Korean J Intern Med > Volume 37(5); 2022 > Article
Park and Cheon: Updates on conventional therapies for inflammatory bowel diseases: 5-aminosalicylates, corticosteroids, immunomodulators, and anti-TNF-α

Abstract

The incidence and prevalence of inflammatory bowel diseases (IBDs) are rapidly increasing worldwide. IBDs are considered an emerging problem not only in Western countries but also in developing counties. The relapses and complications of active IBD mandate various medications. Nevertheless, hospitalization, emergency room visits, or surgery may be required, resulting in a socioeconomic burden. Great advances have been made in the development of new therapeutic options for IBD to achieve induction and maintenance remission. Nevertheless, conventional therapy is still the mainstay in the treatment of IBD. This review article provides an update on recent advances in conventional therapies, including 5-aminosalicylates, corticosteroids, immunomodulators, and anti-tumor necrosis factor-α agents to treat IBD.

INTRODUCTION

Inflammatory bowel diseases (IBDs), including ulcerative colitis (UC) and Crohn’s disease (CD), are characterized by chronic immune-mediated intestinal inflammation of the gastrointestinal tract [1]. Despite the emergence of new biological agents and small molecules for treating IBDs, conventional therapies including 5-aminosalicylates (5-ASA), corticosteroids, immunomodulators, and anti-tumor necrosis factor-α (TNF-α) agents are still the mainstay to induce and maintain clinical remission of IBD because of their effectiveness, safety, and acceptable cost [2]. Medical treatment, personalized care through a multidisciplinary team approach, and validated information for patients is critical for IBD treatment [3,4]. The International Organization for the Study of Inflammatory Bowel Diseases updated the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE)-II statement to optimize IBD treatment; symptomatic remission and normalization of C-reactive protein levels were short-term targets, decreasing the calprotectin level was an intermediate-term target, and endoscopic healing and normalization of quality of life were long-term targets [5,6]. Here, we review the updated data on the current experience and outcomes of conventional IBD therapies for optimizing medical treatments suggested by the STRIDE-II statement. This review provides an overview and practical treatment tips for conventional IBD therapies useful at primary and secondary medical institutions.

5-ASA

Various 5-ASA-based drug formulations

5-ASA-based drugs include sulfasalazine, olsalazine, balsalazide, and mesalamine and are effective, safe, and inexpensive drugs for treating IBD, particularly UC (Table 1) [7]. Sulfasalazine (brand names include Salazopyrin, Hanlim Pharm Co., Seoul, Korea), which is an azo-bonded prodrug of sulfapyridine, and 5-ASA were originally proposed to treat rheumatoid arthritis in the late 1930s [8]. 5-ASA is metabolized from sulfasalazine by the gut bacterial enzyme azoreductase and was reported to ameliorate the intestinal inflammation of IBD patients in the mid-1970s, but sulfapyridine originating from sulfasalazine can cause intolerance to sulfapyridine and allergic reactions, including fever, nausea, vomiting, headache, angioedema, and diarrhea, and its use is gradually decreasing [9]. However, patients with rheumatic diseases, such as rheumatoid arthritis or ankylosing spondylitis, continue to use this drug because it treats arthritis and UC at the same time.
Second-generation azo-bonded 5-ASA drugs, which do not contain sulfapyridine, were developed in the early 1980s and these are converted into 5-ASA in the colon similar to sulfasalazine. Olsalazine (brand names include Dipentum, Pharmacia AB, Uppsala, Sweden) is a 5-ASA dimer and balsalazide (brand names include Colazal, Chong Kun Dang, Seoul, Korea) is an azo-bonded prodrug of 4-amino-benzoyl-β-alanine and 5-ASA. The adverse events of balsalazide are significantly less frequent than those of sulfasalazine [10].
In addition, the most commonly used 5-ASA-based drug, mesalamine (brand names include Asacol, Salofalk, Mezavant, and Pentasa, Ferring, Saint-Prex, Switzerland) has been developed in various newer formulations [11]. To reach the colon mucosa without being absorbed in the stomach and small intestine, 5-ASA must be bound to a drug-delivery system or prodrug [12,13]. Asacol, consisting of 5-ASA coated with Eudragit S100 (Daewoong, Seoul, Korea), which is a polymethacrylate copolymer that only dissolves at pH ≥ 7, targets the terminal ileum and colon [14]. OPTICORE coated 1,600 mg Asacol tablets were recently developed, and are composed of the two-trigger release technology [15]. The Phloral in the outer layer is composed of Eudragit S100, which is a polymethacrylate copolymer that only dissolves at pH ≥ 7, but is also resistant to starch and serves as an energy source for colonic microbiota, to trigger rapid release [15]. In addition, the Duocoat inner layer, which is composed of a partially neutralized enteric polymer with a buffer salt, promotes the dissolution of the outer enteric polymer layer [15]. Salofalk consisting of 5-ASA coated with Eudragit L100, which only dissolves at pH ≥ 6, targets the distal ileum and colon [14]. Mezavant consists of 5-ASA and the MMX system (Takeda, Tokyo, Japan) coated combination of Eudragit S100 and Eudragit L100, which delays and prolongs administration of mesalazine throughout the colon and only dissolves at pH ≥ 7 [16]. Pentasa consists of 5-ASA coated with a semipermeable ethylcellulose membrane. It is sensitive to moisture, released in a time-dependent manner, and targets from the duodenum through the small bowel to the colon [17]. The new Pentasa sachet formulations (1 and 2 g) were recently launched and are expected to improve medication adherence in patients who have difficulty swallowing large tablets [18].

