配色: 字号:
2023+日本专家共识:炎症性肠病患者的疫苗接种
2023-01-17 | 阅:  转:  |  分享 
  
ORIGINAL ARTICLE—ALIMENTARY TRACT

Expert consensus on vaccination in patients with inflammatory

bowel disease in Japan

Takashi Ishige

1

?

Toshiaki Shimizu

2

?

Kenji Watanabe

3

?

Katsuhiro Arai

4

?

Koichi Kamei

5

?

Takahiro Kudo

6

?

Reiko Kunisaki

7

?

Daisuke Tokuhara

8

?

Makoto Naganuma

9

?

Tatsuki Mizuochi

10

?

Atsuko Murashima

11

?

Yuta Inoki

5

?

Naomi Iwata

12

?

Itaru Iwama

13

?

Sachi Koinuma

14

?

Hirotaka Shimizu

4

?

Keisuke Jimbo

6

?

Yugo Takaki

15

?

Shohei Takahashi

16

?

Yuki Cho

17

?

Ryusuke Nambu

13

?

Daisuke Nishida

7

?

Shin-ichiro Hagiwara

18

?

Norikatsu Hikita

17

?

Hiroki Fujikawa

4

?

Kenji Hosoi

19

?

Shuhei Hosomi

20

?

Yohei Mikami

21

?

Jun Miyoshi

22

?

Ryusuke Yagi

1

?

Yoko Yokoyama

23

?

Tadakazu Hisamatsu

22

Received: 30 July 2022 / Accepted: 28 December 2022

C211 Japanese Society of Gastroenterology 2023

Abstract Immunosuppressive therapies can affect the

immune response to or safety of vaccination in patients

with inflammatory bowel disease (IBD). The appropriate-

ness of vaccination should be assessed prior to the initia-

tion of IBD treatment because patients with IBD frequently

undergo continuous treatment with immunosuppressive

drugs. This consensus was developed to support the

decision-making process regarding appropriate vaccination

for pediatric and adult patients with IBD and physicians by

providing critical information according to the published

literature and expert consensus about vaccine-pre-

ventable diseases (VPDs) [excluding cervical cancer and

coronavirus disease 2019 (COVID-19)] in Japan. This

consensus includes 19 important clinical questions (CQs)

& Takashi Ishige

ishiget@gunma-u.ac.jp

1

Department of Pediatrics, Gunma University Graduate

School of Medicine, 3-39-22, Showa-Machi, Maebashi,

Gunma 371-8511, Japan

2

Department of Pediatrics and Adolescent Medicine, Juntendo

University Graduate School of Medicine, Tokyo, Japan

3

Division of Gastroenterology and Hepatology, Department of

Internal Medicine, Hyogo Medical University, Nishinomiya,

Japan

4

Division of Gastroenterology, Center for Pediatric

Inflammatory Bowel Disease, National Center for Child

Health and Development, Tokyo, Japan

5

Division of Nephrology and Rheumatology, National Center

for Child Health and Development, Tokyo, Japan

6

Department of Pediatrics, Juntendo University Faculty of

Medicine, Tokyo, Japan

7

Inflammatory Bowel Disease Center, Yokohama City

University Medical Center, Yokohama, Japan

8

Department of Pediatrics, Wakayama Medical University,

Wakayama, Japan

9

Department of Gastroenterology and Hepatology, Kansai

Medical University, Osaka, Japan

10

Department of Pediatrics and Child Health, Kurume

University School of Medicine, Kurume, Fukuoka, Japan

11

Center for Maternal-Fetal, Neonatal and Reproductive

Medicine, National Center of Child Health and Development,

Tokyo, Japan

12

Department of Infection and Immunology, Aichi Children’s

Health and Medical Center, Obu, Japan

13

Division of Gastroenterology and Hepatology, Saitama

Children’s Medical Center, Saitama, Japan

14

Japan Drug Information Institute in Pregnancy, National

Center of Child Health and Development, Tokyo, Japan

15

Department of Pediatrics, Japanese Red Cross Kumamoto

Hospital, Kumamoto, Japan

16

Department of Pediatrics, Kyorin University School of

Medicine, Tokyo, Japan

17

Department of Pediatrics, Osaka Metropolitan University

Graduate School of Medicine, Osaka, Japan

18

Department of Pediatric Gastroenterology, Nutrition and

Endocrinology, Osaka Women’s and Children’s Hospital,

Osaka, Japan

19

Division of Gastroenterology, Tokyo Metro Children’s

Medical Center, Tokyo, Japan

20

Department of Gastroenterology, Osaka Metropolitan

University Graduate School of Medicine, Osaka, Japan

123

J Gastroenterol

https://doi.org/10.1007/s00535-022-01953-w

on the following 4 topics: VPDs (6 CQs), live attenuated

vaccines (2 CQs), inactivated vaccines (6 CQs), and vac-

cination for pregnancy, childbirth, and breastfeeding (5

CQs). These topics and CQs were selected under unified

consensus by the members of a committee on

intractable diseases with support by a Health and Labour

Sciences Research Grant. Physicians should provide nec-

essary information on VPDs to their patients with IBD and

carefully manage these patients’ IBD if various risk factors

for the development or worsening of VPDs are present.

This consensus will facilitate informed and shared deci-

sion-making in daily IBD clinical practice.

Keywords Ulcerative colitis C1 Crohn’s disease C1

Immunization C1 Vaccine-preventable disease

Introduction

Vaccination protects vaccinated persons and those around

them who are vulnerable to disease, reducing the risk of

disease spread among family members, colleagues, and

other people in the community. The recent progress in the

treatment of inflammatory bowel disease (IBD) has

allowed more patients to receive immunosuppressive

treatments (Table 1) than in the past. Because immuno-

suppressants often affect the safety and immunogenicity of

vaccination and are often difficult to discontinue, the

necessity of vaccination should be evaluated immediately

after the diagnosis of IBD. This consensus was established

to provide critical information required for clinicians to

plan appropriate preventive care for patients with IBD. The

information in the consensus is based on a literature review

and the opinions of experts in IBD and vaccination.

Because of the lack of sufficient data, most of the previous

guidelines on vaccination of patients with IBD considered

only limited evidence of vaccine safety and effectiveness in

IBD populations. In addition, evidence from individual

countries is needed because routine vaccine schedules and

sanitation measures vary from country to country; however,

such evidence is scarce. Because close attention is essential

to avoid vaccine-related adverse events [e.g., infection by the

vaccine virus after administration of a live attenuated vac-

cine (LAV)], these consensus statements are based on

reviews of manuscripts, accumulation of evidence, and

discussion among members; however, they are not based on

assessment of the certainty of the evidence. This consensus

consists of the following sections: vaccine-preventable dis-

eases (VPDs), LAVs, inactivated vaccines, and vaccination

during pregnancy and breastfeeding. Information about

severe acute respiratory syndrome coronavirus 2 (SARS-

CoV-2) vaccination is provided elsewhere. Likewise, human

papilloma virus vaccination is not described in this consen-

sus because at the time of this writing, the recommendation

for the vaccine had been temporarily suspended until Octo-

ber 2021.

Although this consensus is based on the infectious dis-

ease status, vaccination policy, and health insurance system

in Japan, it will facilitate informed and shared decision-

making in daily IBD clinical practice.

Methods

A Health and Labour Sciences Research Grant for Research

on Intractable Diseases from the Ministry of Health, Labour,

and Welfare of Japan was obtained to support the develop-

ment of a task force for establishment of an expert consensus

on vaccination in patients with IBD. First, a steering com-

mittee comprising seven pediatric gastroenterologists, four

21

Division of Gastroenterology and Hepatology, Department of

Internal Medicine, Keio University School of Medicine,

Tokyo, Japan

22

Department of Gastroenterology and Hepatology, Kyorin

University School of Medicine, Tokyo, Japan

23

Department of Intestinal Inflammation Research, Hyogo

College of Medicine, Nishinomiya, Hyogo, Japan

Table 1 List of immunosuppressive and non-immunosuppressive

treatments defined in this consensus

Immunosuppressive agents

Corticosteroids (prednisolone, methylprednisolone, budesonide)

Azathioprine

6-Mercaptopurine

Tacrolimus

Cyclosporine

Infliximab (including biosimilar)

Adalimumab (including biosimilar)

Golimumab

Ustekinumab

Tofacitinib

Non-immunosuppressive agents

5-ASA

Sulfasalazine

Enteral nutrition formula

Granulocyte and monocyte adsorption apheresis/

leukocytapheresis

Darvadstrocel

Vedolizumab

a

5-ASA 5-aminosalicylic acid

a

Vedolizumab is not included among the immunosuppressive agents

because its effect is gut-selective, whereas specific consideration for

opportunistic infections is recommended

123

J Gastroenterol

physician gastroenterologists, and one expert in women’s

health and medication during pregnancy was formed. The

committee was charged to develop questions for a systematic

literature search. The literature review team then performed

the systematic literature search using PubMed and Ichushi

(www.jamas.or.jp). Gray literature, including alerts from the

government or statistical data from the National Institute of

Infectious Diseases, was included. The review team com-

prised 20 physicians, including pediatric gastroenterologists,

physician gastroenterologists, and women’s health experts.

The review team was divided into four sections; vaccine-

preventable diseases (VPDs), LAVs, inactivated vaccines,

and vaccination during pregnancy and breastfeeding and

made drafts of commentaries. The commentaries were dis-

cussed with the steering committee members, who were also

divided into those sections, and statements were established.

A Web meeting attended by all the committee members was

then conducted to establish a consensus among the members.

After a few corrections were made based on the discussion at

the meeting, agreement among all the committee members

was confirmed electronically. The manuscript was made

available to all the intractable diseases grant members for

commenting before submission. However, the strength of the

recommendations was not adjudicated in this consensus.

Vaccine-preventable diseases

VPDs are diseases that are preventable by vaccination.

Table 2 and Fig. 1 show the currently available vaccines and

the recommended schedule of vaccination in Japan. Infants

and people of advanced age are susceptible to VPDs and tend

to have more severe outcomes of VPDs compared with

healthy adults. Patients with IBD also have an increased risk

of the development or worsening of VPDs than do healthy

individuals; therefore, physicians should provide necessary

information on VPDs to their patients with IBD and carefully

manage these patients if various risk factors for the devel-

opment or worsening of VPDs are present (e.g., advanced

age or current treatment with immunosuppressive therapies).

Before deciding to vaccinate a patient with IBD, it is nec-

essary to obtain the patient’s vaccination record and history

of VPDs. Antibody testing should then be taken into con-

sideration, especially for hepatitis B, measles, rubella,

mumps, and varicella, although such testing is not covered

by health insurance in Japan.

Q1. Are patients with IBD at increased risk for

VPDs?

[Statement]

(1) Patients with IBD have an increased risk of

the development or worsening of VPDs

compared with healthy individuals.

(2) Particularly among patients with IBD, the

risk of VPDs is higher in advanced-age

populations; undernourished patients; patients

with comorbidities such as chronic diseases

(e.g., diabetes and kidney disease), primary

immunodeficiencies, and HIV infection; and

patients receiving immunosuppressive

therapies.

Table 2 Vaccines available in Japan

Routine vaccination

Live attenuated vaccine

Bacille Calmette-Guerin (BCG)

Measles (or measles-rubella combined)

Rubella

Varicella rotavirus

Inactivated vaccine

Diphtheria, pertussis, tetanus, and polio (DPT-IPV)

Diphtheria, pertussis, tetanus (DPT)

Polio

Diphtheria-tetanus (DT)

Japanese encephalitis

Human influenza type b (Hib)

Hepatitis B

Human papilloma virus (2- and 4-valent vaccines)

Influenza

Pneumococcus (13- and 23-valent vaccines)

COVID-19 (mRNA and viral vector vaccines)

Voluntary vaccination

Live attenuated vaccine

Mumps

Yellow fever

Shingles (live attenuated varicella)

Inactivated vaccine

Tetanus

Diphtheria

Hepatitis A

Meningococcus

Rabies

Shingles (recombinant zoster)

Human papilloma virus (9-valent vaccine)

J Gastroenterol

123

[Commentary]

Among patients with IBD, the risk of opportunistic

infections is higher in advanced-age populations;

undernourished patients; patients with comorbidities such

as chronic diseases (e.g., diabetes and kidney disease),

primary immunodeficiency, and HIV infection; and

patients receiving immunosuppressive therapy [1–3]. In

particular, immunosuppressive therapy increases the risk of

opportunistic infections, and the immunosuppressive ther-

apy-mediated risk of infection is known to be even higher

in patients receiving combination therapies (e.g., thiopuri-

nes plus steroids or thiopurines plus steroids plus inflix-

imab) [1–3]. In addition, individuals of advanced age

sometimes do not know their vaccination record in spite of

their increased risk of infection; thus, it is very important to

prevent older patients with IBD from developing VPDs of

an infectious nature.

