dis 1985; 5:60.
  107. Burrows DA, O'Neil TJ, Sorrells TL. Successful twin pregnancy after renal transplant maintained on cyclosporine A immunosuppression. Obstet Gynecol 1988; 72:459.
  108. Löwenstein BR, Vain NW, Perrone SV, et al. Successful pregnancy and vaginal delivery after heart transplantation. Am J Obstet Gynecol 1988; 158:589.
  109. Ziegenhagen DJ, Crombach G, Dieckmann M, et al. [Pregnancy during cyclosporin medication following a kidney transplant]. Dtsch Med Wochenschr 1988; 113:260.
  110. al-Khader AA, Absy M, al-Hasani MK, et al. Successful pregnancy in renal transplant recipients treated with cyclosporine. Transplantation 1988; 45:987.
  111. Sims CJ, Porter KB, Knuppel RA. Successful pregnancy after a liver transplant. Am J Obstet Gynecol 1989; 161:532.
  112. Kossoy LR, Herbert CM 3rd, Wentz AC. Management of heart transplant recipients: guidelines for the obstetrician-gynecologist. Am J Obstet Gynecol 1988; 159:490.
  113. Munoz-Flores-Thiagarajan KD, Easterling T, Davis C, Bond EF. Breast-feeding by a cyclosporine-treated mother. Obstet Gynecol 2001; 97:816.
  114. Moretti ME, Sgro M, Johnson DW, et al. Cyclosporine excretion into breast milk. Transplantation 2003; 75:2144.
  115. Cockburn I, Krupp P, Monka C. Present experience of Sandimmun in pregnancy. Transplant Proc 1989; 21:3730.
  116. Bar Oz B, Hackman R, Einarson T, Koren G. Pregnancy outcome after cyclosporine therapy during pregnancy: a meta-analysis. Transplantation 2001; 71:1051.
  117. Petri M. Immunosuppressive drug use in pregnancy. Autoimmunity 2003; 36:51.
  118. Nyberg G, Haljamäe U, Frisenette-Fich C, et al. Breast-feeding during treatment with cyclosporine. Transplantation 1998; 65:253.
  119. Osadchy A, Koren G. Cyclosporine and lactation: when the mother is willing to breastfeed. Ther Drug Monit 2011; 33:147.
  120. Constantinescu S, Pai A, Coscia LA, et al. Breast-feeding after transplantation. Best Pract Res Clin Obstet Gynaecol 2014; 28:1163.
  121. Briggs GG, Freeman RK, Yaffe SJ. Drugs in Pregnancy and Lactation, 9th, Lippincott Williams & Wilkins, Philadelphia 2011. p.1381.
  122. Kainz A, Harabacz I, Cowlrick IS, et al. Review of the course and outcome of 100 pregnancies in 84 women treated with tacrolimus. Transplantation 2000; 70:1718.
  123. Jain A, Venkataramanan R, Fung JJ, et al. Pregnancy after liver transplantation under tacrolimus. Transplantation 1997; 64:559.
  124. Jain AB, Shapiro R, Scantlebury VP, et al. Pregnancy after kidney and kidney-pancreas transplantation under tacrolimus: a single center's experience. Transplantation 2004; 77:897.
  125. Kitada A, Nakai T, Fukui S, et al. Safety of tacrolimus use during pregnancy and related pregnancy outcomes in patients with systemic lupus erythematosus: A retrospective single-center analysis in Japan. Lupus 2023; 32:352.
  126. French AE, Soldin SJ, Soldin OP, Koren G. Milk transfer and neonatal safety of tacrolimus. Ann Pharmacother 2003; 37:815.
  127. Gardiner SJ, Begg EJ. Breastfeeding during tacrolimus therapy. Obstet Gynecol 2006; 107:453.
  128. Kart Köseoglu H, Yücel AE, Künefeci G, et al. Cyclophosphamide therapy in a serious case of lupus nephritis during pregnancy. Lupus 2001; 10:818.
  129. Berry DL, Theriault RL, Holmes FA, et al. Management of breast cancer during pregnancy using a standardized protocol. J Clin Oncol 1999; 17:855.
  130. Ring AE, Smith IE, Jones A, et al. Chemotherapy for breast cancer during pregnancy: an 18-year experience from five London teaching hospitals. J Clin Oncol 2005; 23:4192.
  131. Clowse ME, Magder L, Petri M. Cyclophosphamide for lupus during pregnancy. Lupus 2005; 14:593.
  132. Wiernik PH, Duncan JH. Cyclophosphamide in human milk. Lancet 1971; 1:912.
  133. Johns DG, Rutherford LD, Leighton PC, Vogel CL. Secretion of methotrexate into human milk. Am J Obstet Gynecol 1972; 112:978.
  134. Skalko RG, Gold MP. Teratogenicity of methotrexate in mice. Teratology 1974; 9:159.
  135. Buckley LM, Bullaboy CA, Leichtman L, Marquez M. Multiple congenital anomalies associated with weekly low-dose methotrexate treatment of the mother. Arthritis Rheum 1997; 40:971.