Mechanisms of action of 5-ASA-based drugs

5-ASA is absorbed in the stomach and the small bowel without a drug-delivery system or prodrug. It is metabolized to an inactive form of N-acetyl-5-aminosalicylic acid by N-acetyltransferase (NAT) and is excreted in the urine or feces [19]. While it has no effect at a low dose because of conversion by NAT, it has a sufficient effect at a higher dose greater than that of the NAT saturation point [19]. It relieves inflammation after it reaches the inflamed terminal ileum or colon.
The mechanisms of action of 5-ASA-based drugs are still not understood, but several plausible hypotheses have been proposed. 5-ASA reduces the synthesis of prostaglandins and leukotrienes by downregulating inducible cyclooxygenase 2/prostaglandin E2 (COX-2/PGE2) signaling [20]. Furthermore, it acts as a peroxisome proliferator-activated receptor γ agonist to inhibit the production of inflammatory cytokines, such as interleukin (IL)-1, IL-6, and TNF-α, as well as the activation of nuclear factor kappa B (NF-κB) [21,22]. It also scavenges reactive oxygen and nitrogen species, which helps reduce intestinal inflammation [23]. In addition, it reduces intestinal inflammation through other mechanisms. These therapeutic effects have the advantage of producing fewer systemic side effects, as they only act locally on the intestinal mucosa and not systemically.

Induction doses of 5-ASA for UC patients

The various 5-ASA-based drug formulations are effective at inducing remission compared to placebo agents in patients with mild to moderate UC, and no differences in clinical efficacy have been reported between the various formulations [24]. The disease extent, patient preference, dose, and formulation are considered when selecting a 5-ASA drug. In patients with mild to moderate ulcerative proctitis, rectal 5-ASA therapy is recommended at 1 g per day for induction [25,26]. A rectal enema with 5-ASA can cover up to a splenic flexure and is, therefore, effective local treatment for left-sided colitis. In addition, oral 5-ASA therapy of ≥ 2.4 g per day is the standard treatment for induction in patients with mild to moderate left-sided colitis or pancolitis [25,26]. In the Assessing the Safety and Clinical Efficacy of a New Dose of 5-ASA (ASCEND) trial, patients treated with 4.8 g per day of 5-ASA achieved more rapid symptomatic relief, biochemical remission, and endoscopic remission compared to patients treated with 2.4 g per day of 5-ASA, but no difference was observed in clinical remission [2729]. Combination therapy with oral and topical 5-ASA is superior to oral 5-ASA monotherapy for inducing mild to moderate UC [30] regardless of the disease extent. Therefore, high-dose oral 5-ASA combined with topical 5-ASA is preferred to treat moderate to severe UC patients.