In a systematic review, the pooled incidence of

invasive pneumococcal disease was 65/100,000 person-

years in patients with chronic inflammatory diseases

(including IBD) compared with 10/100,000 in healthy

controls [4]. In terms of Haemophilus influenzae type b

(Hib), a cohort study showed no significant difference in

the Hib-mediated mortality rate between IBD and non-

IBD groups, but hospitalizations due to Hib infection

were significantly higher in patients with than without

IBD with an adjusted odds ratio of 1.34 [95% confidence

interval (CI) 1.16–1.55] [5]. In terms of herpes zoster,

which has been preventable by recombinant herpes zoster

vaccine for populations of advanced age (C 50 years)

since January 2020 in Japan, a cohort study demon-

strated that the prevalence of herpes zoster in patients

with IBD was significantly higher than that of healthy

subjects [6, 7]. With respect to human papillomavirus

disease, a systematic review of five cohort studies and

Fig. 1 Standard vaccine schedule for children in Japan (as of October 2020). These recommendations are derived from the Japan Pediatric

Society

J Gastroenterol

123

three case–control studies clarified that patients with IBD

on immunosuppressive therapy had an increased risk of

cervical high-grade dysplasia/cancer compared with the

general population [8]. In a comparative study, the long-

term hospitalization rate due to seasonal influenza was

significantly higher in patients with than without IBD

(5.40% vs. 1.85%, respectively; P\0.001) [9]. Several

studies of hepatitis B virus (HBV) revealed a higher risk

of HBV infection in patients with IBD than in healthy

individuals [10, 11], whereas other studies showed no

significant difference in HBV infection between indi-

viduals with and without IBD [12, 13]. The Japan

Society of Hepatology guidelines for the management of

HBV infection cautions clinicians about the risk of HBV

reactivation in HBV carrier patients receiving immuno-

suppressive therapy; the guidelines thus recommend

testing patients for HBV antibody prior to immunosup-

pressive therapy [14]. There is no evidence indicating a

higher risk of diphtheria, tetanus, pertussis, or polio in

patients with IBD than in healthy populations, possibly

because of the low prevalence of these diseases or

uneven distribution of cases [1].

No studies have analyzed the risks of measles, rubella,

and mumps between patients with IBD and healthy

individuals, but one study revealed a higher risk of

severe measles infection in immunocompromised patients

than in healthy individuals [15]. In terms of varicella

zoster virus (VZV), one report revealed 5 deaths among

20 patients with IBD (16 adults and 4 children) receiving

immunosuppressive therapy [16]. In a retrospective study

of pediatric patients with IBD, the rate of hospitalization

following a primary VZV infection was higher in

patients with than without IBD [17]. A Japanese

nationwide study that collected data on episodes of

measles, rubella, varicella, and mumps in pediatric

patients receiving immunosuppressive therapy for kidney

disease, rheumatoid arthritis, gastrointestinal disease, or

post-organ transplantation showed that 4 of 47 hospital-

ized pediatric patients (43 with VZV and 4 with mumps)

receiving immunosuppressive therapy died of dissemi-

nated VZV infection [18]. This finding indicated a risk

of worsening VZV infection in patients with IBD

receiving immunosuppressive therapy. In terms of rota-

virus, no study has analyzed the risks and severity of

rotavirus infection between patients with IBD and heal-

thy individuals. However, a case–control study of 4584

patients with rotavirus infection showed that immuno-

compromised patients had a 7.4-fold higher risk of

hospitalization than the general population [19].

Q2. Which patients with IBD are at increased

risk for VPDs?

[Statement]

(1) Patients who have IBD with primary

immunodeficiencies are at increased risk for

VPDs and LAV-induced infections.

Particularly, 7–10% of pediatric patients who

are diagnosed with IBD before the age of

6 years have monogenic IBD, and most of them

have associated immunodeficiencies; therefore,

these patients should be referred to specialists

for vaccination decisions in addition to IBD

management.

(2) Advanced-age patients with IBD tend to

have impaired immune function and associated

comorbidities and complications; they are thus

at increased risk for infectious diseases. These

patients are recommended to receive necessary

vaccinations at appropriate time points.

[Commentary]

Patients who have IBD with primary immunodefi-

ciency and advanced-age patients with IBD are at high

risk for VPDs. Primary immunodeficiency is a broad

category of diseases in which immune resistance to

pathogens is impaired at birth, and more than 400 dif-

ferent types of primary immunodeficiencies have been

identified [20]. IBD caused by monogenic germline

mutations (i.e., monogenic IBD) was recently found to

be present in approximately 7–10% of patients with IBD

diagnosed before the age of 6 years, and most of these

cases of monogenic IBD were found to be comorbid

with primary immunodeficiency disease [21]. In one

systematic review, 44.7% of 750 patients with mono-

genic IBD had severe or atypical infections [22]. In

Japan, targeted genetic panel tests for certain causative

genes have been covered by national health insurance

since 2018, making genetic diagnosis possible.

Vaccination programs for patients with IBD vary

widely depending on the primary immunodeficiency

disease. For example, LAVs are contraindicated in

patients with primary immunodeficiency diseases affect-

ing cellular immunity (such as severe combined

immunodeficiency and Wiskott–Aldrich syndrome)

because of the risk of life-threatening vaccine-induced

J Gastroenterol

123

illness. However, inactivated vaccines can be adminis-

tered to patients with primary immunodeficiency,

although the antibody levels after vaccination vary

depending on the etiology of the primary immunodefi-

ciency [23]. In patients with chronic granulomatous

disease, Bacille Calmette-Gue′rin (BCG) is contraindi-

cated because BCG vaccination may lead to the devel-

opment of disseminated BCG disease, resulting in severe

outcomes [24].

Therefore, if patients with IBD are suspected to have

monogenic germline mutations or primary immunodefi-

ciency because of very early onset (\6 years of age), a

family history of IBD or primary immunodeficiency,

refractory anal lesions, or strong resistance to conventional

IBD treatment, referral of these patients to specialized

facilities for appropriate vaccination programs and sched-

ules is recommended.

Advanced-age patients with IBD are at higher risk for

VPDs than younger patients with IBD and healthy

advanced-age individuals because of their reduced

immunity and higher prevalence of comorbidities/com-

plications such as diabetes, renal impairment, and low

nutritional status [25]. Generally, individuals C 65 years

of age are at increased risk for invasive pneumococcal

infection and seasonal influenza virus, and those C 50

years of age are at increased risk for reactivation of

VZV [26–28]. Immunosuppressive therapy has been

shown to increase the risk of severe infections in indi-

viduals of advanced age. One study demonstrated that

advanced-age patients with IBD receiving infliximab and

adalimumab had a 20-fold higher incidence of severe

infections than did patients of the same age with IBD

but without infliximab and adalimumab therapy [29]. In

terms of herpes zoster as an adverse event associated

with tofacitinib therapy, tofacitinib-treated patients of

advanced age (C 65 years) with ulcerative colitis have

been shown to have higher rates of herpes zoster with an

odds ratio of 9.55 (95% CI 4.77–17.08) than tofacitinib-

treated younger patients (\65 years of age) [30]. In

particular, physicians should be aware of the risk of

VPDs, including opportunistic infections, and should take

into consideration appropriate vaccination planning in

patients with refractory IBD receiving combinations of

steroids, thiopurines, and biologic agents.

Q3. Should physicians obtain vaccination

records and the history of VPDs in patients with

IBD at the time of IBD diagnosis?

[Statement]

At the time of IBD diagnosis, physicians should

obtain the patient’s vaccination records and

history of VPDs, such as measles, rubella,

varicella (herpes zoster, especially in adults

aged ≥ 50 years), mumps, hepatitis B, and

pneumococcal infection.

[Commentary]

Patients with IBD are at risk of developing opportunistic

infections and aggravating VPDs during treatment

depending on age, nutritional status, and immunosuppres-

sive therapy [31–36]. Nevertheless, evaluation of immunity

to VPDs seems to be insufficient in patients with IBD

[37, 38].

To the extent possible, LAVs against VPDs (e.g.,

measles, rubella, varicella, and mumps) should be given

before starting immunosuppressive therapy in patients with

IBD [1]. For this purpose, it is necessary to confirm the

vaccination records and history of VPDs at the time of IBD

diagnosis. In particular, knowing the history of varicella or

herpes zoster is important to determine the risk of devel-

oping herpes zoster while receiving immunosuppressive

therapy. To prevent herpes zoster in patients of advanced

age, recombinant herpes zoster vaccine has been available

to adults aged C 50 years since 2020 in Japan. Thus, it is

also important to confirm the vaccination history of herpes

zoster vaccine in advanced-age patients with IBD.

A program for HBV as a routine vaccination in infants

has been in place since 2016 in Japan; thus, there remains a

substantial HBV-unvaccinated population at risk of sexu-

ally transmitted HBV at or after adolescence. Patients

receiving immunosuppressive therapy are at risk of HBV

reactivation and aggravation of HBV infection [39, 40],

and it is, therefore, necessary to obtain the HBV vaccina-

tion history and provide appropriate information to patients

at the time of IBD diagnosis. In addition to HBV, it is

appropriate to obtain all of the patient’s vaccination history

to fully understand the risk of aggravation of other VPDs.

J Gastroenterol

123

Q4. Should physicians perform antibody tests

to determine the patient’s immunity to VPDs?

[Statement]

(1) Hepatitis B screening is recommended for

all patients with IBD at the time of diagnosis of

IBD.

(2) In terms of VPDs (varicella, herpes zoster,

measles, rubella, and mumps) other than

hepatitis B, antibody testing should be taken

into consideration to assess immunity against

these VPDs in patients with IBD without a

history of or vaccination records for these

VPDs.

(3) Even if hepatitis B screening and antibody

testing for VPDs are not performed at the time

of IBD diagnosis, physicians should take these

screening and antibody tests into consideration

before or after the initiation of

immunosuppressive therapies.

[Commentary]

Several cohort studies worldwide have indicated that

the prevalence of HBV and hepatitis C virus (HCV) in

patients with IBD is similar to that in the general pop-

ulation [2, 41, 42]. Reactivation of HBV is a well-known

complication of immunosuppression, and an increased

incidence of liver failure due to viral reactivation has

been described in patients with IBD under immunosup-

pression [39, 40]. The European Crohn’s and Colitis

Organisation (ECCO) guideline recommends serological

HBV screening for all patients at diagnosis of IBD [2],

and Pittet et al. [43] recommended serological HBV

screening including hepatitis B surface (HBs) antigen,

anti-HBs immunoglobulin G (IgG), and anti-hepatitis B

core IgG for adults with IBD in Switzerland. Although

HCV is not a VPD, the guidelines recommend serolog-

ical screening for HCV using antibody testing in patients

with IBD at diagnosis. If possible, HCV-RNA testing

should be taken into consideration for HCV screening

[2, 44].

Patients with IBD who are undergoing immunosup-

pressive therapy and seronegative for VZV IgG are at

risk of severe varicella and thus require prompt post-

exposure prophylaxis in the event of exposure. At

diagnosis of IBD, patients should be screened for their

susceptibility to primary varicella infection by obtaining

a thorough history. We suggest anti-herpes zoster IgG

antibody testing for patients without a clear history of

varicella, herpes zoster, or receipt of varicella vaccine

[2]. We also suggest testing for antibodies to measles,

rubella, mumps, and VZV at diagnosis of IBD at least

once, although as of 2021, insurance coverage of anti-

body testing is not approved for patients other than those

who are suspected to have infection in Japan [43].

At diagnosis of IBD, or at least prior to commence-

ment of immunosuppressive therapy including vedolizu-

mab, we suggest testing for antibodies to HBV, VZV,

measles, rubella, and mumps. For seronegative patients

with IBD, we suggest administration of the course of

vaccines.

Q5. Should physicians give vaccination or

additional booster shots to patients with IBD

who have low antibody titers?

[Statement]

When the serum titers of antibody to hepatitis

B, measles, rubella, mumps, or varicella are

low, vaccination or additional booster shots to

acquire protection against these VPDs need to

be taken into consideration at the proper time

point in patients with IBD.

[Commentary]

Patients with IBD have increased risks of oppor-

tunistic infections and development or worsening of

VPDs according to their age (e.g., advanced age),

nutritional status, and immunosuppressive therapies.