  136. Milunsky A, Graef JW, Gaynor MF Jr. Methotrexate-induced congenital malformations. J Pediatr 1968; 72:790.
  137. Kozlowski RD, Steinbrunner JV, MacKenzie AH, et al. Outcome of first-trimester exposure to low-dose methotrexate in eight patients with rheumatic disease. Am J Med 1990; 88:589.
  138. Ostensen M, Hartmann H, Salvesen K. Low dose weekly methotrexate in early pregnancy. A case series and review of the literature. J Rheumatol 2000; 27:1872.
  139. 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.
  140. Lewden B, Vial T, Elefant E, et al. Low dose methotrexate in the first trimester of pregnancy: results of a French collaborative study. J Rheumatol 2004; 31:2360.
  141. Weber-Schoendorfer C, Chambers C, Wacker E, et al. Pregnancy outcome after methotrexate treatment for rheumatic disease prior to or during early pregnancy: a prospective multicenter cohort study. Arthritis Rheumatol 2014; 66:1101.
  142. Thorne JC, Nadarajah T, Moretti M, Ito S. Methotrexate use in a breastfeeding patient with rheumatoid arthritis. J Rheumatol 2014; 41:2332.
  143. Sifontis NM, Coscia LA, Constantinescu S, et al. Pregnancy outcomes in solid organ transplant recipients with exposure to mycophenolate mofetil or sirolimus. Transplantation 2006; 82:1698.
  144. www.rocheusa.com/products/cellcept/pregnancy_notice.pdf (Accessed on September 14, 2011).
  145. Hoeltzenbein M, Elefant E, Vial T, et al. Teratogenicity of mycophenolate confirmed in a prospective study of the European Network of Teratology Information Services. Am J Med Genet A 2012; 158A:588.
  146. Klieger-Grossmann C, Chitayat D, Lavign S, et al. Prenatal exposure to mycophenolate mofetil: an updated estimate. J Obstet Gynaecol Can 2010; 32:794.
  147. US Food and Drug Administration Information for Healthcare Professionals: Mycophenolate Mofetil (marketed as CellCept) and Mycophenolic Acid (marketed as Myfortic) http://www.fda.gov/drugs/drugsafety/postmarketdrugsafetyinformationforpatientsandproviders/ucm124776.htm (Accessed on June 02, 2014).
  148. Perez-Aytes A, Ledo A, Boso V, et al. In utero exposure to mycophenolate mofetil: a characteristic phenotype? Am J Med Genet A 2008; 146A:1.
  149. Brent RL. Teratogen update: reproductive risks of leflunomide (Arava); a pyrimidine synthesis inhibitor: counseling women taking leflunomide before or during pregnancy and men taking leflunomide who are contemplating fathering a child. Teratology 2001; 63:106.
  150. De Santis M, Straface G, Cavaliere A, et al. Paternal and maternal exposure to leflunomide: pregnancy and neonatal outcome. Ann Rheum Dis 2005; 64:1096.
  151. Heine K, Poets CF. A pair of twins born after maternal exposure to leflunomide. J Perinatol 2008; 28:841.
  152. Neville CE, McNally J. Maternal exposure to leflunomide associated with blindness and cerebral palsy. Rheumatology (Oxford) 2007; 46:1506.
  153. Chambers CD, Johnson DL, Robinson LK, et al. Birth outcomes in women who have taken leflunomide during pregnancy. Arthritis Rheum 2010; 62:1494.
  154. Cassina M, Johnson DL, Robinson LK, et al. Pregnancy outcome in women exposed to leflunomide before or during pregnancy. Arthritis Rheum 2012; 64:2085.
  155. Fukushima R, Kanamori S, Hirashiba M, et al. Teratogenicity study of the dihydroorotate-dehydrogenase inhibitor and protein tyrosine kinase inhibitor Leflunomide in mice. Reprod Toxicol 2007; 24:310.
  156. Koren G, Florescu A, Costei AM, et al. Nonsteroidal antiinflammatory drugs during third trimester and the risk of premature closure of the ductus arteriosus: a meta-analysis. Ann Pharmacother 2006; 40:824.
  157. Heymann MA, Rudolph AM, Silverman NH. Closure of the ductus arteriosus in premature infants by inhibition of prostaglandin synthesis. N Engl J Med 1976; 295:530.
  158. Corby DG. Aspirin in pregnancy: maternal and fetal effects. Pediatrics 1978; 62:930.
  159. Ostensen M. Nonsteroidal anti-inflammatory drugs during pregnancy. Scand J Rheumatol Suppl 1998; 107:128.
  160. Clark JH, Wilson WG. A 16-day-old breast-fed infant with metabolic acidosis caused by salicylate. Clin Pediatr (Phila) 1981; 20:53.