Maintenance doses of 5-ASA for UC patients

In patients with mild to moderate ulcerative proctitis, 0.5 to 1 g per day of rectal 5-ASA therapy is recommended for maintenance remission [25,26]. In patients with mild to moderate left-sided colitis or pancolitis, ≥ 2 g per day of oral 5-ASA therapy is the standard treatment for maintenance remission [25,26]. A dose escalation of 5-ASA from 1.2 g per day to 2.4 g per day or 2.4 g per day to 4.8 g per day reduces fecal calprotectin levels and is associated with less frequent disease relapse [31]. Combination therapy of oral and topical 5-ASA is superior to oral 5-ASA monotherapy for maintenance of mild to moderate UC [32]. The efficacy and adherence between once-daily and divided dosing were not different in several meta-analyses [33,34]. However, a randomized control trial once-daily dosing group achieved a higher rate of clinical remission and adherence compared to a three-times-daily dosing group [35]. Simplifying drug dosing would improve real-world long-term drug adherence for maintenance therapy [36,37].

Use of 5-ASA in CD patients

The use of 5-ASA in CD patients is common in routine clinical practice, but recent evidence suggests no benefit of 5-ASA compared to placebo for induction and maintenance in CD patients [38,39]. To minimize financial burden and diminish unnecessary drug use, the Stopping Aminosalicylate Therapy in Inactive Crohn’s Disease (STATIC) study, which is an ongoing open-label, randomized, noninferiority randomized trial that compares a continuing group and a withdrawing group with CD during remission [40]. Nevertheless, the safety and effectiveness of 5-ASA for luminal CD patients has led physicians to use it in some cases, particularly in mild colonic disease, to avoid corticosteroids [41]. In a previous study, high-dose mesalamine was superior to placebo, but sulfasalazine was not significantly superior to placebo [41].
In one previous study, the clinical outcomes were not different between a 5-ASA withdrawal group and a 5-ASA continuation group of IBD patients initiated on immunomodulators or biologic agents [42]. Although further large-scale prospective studies are needed, discontinuing 5-ASA should be considered for patients in remission who are receiving immunomodulators or biologic agents.

Side effects of 5-ASA-based drugs

5-ASA-based drugs are generally safe and tolerable, but adverse reactions occur in some cases. 5-ASA can cause fever, headache, rash, vomiting, paradoxical diarrhea, muscle pain, and abdominal pain [43]. It rarely causes serious side effects, including pleuritis, pericarditis, pancreatitis, interstitial nephritis, or hepatotoxicity [44]. Side effects usually occur between 1 and 4 weeks and improve immediately after discontinuation. They are more common when taking sulfasalazine than when taking mesalamine. Sulfasalazine-treated patients should take a folic acid supplement because this drug decreases folic acid absorption.

CORTICOSTEROIDS

Corticosteroids are effective, rapidly acting drugs that induce remission of active moderate to severe IBD. They rapidly inhibit intestinal inflammation by reducing intestinal permeability, decreasing TNF-α production, and blocking NF-κB [45]. Systemic corticosteroids, including 40 to 60 mg (or 0.5 to 1 mg/kg) per day of prednisolone or 40 to 60 mg per day of methylprednisolone or 300 to 400 mg per day of hydrocortisone, should be initiated and tapered over 8 to 12 weeks depending on the initial drug response [24,26]. Steroid administration for < 3 weeks and prednisolone < 15 mg/day usually do not induce active IBD [46]. Long-term steroid use for more than 3 months is not recommended due to a lack of an effect in preventing flare-ups [47].
Despite the effectiveness of corticosteroids, their long-term use should be avoided because of their short-term adverse effects, such as acne, headache, electrolyte imbalance, hyperglycemia, and hypertension, and their long-term adverse effects, such as susceptibility to infection, osteoporosis, aseptic joint necrosis, and adrenal insufficiency [48]. Budesonide, a synthetic glucocorticosteroid with a high affinity for the glucocorticoid receptor, has higher topical potency and lower systemic bioavailability than systemic corticosteroids [49]. Budesonide MMX at a daily dose of 9 mg achieves clinical remission in active mild to moderate left-sided UC and in ileocecal CD patients who fail 5-ASA-based therapy [49,50]. However, this drug is currently not available in Korea. Beclomethasone dipropionate, a second-generation corticosteroid, at a 5 mg daily dose for 4 weeks and every other day for an additional 4 weeks is not inferior to 40 mg prednisolone per day for the initial 2 weeks and tapering 10 mg every 2 weeks during the 8 weeks of treatment for active left-sided or extensive UC [51,52]. Topical steroid agents, including beclomethasone dipropionate, are also beneficial for patients with 5-ASA refractory UC [53]. Topical 5-ASA has demonstrated effectiveness for inducing a clinical response or remission and preventing relapse in several studies. In a previous study, combined topical steroids and 5-ASA was more effective than topical 5-ASA or topical steroids alone for inducing a response (100% of patients with combination vs. 70% with beclomethasone alone and 76% with 5-ASA alone) [54]. These oral locally active steroids could be an alternative therapeutic option to reduce systemic side effects [55]. Corticosteroid-dependent patients who initially respond to corticosteroids but relapse after discontinuation or tapering or corticosteroid-refractory patients who do not respond to corticosteroids should consider the use of immunomodulators or biologic agents to reduce long-term inappropriate exposure to corticosteroids [56,57].