Therefore, maintaining protective immunity in patients

with IBD by vaccination is critical to optimize these

patients’ outcomes [1, 2, 25, 38, 43, 45–49]. For patients

with IBD not on immunosuppressive therapy, we suggest

administration of live and inactivated vaccines. For

patients on immunosuppressive therapy, LAVs are not

recommended [1, 45]. In terms of HBV vaccine for

patients with IBD, we suggest administration of a three-

dose vaccination series (at 0, 1, and 6 months) when the

anti-HBs antibody titer declines to\10 mIU/mL. When

the anti-HBs antibody titer is still\10 mIU/mL at

1–2 months after a three-dose vaccination series in

J Gastroenterol

123

patients with IBD, we suggest administration of an

additional three-dose vaccination series. For patients with

an immune response to HBV vaccine (anti-HBs antibody

titer of C 10 mIU/mL), we suggest no further antibody

testing and no booster shots [14, 50]. The test sensitivity

and the cut-offs for protective immunity to measles,

rubella, and VZV vary among the techniques used, such

as enzyme immunoassays [enzyme immunoassay (EIA)

and enzyme-linked immunosorbent assay (ELISA)],

hemagglutination inhibition assays, particle agglutination

assays, and neutralization tests [50]. Based on an

understanding of these problems, we suggest using the

antibody titers necessary to consider the need for vac-

cinations that are documented in several domestic

guidelines for vaccination of patients with IBD. For

example, based on a guideline for vaccination from the

Japan Society for Hematopoietic Cell Transplantation,

the ELISA IgG antibody titers necessary to consider the

need for vaccinations against measles, rubella, and

varicella are B 4.0, B 5.0, and B 5.0 IU/mL, respec-

tively [51]. In another study and guideline, the EIA IgG

antibody titer necessary to consider the need for vacci-

nations against measles, rubella, mumps, and varicella

was B 4.0 for all [52]. The United States Centers for

Disease Control and Prevention (CDC) stated that the

sensitivity of ELISA to detect antibody to VZV varies

according to the antigens used and that ELISA is,

therefore, not accurate enough for evaluation of immu-

nity against VZV. The CDC recommends purified gly-

coprotein ELISA as a more sensitive method; however,

testing with purified glycoprotein ELISA is not com-

mercially available. Thus, it is necessary to appropriately

interpret the examined VZV antibody titers based on an

understanding of these problems raised by commercial

assays.

Although the sensitivities and cut-off points of anti-

body titers vary according to the testing methods, we

suggest administration of vaccination or booster shots to

patients with IBD when the ELISA (or EIA) IgG anti-

body titers are\4–5 against measles, rubella, mumps,

and varicella. In terms of the Hib vaccine, herpes zoster

vaccine (recombinant vaccine), influenza vaccine, Strep-

tococcus pneumoniae vaccine, meningococcal vaccine,

human papillomavirus vaccine, and diphtheria, tetanus,

and pertussis vaccine combined with inactivated polio-

virus (DTaP-IPV) vaccine, we suggest that vaccination is

based not on the antibody titers but instead on the pre-

vious vaccination records [45, 49, 51].

Q6. Should physicians perform antibody testing

after vaccination in patients with IBD?

[Statement]

(1) In addition to humoral immunity, cellular

and mucosal immunity play crucial roles in

protection against VPDs. Therefore, serum

antibody titers cannot explain all aspects of

protective immunity, and antibody testing has

resultant limitations in the assessment of

vaccine-mediated immunity.

(2) Anti-HBs antibody testing is useful for

assessing hepatitis B vaccine-mediated

immunity and making decisions regarding

additional booster shots.

(3) Antibody tests for measles, rubella, mumps,

and VZV are useful for decision-making

regarding vaccination, but these tests are

limited in the assessment of vaccine-mediated

immunity.

[Commentary]

Not only humoral immunity with antibodies but also

physical barriers (e.g., mucus and epithelial cells) at the

site of infection of pathogenic microorganisms and various

immune cells involved in mucosal immunity are important

in the prevention of infection. In other words, it is difficult

to predict the severity of the infection or whether it can be

prevented based only on antibody titers [53], therefore;

evaluation of the immune status after vaccination should be

judged comprehensively based on each patient’s age,

nutritional status, and administration of drugs such as

immunosuppressive agents. Measuring antibody titers after

vaccination or during the course of IBD treatment is rela-

tively easy and objective and can be used to make decisions

on additional vaccination. Notably, however, the sensitivity

of antibody measurement and the optimal cutoff value of

the antibody for prevention of infection vary depending on

the measurement technique.

Regarding HBV, the Canadian Association of Gas-

troenterology and the ECCO recommend that all patients

with IBD receive a three-dose series of HBV vaccinations

and that the anti-HBs IgG antibody titer be measured

4–12 weeks after the third vaccination to achieve an anti-

J Gastroenterol

123

HBs antibody level of[10 IU/L [2, 45]. HBV vaccination

of all infants has been routine in Japan since 2016 to pre-

vent horizontal transmission, but people born before that

date (excluding those born to HBs antigen-positive moth-

ers) are unlikely to have HBV immunity. It is thus

important to confirm patients’ birth year and vaccination

records. The vaccination guideline for healthcare workers

issued by the Japanese Society for Infection Prevention and

Control also recommends confirmation of the anti-HBs IgG

antibody titer after a three-dose series of HBV vaccination

and, if necessary, consideration of an additional booster to

achieve an anti-HBs antibody level of C 10 IU/L [50].

Regarding measles, rubella, and mumps, the CDC does

not recommend serologic testing pre- and post-vaccination

because of the low sensitivity of such testing to detect

antibodies [54]. There is reportedly no difference in anti-

body levels after vaccination for measles, rubella, and

mumps viruses between patients with IBD on immuno-

suppressive therapy and healthy controls [55]. Conse-

quently, measuring antibody titers after vaccination in

these patients might be useful to determine primary vaccine

failure, in which a person does not develop protective

immunity after vaccination, or secondary vaccine failure,

in which a person loses an initially acquired immunity [56].

However, the usefulness of routine antibody titer mea-

surement is limited because the rate of antibody acquisition

after vaccination is generally very high.

With regards to VZV, the CDC does not recommend

pre- and post-vaccination serologic testing because of the

low sensitivity of antibody detection after vaccination [57].

Because no correlations between the viral antibody titer

and the risk of developing herpes zoster or the severity of

herpes zoster/postherpetic neuralgia have been reported

[58, 59], the usefulness of antibody titer measurement after

vaccination is low.

Live attenuated vaccines

LAVs are made of pathogenic viruses or bacteria with

reduced toxicity. Because they contain pathogenic organ-

isms, the risk of infection due to the vaccine strain cannot

be eliminated. As a rule, therefore, immunocompromised

or immunosuppressed patients and pregnant woman should

not receive LAVs because of safety concerns.

In this section, immunosuppressive treatment that

requires attention with respect to the concomitant use of

LAVs will be explored. Furthermore, the recommended

duration between the last LAV and introduction of

immunosuppressive treatment as well as the duration

between the cessation of immunosuppressive treatment and

LAV administration will be discussed.

LAVs for pregnant women will be discussed in another

section. Yellow fever vaccine, which is administered to

individuals who travel to endemic areas, will not be dis-

cussed; its risks and benefits should be considered in the

clinical setting as needed.

Although live varicella vaccine is approved for the

prevention of herpes zoster in people aged C 50 years, the

recently approved inactivated herpes zoster vaccine can be

used when an LAV is contraindicated.

Vaccinating family members and surrounding individ-

uals could decrease the risk of infection in patients who

should not receive LAVs.

Q7. Can patients on immunosuppressive

treatment receive LAVs?

[Statement]

In principle, LAVs should not be administered

to patients receiving immunosuppressive

treatment.

[Commentary]

Effectiveness of LAVs in patients receiving immuno-

suppressive treatment

Clinical data regarding the efficacy of LAVs in patients

receiving immunosuppressive treatment are limited [1, 2].

However, several prospective interventional studies have

focused on the resultant antibody elevation after LAV

administration in patients who have undergone organ

transplantation and in those receiving immunosuppressive

treatments, including biologics.

In a Japanese study, the seroconversion rate for measles,

rubella, varicella, and mumps vaccines was 80.0%, 100%,

59.1%, and 62.9%, respectively, in children and young

adults receiving immunosuppressants for IBD or other

conditions if their immunological condition met the criteria

of a CD4 cell count of C 500/mm

3

, stimulation index of

phytohemagglutinin-induced lymphocyte proliferation of

C 101.6, and serum IgG level of C 300 mg/dL. Among 32

patients, only one 5-year-old boy with Crohn’s disease

receiving azathioprine developed a breakthrough varicella

infection [52]. Another study revealed the probable effi-

cacy of live varicella vaccines for adults with IBD

receiving azathioprine [60].

A large prospective randomized controlled study

(VERVE trial) of the efficacy of live varicella vaccine in

patients receiving anti-tumor necrosis factor (anti-TNF)

agents is ongoing in the United States.

J Gastroenterol

123

Accumulation of further evidence for the efficacy of

LAVs in patients receiving immunosuppressive treatment

is expected.

Safety of LAVs in patients receiving immunosuppres-

sive treatment

Administration of LAVs to patients with IBD receiv-

ing immunosuppressive treatment in Japan should follow

the package insert of each drug (Table 3) as well as the

‘‘Immunization guideline for children post-organ trans-

plantation and under immunosuppressive treatment

2014’’ [61]. Thus, in principle, LAVs are not recom-

mended for patients receiving immunosuppressive treat-

ments other than vedolizumab. However, clinical

research on the use of LAVs in patients receiving

Table 3 Description of vaccination in the package insert for the treatment of inflammatory bowel disease

Trade name Generic name Prescribing information

Steroids

PREDONINE Prednisolone

Prednisolone

Sodium succinate

Important precautions: do not administer live vaccines to patients receiving long-term or high-

dose steroids or within 6 months after discontinuation

PREDONEMA

Enema

Prednisolone

Sodium phosphate

STERONEMA Betamethasone

Sodium phosphate

RINDERON

suppositories

Betamethasone

Solu-medrol for

intravenous use

Methylprednisolone

Sodium succinate

Contraindications: do not administer live vaccines or attenuated live vaccines to patients

receiving immunosuppressive therapy of this medicine

RECTABUL rectal

foam

Budesonide Important precautions: when administering a live vaccine to a patient receiving this medicine,

the patient’s immunological condition should be examined, and extreme caution should be

exercised

Zentacoat Budesonide None stated

Immunomodulators

IMURAN/AZANIN Azathioprine Contraindications for co-administration: there is a risk of breakthrough infection in

immunosuppressed patients who receive live vaccines. The live vaccine may increase and

become pathogenic if administered to

LEUKERIN Mercaptopurine

Hydrate

Immunosuppressants

Prograf Tacrolimus Hydrate Contraindications for co-administration: do not inoculate live vaccines

Sandimmun Ciclosporin

METHOTREXATE Methotrexate Important precautions: do not inoculate live vaccines while receiving this medicine

Biological products

REMICADE Infliximab Important precautions: do not inoculate live vaccines while receiving this medicine

Pregnant women: caution should be exercised when live vaccines are administered to infants

born to patients who have received this medicine

HUMIRA Adalimumab Important precautions: do not inoculate live vaccines

Pregnant women: caution should be exercised when live vaccines are administered to infants

born to patients who have received this medicine

Simponi Golimumab Important precautions: do not inoculate live vaccines while receiving this medicine

Pregnant women: caution should be exercised when live vaccines are administered to infants

born to patients who have received this medicine

Stelara Ustekinumab Important precautions: do not inoculate live vaccines while receiving this medicine

ENTYVIO Vedolizumab Precautions for co-administration: if symptoms based on the live vaccine appear after

vaccination, appropriate measures should be taken. The possibility of infection caused by live

vaccines cannot be ruled out

XELJANZ Tofacitinib citrate Important precautions: do not inoculate live vaccines while receiving this medicine

J Gastroenterol

123

immunosuppressive treatment has been conducted in

several countries, including Japan, and a consensus and

social system to support the use of LAVs in patients

who may benefit from them are needed.

In principle, LAVs are not recommended for patients

with immunosuppressive conditions, particularly cellular

immune deficiency, because of the risk of lethal viral

infection by the vaccine strain. Therefore, LAVs are

contraindicated in patients undergoing immunosuppres-

sive treatment. Some lethal events after administration of

LAVs, including yellow fever and BCG vaccines, have

been reported in this population [62, 63].

Although a few reports have addressed the safety of

LAVs for patients with IBD receiving immunosuppres-

sants, the safety of measles, rubella, varicella, and

mumps was discussed in a large systematic review and

other reports [52, 60, 64–66]. The BCG and rotavirus

vaccines are supposed to be given before 1 year of age.

Patients with infantile-onset IBD receiving immunosup-

pressive treatment and infants born of mothers receiving

immunosuppressive treatment require special attention.

Primary immunodeficiency should be ruled out in

patients with infantile-onset IBD, and in principle, LAVs

should not be administered in this high-risk population

[61]. The ‘‘Pregnancy and delivery’’ section addresses

the care of infants born of mothers receiving immuno-

suppressive treatment. No reports have described the

administration of BCG and rotavirus vaccine to infants

with IBD receiving immunosuppressive treatment.