  161. Heymann MA. Non-narcotic analgesics. Use in pregnancy and fetal and perinatal effects. Drugs 1986; 32 Suppl 4:164.
  162. Ostensen M, Ramsey-Goldman R. Treatment of inflammatory rheumatic disorders in pregnancy: what are the safest treatment options? Drug Saf 1998; 19:389.
  163. Abatacept (Orencia) for rheumatoid arthritis. Med Lett Drugs Ther 2006; 48:17.
  164. Physician's Desk Reference, Edition 58. Montvale, NJ, Thomson PDR, 2004; 31:1017.
  165. Berger CT, Recher M, Steiner U, Hauser TM. A patient's wish: anakinra in pregnancy. Ann Rheum Dis 2009; 68:1794.
  166. Fischer-Betz R, Specker C, Schneider M. Successful outcome of two pregnancies in patients with adult-onset Still's disease treated with IL-1 receptor antagonist (anakinra). Clin Exp Rheumatol 2011; 29:1021.
  167. Brien ME, Gaudreault V, Hughes K, et al. A Systematic Review of the Safety of Blocking the IL-1 System in Human Pregnancy. J Clin Med 2021; 11.
  168. Petri M, Landy H, Clowse MEB, et al. Belimumab use during pregnancy: a summary of birth defects and pregnancy loss from belimumab clinical trials, a pregnancy registry and postmarketing reports. Ann Rheum Dis 2023; 82:217.
  169. Sammaritano LR, Bermas BL, Chakravarty EE, et al. 2020 American College of Rheumatology Guideline for the Management of Reproductive Health in Rheumatic and Musculoskeletal Diseases. Arthritis Care Res (Hoboken) 2020; 72:461.
  170. Egeberg A, Iversen L, Kimball AB, et al. Pregnancy outcomes in patients with psoriasis, psoriatic arthritis, or axial spondyloarthritis receiving ixekizumab. J Dermatolog Treat 2022; 33:2503.
  171. Pistilli B, Bellettini G, Giovannetti E, et al. Chemotherapy, targeted agents, antiemetics and growth-factors in human milk: how should we counsel cancer patients about breastfeeding? Cancer Treat Rev 2013; 39:207.
  172. Bragnes Y, Boshuizen R, de Vries A, et al. Low level of Rituximab in human breast milk in a patient treated during lactation. Rheumatology (Oxford) 2017; 56:1047.
  173. Friedrichs B, Tiemann M, Salwender H, et al. The effects of rituximab treatment during pregnancy on a neonate. Haematologica 2006; 91:1426.
  174. Kimby E, Sverrisdottir A, Elinder G. Safety of rituximab therapy during the first trimester of pregnancy: a case history. Eur J Haematol 2004; 72:292.
  175. Herold M, Schnohr S, Bittrich H. Efficacy and safety of a combined rituximab chemotherapy during pregnancy. J Clin Oncol 2001; 19:3439.
  176. Chakravarty EF, Murray ER, Kelman A, Farmer P. Pregnancy outcomes after maternal exposure to rituximab. Blood 2011; 117:1499.
  177. Warren RB, Reich K, Langley RG, et al. Secukinumab in pregnancy: outcomes in psoriasis, psoriatic arthritis and ankylosing spondylitis from the global safety database. Br J Dermatol 2018; 179:1205.
  178. Full prescribing information for Actemra (tocilizumab) www.gene.com/gene/products/information/actemra/pdf/pi.pdf (Accessed on June 09, 2010).
  179. Rubbert-Roth A, Goupile PM, Moosavi S, et al. First experiences with pregnancies in RA patients receiving tocilizumab therapy (abstract). Arthritis Rheum 2010; Suppl:384.
  180. Nakajima K, Watanabe O, Mochizuki M, et al. Pregnancy outcomes after exposure to tocilizumab: A retrospective analysis of 61 patients in Japan. Mod Rheumatol 2016; 26:667.
  181. Weber-Schoendorfer C, Schaefer C. Pregnancy outcome after tocilizumab therapy in early pregnancy-a case series from the German Embryotox Pharmacovigilance Center. Reprod Toxicol 2016; 60:29.
  182. Gorodensky JH, Bernatsky S, Afif W, et al. Ustekinumab Safety in Pregnancy: A Comprehensive Review. Arthritis Care Res (Hoboken) 2023; 75:930.
  183. Lund T, Thomsen SF. Use of TNF-inhibitors and ustekinumab for psoriasis during pregnancy: A patient series. Dermatol Ther 2017; 30.
  184. Cortes X, Borrás-Blasco J, Antequera B, et al. Ustekinumab therapy for Crohn's disease during pregnancy: a case report and review of the literature. J Clin Pharm Ther 2017; 42:234.
  185. Clowse ME, Feldman SR, Isaacs JD, et al. Pregnancy Outcomes in the Tofacitinib Safety Databases for Rheumatoid Arthritis and Psoriasis. Drug Saf 2016; 39:755.
Topic 7998 Version 44.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