IMMUNOMODULATORS

Conventional immunomodulators, such as azathioprine (AZT), 6-mercaptopurine (6-MP), and methotrexate (MTX), are recommended for the maintenance of remission in patients with IBD who fail 5-ASA-based drugs and are dependent on or refractory to corticosteroids [58]. The efficacy of 2.0 to 2.5 mg/kg AZT and 6.0 to 1.5 mg/kg 6-MP once per day was confirmed for the maintenance of CD remission in a clinical study [59]. It is important to start with a low dose and increase to the target dose because the effect is often insufficient while maintaining a low dose. In addition, thiopurine monotherapy but not MTX is effective for long-term maintenance in UC [60]. The combined therapy of an anti-TNF-α agent and an immunomodulator reduces the immunogenicity and efficacy of anti-TNF-α agents in UC and CD [61]. Combination therapy with allopurinol and AZT decreases hepatotoxicity and increases effectiveness by reducing the AZT dose [62]. The subsequent leukocytopenia, thrombocytopenia, or pancytopenia caused by increases in active metabolites should be carefully monitored.
Thiopurines are metabolized to 6-thioguanine nucleotides (6-TGN) with therapeutic effects and to 6-methylmercaptopurine ribonucleotides without any therapeutic effect but are related to adverse events [63]. Therapeutic drug monitoring (TDM) and 6-TGN measurement have been suggested to optimize the efficacy of thiopurine therapy before step-up medical therapy [64,65]. In addition, 10% to 28% of IBD patients discontinue thiopurine therapy because of intolerable short-term adverse events or long-term adverse events including cervical neoplasia [66]. AZT-intolerant patients who develop dose-dependent side effects, including nausea, vomiting, hepatotoxicity, leukopenia, and pancreatitis, can switch to 6-MP therapy to gain comparable efficacy and tolerability with a half-dose of AZT [67,68].
Leucopenia needs special attention. Thiopurine S-methyl-transferase (TPMT) gene variants in Western countries have been associated with leukopenia, but are very rare in East Asian countries [69]. In East Asian countries, nudix hydrolase 15 (NUDT15) gene variants (T/T genotype) are more frequent than TPMT gene variants and they more accurately predict severe thiopurine-related leukopenia [70]. Blood tests for full blood counts, renal, and liver biochemistry, TDM of thiopurines, and genotyping of NUDT15 and TPMT in routine practice could improve the efficacy and safety of IBD patients [71,72]. In particular, genotyping NUDT15 is essential, and if it is not available, close monitoring should be performed with a complete blood count test at intervals of 1 to 2 weeks for the first 4 weeks.
MTX is often considered a second-line drug when there is resistance or intolerance to the use of AZT/6-MP, and its effectiveness has been proven [73]. In a recent retrospective study, MTX had similar effects in inducing and maintaining remission and achieving mucosal healing compared to AZT/6-MP in CD patients [74]. By contrast, MTX monotherapy is not recommended for maintaining remission in UC patients [75]. In one study, corticosteroid-free remission did not improve in an infliximab (IFX) and MTX combination group compared to an IFX monotherapy group, but MTX lowered the immunogenicity of IFX, which reflects the long-term durability of IFX [76]. Combination therapy with IFX and MTX therapy is recommended to reduce immunogenicity in IBD patients [75]. The American College of Gastroenterology guidelines suggest 15 to 25 mg once weekly parenteral MTX treatment as monotherapy and 12.5 to 15 mg oral MTX once weekly as combination therapy for CD patients [77]. In addition, MTX is superior to AZT/6-MP in that it is administered once a week, the dosing effect appears more quickly, and the incidence of tumors is low. Therefore, adolescent or young male patients and those with a homozygous mutation in NUDT15 should receive MTX therapy. The adverse events of MTX include nausea, vomiting, diarrhea, hepatotoxicity, and cytopenia. Concomitant administration of folate of at least 5 mg per week (1 mg daily) helps reduce the side effects on the digestive system. MTX should not be used in women planning to become pregnant, because it may cause teratogenesis. It must be discontinued for 3 to 6 months before planning pregnancy.
Due to the increased infection risk with immunomodulators, IBD patients should undergo a screening test for hepatitis B virus (HBV), hepatitis C virus (HCV), human immunodeficiency virus (HIV), and active or latent tuberculosis (Table 2) [24]. Vaccination history should be checked, as live vaccinations are contraindicated during immunosuppressive therapy [24]. Live vaccinations should be administered at least 4 weeks before the start of immunomodulators or 3 to 6 months after stopping the immunomodulators.