Patients with IBD are thought to be at high risk of VPDs

[67, 68]. Patients with very-early-onset IBD under

immunosuppressive treatment are at particularly high risk

if they miss or receive an insufficient number of immu-

nizations. They may not receive contraindicated vaccines,

such as LAVs.

In general, patients receiving immunosuppressive ther-

apy are at high risk of serious infection [31], and preven-

tion of VPDs should be emphasized; however, LAVs

cannot be administered if the instructions in the package

inserts are followed. To change this situation, an ongoing

study is evaluating the safety and efficacy of LAVs in

patients under immunosuppressive treatment [52].

Although the acceptable immunological condition for safe

LAV administration in patients under immunosuppressive

treatment requires further evaluation, studies have sug-

gested that LAVs can be safely administered if these

patients’ cellular and humoral immunological parameters

are within normal levels.

Q8-1. Are LAVs prioritized over treatment of

underlying diseases?

Q8-2. What is the recommended duration

between administration of LAVs and

introduction of immunosuppressive treatment?

Q8-3. What is the recommended duration

between cessation of immunosuppressive

treatment and administration of LAVs?

[Statement]

(A8-1) Disease control should be prioritized

over LAV administration.

(A8-2) Immunosuppressive treatment should

be initiated at least 3 weeks after LAV

administration.

(A8-3) LAVs should be administered at least

3 months after the cessation of

immunosuppressive treatment.

[Commentary]

LAVs for patients without prior administration and

without detectable antibody should be discussed on an

individual-patient basis considering the disease activity and

importance of vaccination. Ideally, LAVs should be

administered before the introduction of immunosuppres-

sive treatment [2, 61]. However, some patients with IBD

require immunosuppressive treatment before completion of

LAVs. Disease control should be prioritized in those

requiring early immunosuppressive treatment, and admin-

istration of LAVs should be considered once immunosup-

pressive treatments are discontinued in the disease course

[1, 69]. LAVs should be administered once disease

remission has been induced without immunosuppressants.

Immunosuppressants should be initiated at least

1 month after LAV administration according to the ECCO

guideline and at least 3 weeks after LAV administration

according to the ‘‘Immunization guideline for children

undergoing organ transplantation and immunocompro-

mised condition’’ (Fig. 2)[2].

For patients under immunosuppressive treatment, LAVs

can be administered at least 3 months after immunosup-

pressive treatment is discontinued as written in the

domestic guideline (Fig. 2)[61]. Those taking high-dose

corticosteroids should not receive LAVs for 6 months after

the discontinuation of steroids according to the package

J Gastroenterol

123

inserts. The ECCO guideline recommends a 1-month

duration between the cessation of immunosuppressive

treatments (corticosteroids, methotrexate, tofacitinib,

cyclosporine, and tacrolimus) and LAV administration

based on the half-life of these immunosuppressants. The

package insert of each LAV does not clearly indicate the

duration between cessation of immunosuppressants and

LAV administration.

Although infection by the vaccine virus of an LAV

could occur in patients under immunosuppressive treat-

ment, there are few reports of lethal or serious infection

except for patients receiving BCG [63]. Therefore, the risks

and benefits of LAV administration in high-risk patients

should be considered on an individual-patient basis.

Inactivated vaccines

Inactivated vaccines are made by isolating and purifying

disease-causing pathogens, such as bacteria and viruses,

and inactivating them with formalin, phenol, heat treat-

ment, or ultraviolet irradiation to eliminate their infectivity

and pathogenicity without affecting their protective anti-

gens. Therefore, serious adverse reactions are unlikely to

occur. However, compared with LAVs, immunity is more

difficult to acquire and is shorter-lasting. Therefore, repe-

ated vaccinations are required to maintain immunity, fol-

lowed by additional immunizations after a certain period of

time. A toxoid is a vaccine in which bacterial toxins are

treated with formalin or other agents to eliminate toxicity.

These vaccines require repeated inoculations similar to

inactivated vaccines. Table 2 shows a list of inactivated

vaccines and toxoids that can be provided in Japan.

In daily medical practice, inactivated vaccines and

toxoids are often collectively referred to as inactivated

vaccines; thus, the term ‘‘inactivated vaccines’’ is used in

this section to refer to both types.

Inactivated vaccines for patients with IBD are consid-

ered effective except in cases of severe disease or use of

high-dose steroids. Furthermore, inactivated vaccines can

be administered during immunosuppressive therapy.

Notably, the possibility of a low antibody acquisition rate

should be considered.

This section describes the relevance of inactivated

vaccines to IBD.

Q9. Is inactivated vaccination recommended

for patients with IBD on immunosuppressive

therapy?

[Statement]

(1) Inactivated vaccination is considered

effective even during immunosuppressive

therapy. However, it is necessary to consider

that the antibody acquisition rate may decrease

during immunosuppressive therapy.

(2) There have been no reports of serious

adverse reactions such as infections associated

with vaccination, and the vaccine can be safely

administered according to the normal schedule.

[Commentary]

Several reports have described inactivated vaccination

of patients with IBD on immunosuppressive therapy,

including reviews [70, 71]. Large numbers of patients have

acquired antibodies, and the safety of this practice has been

deemed adequate.

Fig. 2 Duration between

immunosuppressive treatment

and live attenuated vaccine

administration [61]

J Gastroenterol

123

Influenza virus vaccines can reportedly reduce the rate

of immune acquisition in patients treated with thiopurines

or anti-TNF agents [72, 73].

In patients receiving the hepatitis B vaccine,

immunomodulatory drugs such as azathioprine and

methotrexate did not decrease the antibody acquisition rate;

the acquisition rate reportedly decreased only in patients

with IBD using anti-TNF agents [74]. In addition, one

study showed that older age is associated with a lower

antibody acquisition rate [75].

Patients with IBD have a high incidence of cervical and

oral cancer, and patients with anal lesions are at high risk of

human papillomavirus-related anal cancer. Therefore, anti-

body acquisition by human papillomavirus vaccination is

important. In a report of the human papillomavirus tetrava-

lent vaccine in 33 pediatric patients with IBD on immuno-

suppressive therapy, 100% of the patients acquired

antibodies to types 6, 11, and 16 and 96% acquired antibodies

to type 18 [76]. These rates were comparable to the antibody

acquisition rate in healthy women, and there were no serious

adverse reactions related to vaccination [76].

In one study, the median seroprotection rate against the

pneumococcal 13-valent diphtheria conjugate vaccine

(PCV13) significantly increased from 43.9% at inclusion to

90.4% (P\0.001) after vaccination. Patients receiving

anti-TNF agents achieved a slightly lower seroprotection

rate (from 44.5 to 86.6%) than patients treated with other

types of immunosuppressive therapy [77]. Furthermore,

patients administered infliximab or combination immuno-

suppressive therapy had significantly lower response rates

against the 23-valent pneumococcal polysaccharide vac-

cine (PPSV23) (57.6% and 62.5%, respectively) compared

with the group on mesalamine (88.6%; P\0.05 for both

comparisons) [78]. Additionally, both PCV13 and PSSV23

were generally safe and well tolerated.

Regarding the safety of the vaccine, a systematic review

of pediatric patients with IBD revealed no infections

caused by the vaccine strain, indicating that the vaccine can

be safely administered [71].

Inactivated vaccines can be safely administered to

patients with IBD on immunosuppressive therapy according

to the normal schedule, with most patients acquiring

antibody.

Q10. In what situations should inactivated

vaccination be avoided?

[Statement]

There are no conditions or situations in which

inactivated vaccination should be obviously

avoided in patients with IBD. Inactivated

vaccination is considered to be beneficial

regardless of the patient’s immunosuppressive

status, but the clinician should consider that the

rate of antibody production may be reduced

during immunosuppressive therapy.

[Commentary]

Inactivated vaccines can be administered to patients

with IBD who are not receiving immunosuppressive ther-

apy, with the same safety considerations as for healthy

patients. However, a systematic review of pediatric patients

with IBD showed that even those on immunosuppressive

therapy can be safely vaccinated without developing

infections caused by vaccine strains [71]. The same results

were reported in a systematic review of pediatric rheumatic

disorders, not IBD [79]. Immunosuppressive therapy cau-

ses a decrease in both cellular (T-cell) and humoral (B-cell)

immunity. In the latter case, both live and inactivated

vaccines are available. In the former case, LAVs are con-

traindicated in principle, and inactivated vaccines may

cause a reduction in antibody production because the

helper T cells may not be able to reproduce the antibody-

producing function of B cells.

A decrease in the immune response due to immuno-

suppressive therapy can reportedly occur when pred-

nisolone is administered at a dose of C 2 mg/kg/day

(C 20 mg/day for 10 kg) for longer than 14 days at a body

weight of\10 kg or when thiopurines or biologic agents

are used [80]. However, the same judgment cannot be

made to every patients because the mechanism of action

and concomitant conditions of the drugs used, immuno-

logical characteristics of individual patients, nutritional

status, and other factors also affect the results.

J Gastroenterol

123

Several reports have described a reduction in inactivated

vaccine-acquired antibody titers during immunosuppres-

sive therapy. In a study of influenza virus vaccines, the

seroconversion rate was lower in pediatric patients with

IBD treated with thiopurines and infliximab than in healthy

subjects [81].

Conversely, a retrospective cohort study of the vaccine

in pediatric patients and a randomized controlled study in

adult patients showed that a sufficient antibody production

rate was obtained even during immunosuppressive therapy

in pediatric patients [82], whereas a sufficient antibody

production rate was observed in adult patients other than

those using anti-TNF agents [73]. In addition, research has

suggested that the efficacy of HBV and pneumococcal

vaccines may be attenuated during the acute phase of the

disease and during immunosuppressive therapy [83].

Q11. Should the influenza vaccine be provided

every year? Should the vaccine be given twice?

[Statement]

High-risk individuals, who are prone to severe

influenza infection, are eligible for regular

influenza vaccination based on the

Immunization Law. In principle, a single

influenza vaccination is sufficient for patients

with IBD aged ≥ 13 years.

[Commentary]

Influenza is an acute febrile infectious disease caused by

the influenza virus. The symptoms are often associated

with high fever, regardless of the vaccination status; the

severity of the disease is often mild to moderate; and the

prognosis is generally relatively good. The disease often

resolves spontaneously with a fatality rate of\0.1%.

Although the effectiveness of the vaccine varies among

seasons, vaccination is recommended by the Japanese

Association for Infectious Diseases for patients at high risk

of complications from influenza (Table 4). In patients with

IBD receiving immunosuppressive therapy, active vacci-

nation is recommended because such patients are consid-

ered to be at high risk of complications.

Guidelines from the Japanese Association for Infectious

Diseases and from the United States recommend that

influenza vaccination be administered by the end of

October [84]. The prevalent influenza strains vary each

year. In addition, the vaccine’s effectiveness in prevention

of the disease is strongest after vaccination and decreases

by approximately 8–9% each subsequent month. Therefore,

annual influenza vaccination is recommended.

In individuals aged C 65 years, the benefit of the vac-

cine is decreased at 30 days post-vaccination. One study

showed that the rate of reduced efficacy in patients of this

age was 10.8% (95% CI 2.6–23.8%) for influenza A

(H3N2), 9.6% (95% CI - 3.3 to 32.7%) for influenza A

(H1N1), and 10.8% (95% CI 1.4–33.9%) for influenza

B/Yamagata. Notably, antibody titers are more likely to

decrease in patients of advanced age because of the slightly

faster rate of reduction than in the overall population of

patients aged C 18 years [84]. Not only patients of

advanced age but also pregnant women, children under

5 years of age, and patients with underlying medical con-

ditions should be cautious because of the possibility of

influenza complications [85].

A randomized comparative study of the influenza vac-

cine in adults with IBD undergoing anti-TNF therapy or

thiopurine therapy was conducted by dividing the patients

into two groups: those who received a single dose and

those who received an additional dose. There was no dif-

ference in the immune response after additional vaccina-

tion with the influenza vaccine, and no additional

vaccination was required [86].

The Japanese Association for Infectious Diseases rec-

ommends a single dose of influenza vaccine for people

aged C 13 years and two doses for children

aged B 12 years.

Table 4 Patients at high risk of complications during influenza

infection

6-Month-old or more and under 5-year-old

65 Years of age or older

Chronic respiratory diseases (bronchial asthma, COPD, etc.)

Cardiovascular disease (excluding hypertension alone)

Chronic kidney, liver, blood, metabolic diseases (diabetes, etc.)