ANTI-TNF-α AGENTS

Indications of anti-TNF-α agents

The development of anti-TNF-α agents in 1998 for CD and 2005 for UC changed the paradigm of medical treatment for moderate to severe IBD patients [78]. Anti-TNF-α agents are recommended for induction and maintenance remission in moderate to severe active IBD patients who are refractory to corticosteroids and/or immunomodulators or dependent on corticosteroids [78]. Anti-TNF-α agents are also effective for treating complex perianal fistulizing CD and preventing the postoperative recurrence of CD [79,80]. However, anti-TNF-α agents are not indicated for patients with an intra-abdominal abscess and/or a fibrotic stricture, and surgical resection is preferred.

Utilization of anti-TNF-α agents

IFX (Remicade, Janssen, Seoul, Korea; Remsima, Celltrion, Incheon, Korea; and Remaloce, SAMSUNG BIOEPIS, Incheon, Korea) is a chimeric monoclonal antibody biologic drug that is infused intravenously at 5 mg/kg over 2 hours (shortened to 1 hour if well tolerated) at 0, 2, and 6 weeks and every 8 weeks thereafter in IBD patients (Table 3) [81,82]. A subcutaneous formulation of the IFX biosimilars, CT-P13 (Remsima) or SB2 (Remaloce), is now available for IBD patients. Subcutaneous CT-P13 has comparable efficacy, safety, and immunogenicity as intravenous CT-P13 [83]. Adalimumab, a completely humanized immunoglobulin G1 monoclonal anti-TNF-α antibody, is administered subcutaneously at an induction dose of 160 mg, followed by 80 mg 2 weeks later, and a maintenance dose of 40 mg every other week for IBD patients [84,85]. The adalimumab biosimilars, SB5 (Adalloce, SAMSUNG BIOEPIS) and CT-P17 (Yuflyma, Celltrion Korea) are now available for IBD patients. Golimumab is generated from genetically engineered mice immunized with human TNF and is administered subcutaneously at an induction dose of 200 mg, followed by 100 mg 2 weeks later, and a maintenance schedule of 100 mg every 4 weeks for UC patients [86].

Adverse events of anti-TNF-α agents

Anti-TNF-α agents are associated with an increase in adverse infectious events in IBD patients [87]. Screening of latent tuberculosis, HBV, HCV, and HIV before beginning an anti-TNF-α agent and preventing pneumocystis jirovecii or herpes zoster infection has benefits in patients treated with an anti-TNF-α agent (Table 2) [8789]. Routine screening of latent tuberculosis with either an interferon-gamma release assay test or the tuberculin skin test combined with a chest X-ray is mandatory for candidates taking an anti-TNF-α agent. In addition, serological screening with hepatitis B surface antigen and the antibody to hepatitis B surface, and the core (anti-HBc) protein should be performed before immunosuppressive therapy. Anti-TNF-α agents are associated with increased risk for hepato-splenic T cell lymphoma and melanoma [90,91]. The immediate infusion reaction during the anti-TNF-α agent course can cause pruritus, rash, headache, chest discomfort, and anaphylaxis. A late infusion reaction 1 to 3 weeks after administration of an anti-TNF-α agent manifests as arthralgia, joint stiffness, and fever. These infusion reactions occur in 3.5% to 38.6% of cases and can be prevented using antipyretics, antihistamines, and/or corticosteroids [92].