Neuromuscular disease (including motor paralysis, convulsions,

dysphagia)

Immunosuppressive state (including those caused by HIV and

drugs)

Pregnancy

Residents of long-term medical treatment facilities

Significant obesity

Patients receiving long-term aspirin

Cancer-bearing patients

COPD chronic obstructive pulmonary disease, HIV human immun-

odeficiency virus

J Gastroenterol

123

Q12. Is the inactivated herpes zoster vaccine

effective in patients with IBD?

[Statement]

The inactivated herpes zoster vaccine is

approved for prevention of herpes zoster in

individuals aged > 50 years. However, data

regarding vaccination in patients with IBD and

receiving immunosuppressive therapy are

insufficient.

[Commentary]

Herpes zoster is a skin disease caused by VZV reacti-

vation. It is characterized by erythema and neuralgia that

usually develop eccentrically. Postherpetic neuralgia is

characterized by hyperalgesia and allodynia that persist

even after herpes zoster has resolved, and many cases of

intractable chronic pain have been recorded. Consequently,

patients’ quality of life markedly decreases.

The only way to prevent herpes zoster and postherpetic

neuralgia is inoculation with the vaccine [87–89]. In Japan,

in addition to the dry attenuated live varicella vaccine,

recombinant herpes zoster vaccine (an inactivated vaccine)

can be used in adults aged[50 years. This inactivated

vaccine has a preventive effect of C 90% against herpes

zoster and postherpetic neuralgia with two doses at an

interval of 2–6 months, and[85% of cases can be pre-

vented for 4 years after vaccination [90, 91]. Therefore, the

inactivated herpes zoster vaccine is recommended to prevent

complications associated with herpes zoster in immunized

adults aged[50 years regardless of their history of live

vaccination or herpes zoster occurrence. This inactivated

vaccine is also preferable to the LAVs for the prevention of

herpes zoster and related complications.

Notably, the concomitant use of LAVs with steroids and

thiopurines is contraindicated, and LAVs are not recom-

mended for patients treated with biologics that affect the

immune system. Instead, the inactivated herpes zoster vac-

cine is prescribed for these patients. The inactivated vaccine

has been approved for adults aged[50 years, but its effi-

cacy is not fully confirmed in patients with IBD, especially

those receiving immunosuppressive therapy [92]. Because

inoculation with the inactivated herpes zoster vaccine is paid

for by the patients themselves, it will be economically

burdensome.

Q13. Should the pneumococcal vaccine be

provided to advanced-age patients with IBD?

[Statement]

Pneumococcal vaccination is recommended for

aged patients, particularly those

aged > 65 years.

[Commentary]

In 2019 in Japan, the ‘‘Concept of Streptococcus pneu-

moniae vaccine for adults aged 65 and over’’ (3rd edition)

was presented by the joint committee of the Japanese Res-

piratory Society and the Japanese Society of Infectious

Diseases. In March 2021, the ‘‘Concept of pneumococcal

vaccination for high-risk persons aged 6 to 64’’ was pre-

sented by the joint committee of the Japanese Respiratory

Society, the Japanese Society of Infectious Diseases, and the

Japanese Society of Vaccination.

Pneumococcal vaccines include PPSV23 and PCV13.

PPSV23 is a routine vaccine for adults aged C 65 years.

Currently, PCV13 inoculation followed by a series of

PPSV23 inoculation (regular or voluntary inoculation) can

be administered. In Japan, the effect of the vaccine within

5 years after PPSV23 inoculation is 27.4% for all types of

pneumococcal pneumonia and 33.5% for serum-type pneu-

mococcal pneumonia [93]. According to a report targeting

invasive pneumococcal disease in patients aged C 15 years

in Japan, the effect of vaccination against invasive pneu-

mococcal disease caused by the serotypes included in the

PPSV23 vaccine is 45%. Age-stratified analysis revealed

that the efficacy of the PPSV23 vaccine serotype is 75% in

individuals aged 15–64 years and 39% in individuals

aged[65 years. Based on this evidence, an improvement in

the PPSV23 inoculation rate in adults aged C 65 years has

been expected; nevertheless, vaccination for individuals

with underlying diseases has not been investigated. Addi-

tionally, pneumococcal vaccines for high-risk persons (such

as patients with rheumatoid arthritis and collagenous dis-

eases) aged 6–64 years have been presented by the joint

committee, indicating the need to recognize attenuation of

immunogenicity of the pneumococcal vaccine in patients

undergoing thiopurine therapy.

Although clear evidence on whether IBD increases the

risk of pneumococcal infection has not been provided [45],

the pneumococcal vaccine is recommended for adult

patients with IBD who are considered to be at a high risk of

pneumococcal infection or who are receiving immuno-

suppressive therapy. This vaccine should also be adminis-

tered before immunosuppressants are given. However,

studies have yet to verify whether pneumococcal vaccina-

tion is recommended for patients with IBD if

J Gastroenterol

123

immunosuppressive treatment is not performed or for

patients with fewer risk factors for pneumococcal infection.

Data are also insufficient to propose the most appropriate

type of vaccine and administration schedule.

Age is considered a risk factor for pneumococcal

infection; thus, pneumococcal vaccination is beneficial for

older patients with IBD, especially those aged C 65 years,

unless they have contraindications for vaccination.

Q14. When inactivated vaccines are provided to

patients with IBD being treated with anti-TNF

agents, should the timing of anti-TNF therapy

and inactivated vaccination be considered?

[Statement]

Inactivated vaccines can be administered on the

same day of anti-TNF therapy.

[Commentary]

Several studies have revealed the efficacy of inactivated

vaccination for patients who have IBD, rheumatoid

arthritis, and other collagenous diseases receiving anti-TNF

agents. Although the antibody acquisition rate of the vac-

cine is slightly reduced in patients using anti-TNF agents

[81, 94], sufficient antibody titers have been obtained, and

inoculation can be safely conducted. These findings show

that the benefits of inactivated vaccination outweigh its

disadvantages. Therefore, inactivated vaccines can be

administered to patients with IBD even when they are

using anti-TNF agents.

Few studies have described the appropriate timing of

inactivated vaccination after anti-TNF agents are given. In a

study of patients with rheumatoid arthritis and ankylosing

spondylitis being treated with infliximab, the antibody

acquisition rates were compared between patients inoculated

with influenza vaccine on the day of infliximab administra-

tion (n = 22) and patients inoculated 3 weeks after inflix-

imab administration (n = 16). The results indicated no

difference in the antibody acquisition rates between the two

groups [95]. Interestingly, in patients with rheumatoid

arthritis, a higher immune response to vaccination against

the H1N1 strain was achieved in the group inoculated on the

day of infliximab administration than in the group inoculated

3 weeks after infliximab administration.

A randomized controlled trial of patients with IBD was

conducted to compare patients inoculated on the day of

infliximab administration (n = 69) with those inoculated

between two doses of infliximab administration (n = 68).

All patients received influenza vaccines [A/California/7/

2009 (H1N1), A/Victoria/361/2011 (H3N2), B/Wisconsin/

1/2010] while undergoing infliximab maintenance therapy.

The proportions of patients with sufficient antibody titers to

prevent influenza infection (hemagglutination inhibition

antibody titer of C 1:40) were 67% versus 66% (P = 0.8)

for H1N1, 43% versus 49% (P = 0.5) for H3N2, and 69%

versus 79% (P = 0.2) for B/Wisconsin/1/2010, respec-

tively. In addition, no significant differences in the exac-

erbation of the current disease or in the incidence of

adverse reactions were observed between the two groups,

and no serious adverse events were observed in either

group [82].

Research on influenza vaccines for patients taking

infliximab is limited. However, some studies have indi-

cated that there is no evidence showing that the schedule of

inactivated vaccination should be staggered in patients

receiving anti-TNF agents. Therefore, inactivated vaccines

can be administered on the same day as anti-TNF agents.

Pregnancy, lactation, and vaccination

Special consideration is required for pregnant and lactating

patients with IBD. Vaccination and infection control are

key issues during these periods. Because VPDs often affect

pregnancy outcomes, vaccination should be appropriately

received before pregnancy if possible. If a patient diag-

nosed with IBD wishes to become pregnant, vaccination

should be administered at the time of IBD diagnosis.

The benefits and risks of vaccination during pregnancy

must be carefully explained to patients. Inactivated vacci-

nes are considered safe during pregnancy. For patients who

are worried about vaccines, accurate information including

the incidence of congenital malformations or spontaneous

abortion among typical births should be provided. The

indication for vaccines for infants born to mothers with

IBD should be considered based on the duration, types, and

placental transfer of immunosuppressive agents used dur-

ing pregnancy.

J Gastroenterol

123

Q15. Which vaccines are recommended for

women with IBD who wish to become

pregnant?

[Statement]

(1) To avoid congenital rubella syndrome,

patients with a low rubella-specific antibody

titer are encouraged to be vaccinated for rubella

before pregnancy.

(2) Varicella and measles vaccination are

recommended before pregnancy if the patient is

confirmed to have no history of vaccination or

infection.

[Commentary]

A rubella epidemic occurred in Japan from 2012 to

2013, resulting in a high incidence of congenital rubella

syndrome. The cause of this epidemic is considered to be

the high rate of lack of sensitization to rubella among men

in their 30 s and 40 s and young pregnant women at that

time. A rubella epidemic and an outbreak of congenital

rubella syndrome were also reported from 2018 to 2019

[96]. Infection with rubella in the first trimester of preg-

nancy can cause congenital rubella syndrome, potentially

resulting in cataracts, glaucoma, congenital heart disease,

and sensorineural hearing loss. Congenital rubella syn-

drome due to reinfection also may occur even if the mother

has been previously vaccinated, although this is rare. In

Japan, rubella vaccination is recommended to prevent the

development of congenital rubella syndrome for women

who wish to become pregnant if their rubella-specific

antibody titer (hemagglutination inhibition method)

is B 1:16.

Because of the risk of severe disease if varicella or

measles is contracted during pregnancy, women with IBD

who have no history or vaccination of these diseases and

wish to become pregnant should be considered for these

vaccines. Because rubella vaccine, measles vaccine,

measles-rubella vaccine, and varicella vaccine are LAVs,

other sections of this document should be consulted to

determine the vaccination plan for patients who are

receiving and/or planning scheduled immunosuppressive

treatment.

Because these vaccines are contraindicated during

pregnancy, patients should be instructed to use contracep-

tion for 2 months after vaccination. If rubella or measles

vaccines are administered to a patient who is unaware that

she is pregnant or when pregnancy occurs within 2 months

of vaccination, the CDC and the Japan Society of Obstet-

rics and Gynecology guidelines indicate that there is no

need to interrupt the pregnancy because no clinically sig-

nificant risk to the fetus has been demonstrated in previous

research [97].

Voluntary rubella vaccination is recommended for

partners of pregnant women, their children, and their other

family members living together who have no history of

rubella or vaccination. To prevent rubella transmission to

the female partner, rubella vaccination is recommended for

men with IBD who have no history or vaccination of

rubella.

Q16. Can pregnant women with IBD be

vaccinated?

[Statement]

Inactivated vaccines can be used for pregnant

women.

[Commentary]

In principle, inoculation of pregnant women using LAVs

should be avoided because of concerns about the possi-

bility of transfer of the vaccine component virus to the

fetus, resulting in infection (see Q15). Vaccines other than

LAVs (e.g., inactivated vaccines) do not cause infection in

the fetus and can be administered as necessary.

Patients with IBD are considered at risk for severe

influenza infection, and annual influenza vaccination is

recommended in both Europe and the United States [2, 80].

Increased risks of the following have been reported in

pregnant women who contract influenza: hospitalization,

maternal mortality, spontaneous abortion, preterm birth,

low birth weight, small for gestational age, and fetal death

[98–101]. The influenza vaccine used in Japan is an inac-

tivated vaccine, which theoretically poses no risk to preg-

nant women or fetuses. Many studies have revealed the

safety of influenza vaccination during pregnancy, showing

that it is not associated with adverse birth events and can

improve pregnancy outcomes [102, 103]. Influenza vacci-

nation of pregnant and postpartum women reduces the

incidence of influenza in infants up to 6 months of age. In

the United States, inactivated influenza vaccination of

pregnant women is recommended during influenza epi-

demics because it is the most effective technique for pre-

venting severe cases of influenza [104].

Based on the above, it is recommended that pregnant

women with IBD, who are considered to be more suscep-

tible to severe influenza, receive inactivated influenza

vaccine when they desire vaccination.

J Gastroenterol

123

Q17. Can an infant born of a woman with IBD

who received immunosuppressive treatment

during pregnancy be vaccinated?

[Statement]

(1) If the mother received a biologic medication

during the second or third trimester, then LAVs

are not recommended until the age of 6 months.

(2) Infants born to mothers with IBD who

received steroids and thiopurines during

pregnancy are recommended to follow the

standard vaccination schedule.

[Commentary]

Currently, four types of inactivated vaccines (Hib,

pneumococcal vaccine, hepatitis B vaccine, and DTaP-IPV

vaccine) and two LAVs (BCG and rotavirus) are recom-

mended for standard vaccination at\1 year of age in

Japan (Fig. 1).

For many biologics, drug concentrations in cord blood

are reportedly higher than those in maternal plasma (in-

fliximab, about 1.6 times; adalimumab, about 1.5 times;

vedolizumab, about 0.8 times; and ustekinumab, about 1.8

times) [105–107]. Because of the long plasma disappear-

ance half-life of biologics, these drugs were still

detectable in the blood at 3–6 months of age in infants born

to mothers with IBD who had received infliximab and

adalimumab in the third trimester of pregnancy [105, 108].

In a single case report describing a child of a mother

exposed to infliximab during pregnancy, the child received

a BCG vaccination at 3 months of age and then died of

disseminated BCG infection at 4.5 months of age [63].

Although some reports have indicated that no major

problems were observed in the children [109, 110],

national and international professional guidelines recom-

mend that infants of mothers who received biologics during

the second or third trimester of pregnancy should avoid

LAVs and BCG vaccination for at least 6 months after

birth [61, 111–113].

Administration of the rotavirus vaccine is recommended

by 14 weeks 6 days after birth to avoid vaccination at

6 months to 2 years of age, which is a susceptible period

for the development of vaccination-associated intussus-

ception. Therefore, it is difficult to actively recommend

vaccination of infants born to mothers with IBD who

received immunosuppressive treatment during pregnancy.

Some reports have indicated no apparent increase in

adverse reactions after rotavirus vaccination by 6 months

of age in infants born to mothers with IBD who had

received biologic agents (infliximab, adalimumab, ustek-

inumab, or vedolizumab) during pregnancy [114, 115].

Administration of inactivated vaccines should follow a

standard vaccination schedule. Several reports have

described an adequate immune response with no adverse

events after inactivated vaccines were given to infants born

to mothers with IBD who had received immunosuppressive

treatment during pregnancy [110, 114, 116].

Placental transfer has been measured for prednisolone

[117], thiopurines [118, 119], and immunosuppressive

drugs (tacrolimus and cyclosporine) [120, 121], and the

cord blood concentrations of all were reportedly lower than

those in maternal plasma. Because the plasma elimination

half-life of these drugs is shorter than that of biologics, the

use of any of these agents until delivery is not considered to

adversely affect either live or inactivated vaccines given

according to the standard vaccination schedule in Japan

[122].

One report described B-cell hypofunction in the cord

blood of an infant born to a mother who had continued

azathioprine during pregnancy, but the infant’s immune

function was normal at 1 month of age [123]. Other reports

have described blood cell abnormalities and decreased

immune function in infants born to mothers who had

continued azathioprine during pregnancy, and all of these

abnormalities normalized within about 2 months

[124, 125].

Q18. Can breastfeeding women with IBD be

vaccinated?

[Statement]

Mothers with IBD can be vaccinated while breastfeeding.

[Commentary]

In Japan, vaccination is considered to have no adverse

effect on the safety of breast milk when live or inactivated

vaccines are administered to nursing women [126].

Although components of the rubella vaccine are reportedly

secreted into breast milk and transient asymptomatic

infection has been observed in infants [127], most infants

did not show clinical symptoms, and the rubella vaccina-

tion of the children was not affected [128, 129].

The yellow fever vaccine is rarely administered in

Japan; however, some reports have described yellow fever

vaccine-associated encephalitis and neurological disorders

in infants after the vaccine was administered to lactating

mothers [130, 131]. The CDC states that lactating mothers

should be vaccinated only if travel to a yellow fever-en-

demic area is unavoidable [126].

J Gastroenterol

123

Q19. Can breastfed infants of women with IBD receiving

immunosuppressive therapy be vaccinated?

[Statement]

(1) Breastfed infants of mothers with IBD

receiving biologics are recommended to be

vaccinated according to the standard schedule.

(2) Breastfed infants of mothers with IBD

receiving steroids and thiopurines are also

recommended to be vaccinated according to the

standard schedule.

[Commentary]

Factors that enhance drug transfer to breast milk include

low blood protein binding, basic drugs, high lipophilicity,

and small molecular weight.

Biologics have very high molecular weights, which

limits their transfer to breast milk. Infliximab and adali-

mumab have been detected in very small amounts by

sensitive assays, but no reports have described adverse

events in breasted infants [132–134]. Because the oral

bioavailability of biologics is extremely low, even if a

small amount of a drug is ingested orally by an infant

through breast milk, it is hardly absorbed and thus poses no

problem for the infant’s vaccination.

Prednisolone and thiopurines have been measured in the

milk of breastfeeding women who are orally receiving

these drugs, and the amount of drug ingested by fully

lactating infants from breast milk is estimated to be about

1–5% of the infant’s therapeutic dose; additionally, the

estimated rate of adverse events in lactating infants is low

[135, 136]. Expert guidelines in North America and Europe

consider prednisolone and thiopurines to be safe for use

during breastfeeding [111, 112, 137–140]. Because of the

low likelihood of affecting the infant’s immune function,

general vaccination of infants is not considered

problematic.

During steroid pulse therapy, the amount of drug

ingested by the infant via breast milk increases with the

dose of medication; therefore, the clinician must carefully

consider whether breastfeeding should be performed. If a

breastfed infant shows symptoms that suggest an adverse

event caused by a drug transferred to breast milk, the

infant’s drug blood level should be measured and immune

function evaluated before vaccination.

Acknowledgements This work was supported in part by a Health

and Labour Sciences Research Grant for Research on

Intractable Diseases from the Ministry of Health, Labour and Welfare

of Japan (Hisamatsu 20316729). The authors thank Angela Morben,

DVM, ELS, from Edanz (https://jp.edanz.com/ac), for editing a draft

of this manuscript.

Declarations

Conflict of interest Any financial relationships with enterprises,

businesses, or academic institutions in the subject matter or materials

discussed in the manuscript are listed as follows. (1) Those from

which the authors or the authors’ spouse, partner, or immediate rel-

atives have individually received any income, honoraria, or any other

types of remuneration: AbbVie, Astellas Pharma, Celgene, Chugai

Pharmaceutical, EA Pharma, Janssen Pharmaceutical, JIMRO, Kissei

Pharmaceutical, Kyorin Pharmaceutical, Mitsubishi Tanabe Pharma,

Mochida Pharmaceutical, Nichi-Iko Pharmaceutical, Nobel Pharma,

Pfizer, Takeda Pharmaceutical, and UCB Japan. (2) Those from

which the authors have received scholarship/research grants: AbbVie,

Alfresa Pharma, AstraZeneca, Chugai Pharmaceutical, Daiichi-San-

kyo, EA Pharma, EP-CRSU, Janssen Pharmaceutical, JIMRO, Kyorin

Pharmaceutical, Mitsubishi Tanabe Pharma, Mochida Pharmaceuti-

cal, Nippon Kayaku, Pfizer, Taiju Life Social Welfare Foundation,

Takeda Pharmaceutical, TERUMO, Vaccination Research Center,

and ZERIA Pharmaceutical. (3) Those from which the authors have

individually received an endowed chair: AbbVie, EA Pharma, Mit-

subishi Tanabe Pharma, and ZERIA Pharmaceutical.

References

1. Benchimol EI, Tse F, Carroll MW, et al. Canadian Association

of Gastroenterology clinical practice guideline for immuniza-

tions in patients with inflammatory bowel disease (IBD)—part

1: live vaccines. Gastroenterology. 2021;161:669–80.

2. Kucharzik T, Ellul P, Greuter T, et al. ECCO guidelines on the

prevention, diagnosis, and management of infections in

inflammatory bowel disease. J Crohn’s Colitis.

2021;15:879–913.

3. Toruner M, Loftus EV, Harmsen WS, et al. Risk factors for

opportunistic infections in patients with inflammatory bowel

disease. Gastroenterology. 2008;134:929–36.

4. van Aalst M, Lo¨tsch F, Spijker R, et al. Incidence of invasive

pneumococcal disease in immunocompromised patients: a sys-

tematic review and meta-analysis. Travel Med Infect Dis.

2018;24:89–100.

5. Stobaugh DJ, Deepak P, Ehrenpreis ED. Hospitalizations for

vaccine preventable pneumonias in patients with inflammatory

bowel disease: a 6-year analysis of the nationwide inpatient

sample. Clin Exp Gastroenterol. 2013;6:43–9.

6. Nugent Z, Singh H, Targownik LE, et al. Herpes zoster infection

and herpes zoster vaccination in a population-based sample of

persons with IBD: is there still an unmet need? Inflamm Bowel

Dis. 2019;25:532–40.

7. Tsai SY, Yang TY, Lin CL, et al. Increased risk of varicella

zoster virus infection in inflammatory bowel disease in an Asian

population: a nationwide population-based cohort study. Int J

Clin Pract. 2015;69:228–34.

8. Allegretti JR, Barnes EL, Cameron A. Are patients with

inflammatory bowel disease on chronic immunosuppressive

therapy at increased risk of cervical high-grade dys-

plasia/cancer? A meta-analysis. Inflamm Bowel Dis.

2015;21:1089–97.

9. Tinsley A, Navabi S, Williams ED, et al. Increased risk of

influenza and influenza-related complications among 140,480

patients with inflammatory bowel disease. Inflamm Bowel Dis.

2019;25:369–76.

10. Chen D, Luo S, Ben Q, et al. Prevalence of hepatitis B and C and

factors for infection and nonimmune in inflammatory bowel

disease patients in China. Eur J Gastroenterol Hepatol.

2017;29:509–15.

J Gastroenterol

123

11. Tolentino YFM, Fogac?a HS, Zaltman C, et al. Hepatitis B virus

prevalence and transmission risk factors in inflammatory bowel

disease patients at Clementino Fraga Filho university hospital.

World J Gastroenterol. 2008;14:3201–6.

12. Ardesia M, Costantino G, Mondello P, et al. Serology of viral

infections and tuberculosis screening in an IBD population

referred to a tertiary centre of Southern Italy. Gastroenterol Res

Pract. 2017;2017:4139656.

13. Chevaux JB, Nani A, Oussalah A, et al. Prevalence of hepatitis

B and C and risk factors for nonvaccination in inflammatory

bowel disease patients in Northeast France. Inflamm Bowel Dis.

2010;16:916–24.

14. Drafting Committee for Hepatitis Management Guidelines, the

Japan Society of Hepatology. Japan Society of Hepatology

guidelines for the management of hepatitis B virus infection:

2019 update. Hepatol Res. 2020;50:791–816.

15. Permar SR, Griffin DE, Letvin NL. Immune containment and

consequences of measles virus infection in healthy and

immunocompromised individuals. Clin Vaccine Immunol.

2006;13:437–43.

16. Cullen G, Baden RP, Cheifetz AS. Varicella zoster virus

infection in inflammatory bowel disease. Inflamm Bowel Dis.

2012;18:2392–403.

17. Adams DJ, Nylund CM. Hospitalization for varicella and zoster

in children with inflammatory bowel disease. J Pediatr.

2016;171:140–5.

18. Kamei K, Miyairi I, Ishikura K, et al. Prospective study of live

attenuated vaccines for patients with nephrotic syndrome

receiving immunosuppressive agents. J Pediatr.

2018;196:217–22.

19. Bruijning-Verhagen P, Nipshagen MD, de Graaf H, et al.

Rotavirus disease course among immunocompromised patients;

5-year observations from a tertiary care medical centre. J Infect.

2017;75:448–54.

20. Tangye SG, Al-Herz W, Bousfiha A, et al. Human inborn errors

of immunity: 2019 update on the classification from the Inter-

national Union of Immunological Societies Expert Committee.

J Clin Immunol. 2020;40:24–64.

21. Pazmandi J, Kalinichenko A, Ardy RC, et al. Early-onset

inflammatory bowel disease as a model disease to identify key

regulators of immune homeostasis mechanisms. Immunol Rev.

2019;287:162–85.

22. Nambu R, Warner N, Mulder DJ, et al. A systematic review of

monogenic inflammatory bowel disease. Clin Gastroenterol

Hepatol. 2022;20:e653–63.

23. Sobh A, Bonilla FA. Vaccination in primary immunodeficiency

disorders. J Allergy Clin Immunol Pract. 2016;4:1066–75.

24. Kawashima H, Hasegawa D, Nakamura M, et al. Hazards of

early BCG vaccination: BCGitis in a patient with chronic

granulomatous disease. Pediatr Int. 2007;49:418–9.

25. Nakase H, Uchino M, Shinzaki S, et al. Evidence-based clinical

practice guidelines for inflammatory bowel disease 2020.

J Gastroenterol. 2021;56:489–526.

26. Pneumococcal vaccination: who and when to vaccinate. In:

Centers for Disease Control and Prevention (CDC). https://

www.cdc.gov/vaccines/vpd/pneumo/hcp/who-when-to-vacci

nate.html. Accessed 8 June 2022.

27. Demicheli V, Jefferson T, Di Pietrantonj C, et al. Vaccines for

preventing influenza in the elderly. Cochrane Database Syst

Rev. 2018;2:CD004876.

28. Gupta G, Lautenbach E, Lewis JD. Incidence and risk factors for

herpes zoster among patients with inflammatory bowel disease.

Clin Gastroenterol Hepatol. 2006;4:1483–90.

29. Cottone M, Kohn A, Daperno M, et al. Advanced age is an

independent risk factor for severe infections and mortality in

patients given anti-tumor necrosis factor therapy for inflamma-

tory bowel disease. Clin Gastroenterol Hepatol. 2011;9:30–5.

30. Winthrop KL, Melmed GY, Vermeire S, et al. Herpes zoster

infection in patients with ulcerative colitis receiving tofacitinib.

Inflamm Bowel Dis. 2018;24:2258–65.

31. Leung VS, Nguyen MT, Bush TM. Disseminated primary

varicella after initiation of infliximab for Crohn’s disease. Am J

Gastroenterol. 2004;99:2503–4.

32. Ritz MA, Jost R. Severe pneumococcal pneumonia following

treatment with infliximab for Crohn’s disease. Inflamm Bowel

Dis. 2001;7:327.

33. Long MD, Martin C, Sandler RS, et al. Increased risk of

pneumonia among patients with inflammatory bowel disease.

Am J Gastroenterol. 2013;108:240–8.

34. Millonig G, Kern M, Ludwiczek O, et al. Subfulminant hepatitis

B after infliximab in Crohn’s disease: need for HBV-screening?

World J Gastroenterol. 2006;12:974–6.

35. Culver E, Travis S. How to manage the infectious risk under

anti-TNF in inflammatory bowel disease. Curr Drug Targets.

2010;11:198–218.

36. Viget N, Vernier-Massouille G, Salmon-Ceron D, et al.

Opportunistic infections in patients with inflammatory bowel

disease: prevention and diagnosis. Gut. 2008;57:549–58.

37. Selby L, Hoellein A, Wilson JF. Are primary care providers

uncomfortable providing routine preventive care for inflamma-

tory bowel disease patients? Dig Dis Sci. 2011;56:819–24.

38. Crawford NW, Catto-Smith AG, Oliver MR, et al. An Australian

audit of vaccination status in children and adolescents with

inflammatory bowel disease. BMC Gastroenterol. 2011;11:87.

39. Loras C, Gisbert JP, M?′nguez M, et al. Liver dysfunction related

to hepatitis B and C in patients with inflammatory bowel disease

treated with immunosuppressive therapy. Gut. 2010;59:1340–6.

40. Park SH, Yang SK, Lim YS, et al. Clinical courses of chronic

hepatitis B virus infection and inflammatory bowel disease in

patients with both diseases. Inflamm Bowel Dis.

2012;18:2004–10.

41. Huang ML, Xu XT, Shen J, et al. Prevalence and factors related

to hepatitis B and C infection in inflammatory bowel disease

patients in China: a retrospective study. J Crohns Colitis.

2014;8:282–7.

42. Harsh P, Gupta V, Kedia S, et al. Prevalence of hepatitis B,

hepatitis C and human immunodeficiency viral infections in

patients with inflammatory bowel disease in north India. Intest

Res. 2017;15:97–102.

43. Pittet LF, Verolet CM, Michetti P, et al. Risk of vaccine-pre-

ventable infections in Swiss adults with inflammatory bowel

disease. Digestion. 2021;102:956–64.

44. Rahier JF, Magro F, Abreu C, et al. Second European evidence-

based consensus on the prevention, diagnosis and management

of opportunistic infections in inflammatory bowel disease.

J Crohn’s Colitis. 2014;8:443–68.

45. Jones JL, Tse F, Carroll MW, et al. Canadian Association of

Gastroenterology clinical practice guideline for immunizations

in patients with inflammatory bowel disease (IBD)-part 2:

inactivated vaccines. Gastroenterology. 2021;161:681–700.

46. Mir FA, Kane SV. Health maintenance in inflammatory bowel

disease. Curr Gastroenterol Rep. 2018;20:23.

47. Vinsard DG, Wakefield D, Vaziri H, et al. Vaccine-pre-

ventable diseases in hospitalized patients with inflammatory

bowel disease: a nationwide cohort analysis. Inflamm Bowel

Dis. 2019;25:1966–73.

48. Ning L, Liu R, Li S, et al. Increased risk of herpes zoster

infection in patients with inflammatory bowel disease: a meta-

analysis of cohort studies. Eur J Clin Microbiol Infect Dis.

2020;39:219–27.

J Gastroenterol

123

49. Gidengil C, Chen C, Parker AM, et al. Beliefs around childhood

vaccines in the United States: a systematic review. Vaccine.

2019;37:6793–802.

50. Mikamo H, Taya K, Ishiguro N, et al. Vaccine guidelines for

healthcare professionals Ver.3. Jpn J Environ Infect.

2020;35:S1–31.

51. JSHCT Guideline Committee. Vaccinations. In: Guideline of the

Japan Society for Hematopoietic Cell Transplantation, 3rd edn.

Tokyo: Japan Society for Hematopoietic Cell Transplantation;

2018.

52. Kamei K, Miyairi I, Ishikura K, et al. Prospective study of live

attenuated vaccines for patients receiving immunosuppressive

agents. PLoS ONE. 2020;15: e0240217.

53. Plotkin SA. Correlates of protection induced by vaccination.

Clin Vaccine Immunol. 2010;17:1055–65.

54. McLean HQ, Fiebelkorn AP, Temte JL, et al. Prevention of

measles, rubella, congenital rubella syndrome, and mumps,

2013: summary recommendations of the Advisory Committee

on Immunization Practices (ACIP). MMWR Recomm Rep.

2013;62:1–34.

55. Caldera F, Misch EA, Saha S, et al. Immunosuppression does

not affect antibody concentrations to measles, mumps, and

rubella in patients with inflammatory bowel disease. Dig Dis

Sci. 2019;64:189–95.

56. Schenk J, Abrams S, Theeten H, et al. Immunogenicity and

persistence of trivalent measles, mumps, and rubella vaccines: a

systematic review and meta-analysis. Lancet Infect Dis.

2021;21:286–95.

57. Marin M, Gu¨ris D, Chaves SS, et al. Prevention of varicella:

recommendations of the Advisory Committee on Immunization

Practices (ACIP). MMWR Recomm Rep. 2007;56:1–40.

58. Asada H, Nagayama K, Okazaki A, et al. An inverse correlation

of VZV skin-test reaction, but not antibody, with severity of

herpes zoster skin symptoms and zoster-associated pain. J Der-

matol Sci. 2013;69:243–9.

59. Imoto K, Okazaki A, Onishi F, et al. VZV skin-test reaction, but

not antibody, is an important predictive factor for postherpetic

neuralgia. J Dermatol Sci. 2015;79:235–40.

60. Khan N, Trivedi C, Kavani H, et al. Efficacy of live attenuated

herpes zoster vaccine in patients with inflammatory bowel dis-

eases. Clin Gastroenterol Hepatol. 2019;17:1341–7.

61. Japanese Society for Pediatric Infectious Diseases. Immuniza-

tion guideline for children post-organ transplantation and under

immunosuppressive treatment 2014. Tokyo: Kyowa kikaku;

2014.

62. Whittembury A, Ramirez G, Herna′ndez H, et al. Viscerotropic

disease following yellow fever vaccination in Peru. Vaccine.

2009;27:5974–81.

63. Cheent K, Nolan J, Shariq S, et al. Case report: fatal case of

disseminated BCG infection in an infant born to a mother taking

infliximab for Crohn’s disease. J Crohns Colitis. 2010;4:603–5.

64. Wasan SK, Zullow S, Berg A, et al. Herpes zoster vaccine

response in inflammatory bowel disease patients on low-dose

immunosuppression. Inflamm Bowel Dis. 2016;22:1391–6.

65. Wichmann A, Cleveland NK, Rubin DT. Safety and efficacy of

live measles vaccine administered to a Crohn’s disease patient

receiving vedolizumab. Am J Gastroenterol. 2016;111:577.

66. Croce E, Hatz C, Jonker EF, et al. Safety of live vaccinations on

immunosuppressive therapy in patients with immune-mediated

inflammatory diseases, solid organ transplantation or after bone-

marrow transplantation—a systematic review of randomized

trials, observational studies and case reports. Vaccine.

2017;35:1216–26.

67. Bernstein CN, Rawsthorne P, Blanchard JF. Population-based

case-control study of measles, mumps, and rubella and inflam-

matory bowel disease. Inflamm Bowel Dis. 2007;13:759–62.

68. Martinelli M, Giugliano FP, Strisciuglio C, et al. Vaccinations

and immunization status in pediatric inflammatory bowel dis-

ease: a multicenter study from the pediatric IBD Porto Group of

the ESPGHAN. Inflamm Bowel Dis. 2020;26:1407–14.

69. Veereman-Wauters G, De Ridder L, Veres G, et al. Risk of

infection and prevention in pediatric patients with IBD: ESP-

GHAN IBD Porto Group commentary. J Pediatr Gastroenterol

Nutr. 2012;54:830–7.

70. Caldera F, Ley D, Hayney MS, et al. Erratum to: Optimizing

immunization strategies in patients with IBD. Inflamm Bowel

Dis. 2021;27: e9.

71. Nguyen HT, Minar P, Jackson K, et al. Vaccinations in

immunosuppressive-dependent pediatric inflammatory bowel

disease. World J Gastroenterol. 2017;23:7644–52.

72. Cullen G, Bader C, Korzenik JR, et al. Serological response to

the 2009 H1N1 influenza vaccination in patients with inflam-

matory bowel disease. Gut. 2012;61:385–91.

73. Shirai S, Hara M, Sakata Y, et al. Immunogenicity of quadri-

valent influenza vaccine for patients with inflammatory bowel

disease undergoing immunosuppressive therapy. Inflamm Bowel

Dis. 2018;24:1082–91.

74. Gisbert JP, Villagrasa JR, Rodr?′guez-Nogueiras A, et al. Effi-

cacy of hepatitis B vaccination and revaccination and factors

impacting on response in patients with inflammatory bowel

disease. Am J Gastroenterol. 2012;107:1460–6.

75. Cossio-Gil Y, Mart?′nez-Go′mez X, Campins-Mart?′ M, et al.

Immunogenicity of hepatitis B vaccine in patients with inflam-

matory bowel disease and the benefits of revaccination. J Gas-

troenterol Hepatol. 2015;30:92–8.

76. Jacobson DL, Bousvaros A, Ashworth L, et al. Immunogenicity

and tolerability to human papillomavirus-like particle vaccine in

girls and young women with inflammatory bowel disease.

Inflamm Bowel Dis. 2013;19:1441–9.

77. Pittet LF, Verolet CM, Michetti P, et al. High immunogenicity

of the pneumococcal conjugated vaccine in immunocompro-

mised adults with inflammatory bowel disease. Am J Gas-

troenterol. 2019;114:1130–41.

78. Fiorino G, Peyrin-Biroulet L, Naccarato P, et al. Effects of

immunosuppression on immune response to pneumococcal

vaccine in inflammatory bowel disease: a prospective study.

Inflamm Bowel Dis. 2012;18:1042–7.

79. Heijstek MW, Ott de Bruin LM, Borrow R, et al. Vaccination in

paediatric patients with auto-immune rheumatic diseases: a

systemic literature review for the European League against

Rheumatism evidence-based recommendations. Autoimmun

Rev. 2011;11:112–22.

80. Farraye FA, Melmed GY, Lichtenstein GR, et al. ACG clinical

guideline: preventive care in inflammatory bowel disease. Am J

Gastroenterol. 2017;112:241–58.

81. Mamula P, Markowitz JE, Piccoli DA, et al. Immune response to

influenza vaccine in pediatric patients with inflammatory bowel

disease. Clin Gastroenterol Hepatol. 2007;5:851–6.

82. Debruyn J, Fonseca K, Ghosh S, et al. Immunogenicity of

influenza vaccine for patients with inflammatory bowel disease

on maintenance infliximab therapy: a randomized trial. Inflamm

Bowel Dis. 2016;22:638–47.

83. Lamb CA, Kennedy NA, Raine T, et al. British Society of

Gastroenterology consensus guidelines on the management of

inflammatory bowel disease in adults. Gut. 2019;68:s1-106.

84. Ferdinands JM, Gaglani M, Martin ET, et al. Waning vaccine

effectiveness against influenza-associated hospitalizations

among adults, 2015–2016 to 2018–2019, United States hospi-

talized adult influenza vaccine effectiveness network. Clin Infect

Dis. 2021;73:726–9.

85. El Omeiri N, Azziz-Baumgartner E, Thompson MG, et al.

Seasonal influenza vaccine effectiveness against laboratory-

J Gastroenterol

123

confirmed influenza hospitalizations—Latin America, 2013.

Vaccine. 2018;36:3555–66.

86. Matsumoto H, Ohfuji S, Watanabe K, et al. Booster influenza

vaccination does not improve immune response in adult

inflammatory bowel disease patients treated with immunosup-

pressives: a randomized controlled trial. J Gastroenterol.

2015;50:876–86.

87. Kimberlin DW, Whitley RJ. Varicella-zoster vaccine for the

prevention of herpes zoster. N Engl J Med. 2007;356:1338–43.

88. Dooling KL, Guo A, Patel M, et al. Recommendations of the

advisory committee on immunization practices for use of herpes

zoster vaccines. MMWR Morb Mortal Wkly Rep.

2018;67:103–8.

89. Harbecke R, Cohen JI, Oxman MN. Herpes zoster vaccines.

J Infect Dis. 2021;224:S429–42.

90. Lal H, Cunningham AL, Godeaux O, et al. Efficacy of an

adjuvanted herpes zoster subunit vaccine in older adults. N Engl

J Med. 2015;372:2087–96.

91. Cunningham AL, Heineman TC, Lal H, et al. Immune responses

to a recombinant glycoprotein E herpes zoster vaccine in adults

aged 50 years or older. J Infect Dis. 2018;217:1750–60.

92. Kochhar GS, Desai A, Caldera FD, et al. Effectiveness of

recombinant zoster vaccine (RZV) in patients with inflammatory

bowel disease. Vaccine. 2021;39:4199–202.

93. Suzuki M, Dhoubhadel BG, Ishifuji T, et al. Serotype-specific

effectiveness of 23-valent pneumococcal polysaccharide vaccine

against pneumococcal pneumonia in adults aged 65 years or

older: a multicentre, prospective, test-negative design study.

Lancet Infect Dis. 2017;17:313–21.

94. Debruyn JCC, Hilsden R, Fonseca K, et al. Immunogenicity and

safety of influenza vaccination in children with inflammatory

bowel disease. Inflamm Bowel Dis. 2012;18:25–33.

95. Elkayam O, Bashkin A, Mandelboim M, et al. The effect of

infliximab and timing of vaccination on the humoral response to

influenza vaccination in patients with rheumatoid arthritis and

ankylosing spondylitis. Semin Arthritis Rheum. 2010;39:442–7.

96. Epidemiological information of rubella in Japan. In: National

institute of infectious diseases. https://www.niid.go.jp/niid/ja/

rubella-m-111/rubella-top/2145-rubella-related/8278-

rubella1808.html. Accessed 8 June 2022.

97. Pregnancy guidelines and recommendations by vaccine. In:

Centers for Disease Control and Prevention (CDC). https://

www.cdc.gov/vaccines/pregnancy/hcp-toolkit/guidelines.html.

Accessed 8 June 2022.

98. Mertz D, Geraci J, Winkup J, et al. Pregnancy as a risk factor for

severe outcomes from influenza virus infection: a systematic

review and meta-analysis of observational studies. Vaccine.

2017;35:521–8.

99. Siston AM, Rasmussen SA, Honein MA, et al. Pandemic 2009

influenza A(H1N1) virus illness among pregnant women in the

United States. JAMA. 2010;303:1517–25.

100. Fell DB, Platt RW, Basso O, et al. The relationship between

2009 pandemic H1N1 influenza during pregnancy and preterm

birth: a population-based cohort study. Epidemiology.

2018;29:107–16.

101. Gunnes N, Gjessing HK, Bakken IJ, et al. Seasonal and pan-

demic influenza during pregnancy and risk of fetal death: a

Norwegian registry-based cohort study. Eur J Epidemiol.

2020;35:371–9.

102. Pasternak B, Svanstrom H, M?lgaard-Nielsen D, et al. Risk of

adverse fetal outcomes following administration of a pandemic

influenza A (H1N1) vaccine during pregnancy. JAMA.

2012;308:165–74.

103. Newsome K, Alverson CJ, Williams J, et al. Outcomes of infants

born to women with influenza A(H1N1)pdm09. Birth Defects

Res. 2019;111:88–95.

104. ACOG Committee Opinion No. 732: Influenza Vaccination

During Pregnancy. Obstet Gynecol. 2018; 131:e109–14.

105. 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;11:286–92.

106. Julsgaard M, Kjeldsen J, Brock B, et al. Letter: vedolizumab

drug levels in cord and maternal blood in women with inflam-

matory bowel disease. Aliment Pharmacol Ther. 2018;48:386–8.

107. Rowan CR, Cullen G, Mulcahy HE, et al. Ustekinumab drug

levels in maternal and cord blood in a woman with Crohn’s

disease treated until 33 weeks of gestation. J Crohns Colitis.

2018;12:376–8.

108. Julsgaard M, Christensen LA, Gibson PR, et al. Concentrations

of adalimumab and infliximab in mothers and newborns, and

effects on infection. Gastroenterology. 2016;151:110–9.

109. Bortlik M, Duricova D, Machkova N, et al. Impact of anti-tumor

necrosis factor alpha antibodies administered to pregnant

women with inflammatory bowel disease on long-term outcome

of exposed children. Inflamm Bowel Dis. 2014;20:495–501.

110. Duricova D, Dvorakova E, Hradsky O, et al. Safety of anti-Tnf-

alpha therapy during pregnancy on long-term outcome of

exposed children: a controlled, multicenter observation. Inflamm

Bowel Dis. 2019;25:789–96.

111. van der Woude CJ, Ardizzone S, Bengtson MB, et al. The

second European evidenced-based consensus on reproduction

and pregnancy in inflammatory bowel disease. J Crohns Colitis.

2015;9:107–24.

112. Nguyen GC, Seow CH, Maxwell C, et al. The Toronto con-

sensus statements for the management of inflammatory bowel

disease in pregnancy. Gastroenterology. 2016;150:734–57.

113. Furer V, Rondaan C, Heijstek MW, et al. 2019 update of

EULAR recommendations for vaccination in adult patients with

autoimmune inflammatory rheumatic diseases. Ann Rheum Dis.

2020;79:39–52.

114. Beaulieu DB, Ananthakrishnan AN, Martin C, et al. Use of

biologic therapy by pregnant women with inflammatory bowel

disease does not affect infant response to vaccines. Clin Gas-

troenterol Hepatol. 2018;16:99–105.

115. Moens A, van der Woude CJ, Julsgaard M, et al. Pregnancy

outcomes in inflammatory bowel disease patients treated with

vedolizumab, anti-TNF or conventional therapy: results of the

European CONCEIVE study. Aliment Pharmacol Ther.

2020;51:129–38.

116. De Lima A, Kanis SL, Escher JC, et al. Hepatitis B vaccination

effective in children exposed to anti-tumour necrosis factor

alpha in utero. J Crohns Colitis. 2018;12:948–53.

117. Beitins IZ, Bayard F, Ances IG, et al. The transplacental passage

of prednisone and prednisolone in pregnancy near term. J Pedi-

atr. 1972;81:936–45.

118. De Boer NKH, Jarbandhan SVA, De Graaf P, et al. Azathioprine

use during pregnancy: unexpected intrauterine exposure to

metabolites. Am J Gastroenterol. 2006;101:1390–2.

119. Jharap B, De Boer NKH, Stokkers P, et al. Intrauterine exposure

and pharmacology of conventional thiopurine therapy in preg-

nant patients with inflammatory bowel disease. Gut.

2014;63:451–7.

120. Zheng S, Easterling TR, Hays K, et al. Tacrolimus placental

transfer at delivery and neonatal exposure through breast milk.

Br J Clin Pharmacol. 2013;76:988–96.

121. Burrows DA, O’Neil TJ, Sorrells TL. Successful twin pregnancy

after renal transplant maintained on cyclosporine A immuno-

suppression. Obstet Gynecol. 1988;72:459–61.

122. Luu M, Benzenine E, Barkun A, et al. Safety of first year vac-

cination in children born to mothers with inflammatory bowel

disease and exposed in utero to anti-TNFa agents: a French

J Gastroenterol

123

nationwide population-based cohort. Aliment Pharmacol Ther.

2019;50:1181–8.

123. Kaneko K, Kawai T, Watanabe N, et al. Spontaneous recovery

from suppressed B cell production and proliferation caused by

intrauterine azathioprine exposure in the fetal period. Lupus.

2019;28:1027–8.

124. Cote′ CJ, Meuwissen HJ, Pickering RJ. Effects on the neonate of

prednisone and azathioprine administered to the mother during

pregnancy. J Pediatr. 1974;85:324–8.

125. Thomas C, Monteil-Ganiere C, Mirallie′ S, et al. A severe

neonatal lymphopenia associated with administration of aza-

thioprine to the mother in a context of Crohn’s disease. J Crohns

Colitis. 2018;12:258–61.

126. ACIP general best practice guidelines for immunization. In:

Centers for Disease Control and Prevention (CDC). https://

www.cdc.gov/vaccines/hcp/acip-recs/general-recs/index.html.

Accessed 9 June 2022.

127. Losonsky GA, Mark Fishaut J, Strussenberg J, et al. Effect of

immunization against rubella on lactation products. II. Mater-

nal–neonatal interactions. J Infect Dis. 1982;145:661–6.

128. Krogh V, Duffy LC, Wong D, et al. Postpartum immunization

with rubella virus vaccine and antibody response in breast-

feeding infants. J Lab Clin Med. 1989;113:695–9.

129. Grillner L, Hedstro¨m CE, Bergstro¨m H, et al. Vaccination

against rubella of newly delivered women. Scand J Infect Dis.

1973;5:237–41.

130. Centers for Disease Control and Prevention (CDC). Transmis-

sion of yellow fever vaccine virus through breast-feeding—

Brazil, 2009. MMWR Morb Mortal Wkly Rep. 2010;59:130–2.

131. Traiber C, Coelho-Amaral P, Fonteles Ritter VR, et al. Infant

meningoencephalitis caused by yellow fever vaccine virus

transmitted via breastmilk. J Pediatr (Rio J). 2011;87:269–72.

132. Ben-Horin S, Yavzori M, Kopylov U, et al. Detection of

infliximab in breast milk of nursing mothers with inflammatory

bowel disease. J Crohns Colitis. 2011;5:555–8.

133. Matro R, Martin CF, Wolf D, et al. Exposure concentrations of

infants breastfed by women receiving biologic therapies for

inflammatory bowel diseases and effects of breastfeeding on

infections and development. Gastroenterology.

2018;155:696–704.

134. Fritzsche J, Pilch A, Mury D, et al. Infliximab and adalimumab

use during breastfeeding. J Clin Gastroenterol. 2012;46:718–9.

135. Drugs and Lactation Database (LactMed). In: NCBI Bookshelf.

https://www.ncbi.nlm.nih.gov/books/NBK501922/. Accessed 9

June 2022.

136. Hale TW. Hale’s medications and mothers’ milk 2021: a manual

of lactational pharmacology. 19th ed. New York: Springer

Publishing; 2021.

137. Constantinescu S, Pai A, Coscia LA, et al. Breast-feeding after

transplantation. Best Pract Res Clin Obstet Gynaecol.

2014;28:1163–73.

138. Skorpen CG, Hoeltzenbein M, Tincani A, et al. The EULAR

points to consider for use of antirheumatic drugs before preg-

nancy, and during pregnancy and lactation. Ann Rheum Dis.

2016;75:795–810.

139. Mahadevan U, Robinson C, Bernasko N, et al. Inflammatory

bowel disease in pregnancy clinical care pathway: a report from

the American Gastroenterological Association IBD parenthood

project working group. Gastroenterology. 2019;156:1508–24.

140. Sammaritano LR, Bermas BL, Chakravarty EE, et al. 2020

American College of Rheumatology guideline for the manage-

ment of reproductive health in rheumatic and musculoskeletal

diseases. Arthritis Rheumatol. 2020;72:529–56.

Publisher’s Note Springer Nature remains neutral with regard to

jurisdictional claims in published maps and institutional affiliations.

Springer Nature or its licensor (e.g. a society or other partner) holds

exclusive rights to this article under a publishing agreement with the

author(s) or other rightsholder(s); author self-archiving of the

accepted manuscript version of this article is solely governed by the

terms of such publishing agreement and applicable law.

J Gastroenterol

123

献花(0)
+1
(本文系金鑫康复堂首藏)