Optimizing anti-TNF-α agents

Although anti-TNF-α agents have advanced the medical treatment of IBD, 10% to 30% of patients experience a primary non-response and do not respond to induction therapy; 23% to 46% of patients eventually experience a loss of response to maintenance therapy [93,94]. TDM is efficacious for optimizing IBD therapy [95]. A recent consensus statement suggested that the IFX trough concentration during week 14 and the maintenance period should be > 3 μg/mL and that the adalimumab trough concentration during week 4 and the maintenance period should be > 5 μg/mL [96]. In the presence of adequate trough drug concentrations, anti-drug antibodies (ADA) are unlikely to be clinically relevant [97]. In the absence of a detectable biologic drug concentration, patients with undetectable/low ADA can optimize drug therapy by combining them with immunomodulators, shortening the dosing interval, and increasing the drug dose. Patients with high ADA levels should consider switching drug classes [97]. Although reactive TDM performed in response to active inflammation based on biochemical, endoscopic, or radiological assessments is recommended, proactive TDM performed in patients regardless of clinical status is currently under active investigation to demonstrate its superiority [98]. Before changing biologic agents, it is necessary to carefully consider the need for surgery, the existence of consistent infections, or the possibility of combined IBD.

CONCLUSIONS

Conventional therapies, including 5-ASA-based drugs, corticosteroids, immunomodulators, and anti-TNF-α agents are the keystone of medical treatment for IBD. New formulations and delivery systems are being developed for 5-ASA-based drugs to improve patient adherence and convenience. Corticosteroids and second-generation corticosteroids have important roles in inducing clinical remission in moderate to severe IBD patients. To minimize the inappropriate use of corticosteroids, optimal timing and dosing of the immunomodulator is required to maintain remission. The NUDT15 gene test and TDM monitoring have helped reduce adverse events and maximize the efficacy of immunomodulators. Anti-TNF-α agents have changed the standard of treating refractory moderate to severe IBD patients. Combination therapy with an anti-TNF-α agent and an immunomodulator helps overcome immunogenicity in IBD patients.

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Table 1
5-ASA preparations
Preparation Drug Formulation Release site
Azo-bonded prodrugs Sulfasalazine (Salazopyrin) Sulfapyridine+5-ASA Colon
Olsalazine (Dipentum) 5-ASA+5-ASA Colon
Basalazide (Colazal) 4-amino-benzoyl-β-alanine+5-ASA Colon
pH-dependent drugs Mesalamine (Asacol) 5-ASA coated with Eudragit S100 Terminal ileum and colon
Mesalamine (Salofalk) 5-ASA coated with Eudragit L100 Distal ileum and colon
Mesalamine (Mezavant) MMX system coated combination with Eudragit S100 and Eudragit L100 Terminal ileum and colon
Time-dependent drugs Mesalamine (Pentasa) 5-ASA coated with a semipermeable membrane of ethycellulose Duodenum, small bowel, and colon

5-ASA, 5-aminosalicylate.

Table 2
Checklist for patient screening for immunomodulators and/or anti-TNF-α agents
General considerations Checkpoint
Contraindications to anti-TNF-α agents Grade 3, 4 heart failure
Previous lymphoma or current malignancy history
Demyelinating disease
History of recurrent infection
Precautions and screening Tuberculosis: tuberculin skin test, chest x ray, interferon gamma release assay
HBV: HBsAg, anti-HBc, anti-HBs
HCV: anti-HCV
HIV: antigen/antibody HIV-1/2 immunoassy
VZV: IgM/IgG anti-VZV
Vaccination status Live vaccination (MMR, herpes zoster, BCG, varicella): contraindicated
Inactive vaccination (COVID-19, DTP, HAV, HPV, influenza, pneumococcus): safe

TNF-α, tumor necrosis factor-α; HBV, hepatitis B virus; HBsAg, hepatitis B surface antigen; anti-HBc, anti-hepatitis B core antibody; anti-HBs, anti-hepatitis B surface antibody; HCV, hepatitis C virus; HIV, human immunodeficiency virus; VZV, varicella zoster virus; IgM, immunoglobulin M; MMR, measles, mumps and rubella; BCG, Bacillus Calmette–Guérin; COVID-19, coronavirus disease 2019; DTP, Diphtheria-Tetanus-Pertussis; HAV, hepatitis A; HPV, human papillomavirus.

Table 3
Anti-TNF-α agents for inflammatory bowel disease
Preparation Drug Route of administration Target disease Induction dose Maintenance dose
Infliximab Remicade Intravenous UC, CD 5 mg/kg at weeks 0, 2, and 6 5 mg/kg every 8 weeks
Remsima (CT-P13) Intravenous UC, CD 5 mg/kg at weeks 0, 2, and 6 5 mg/kg every 8 weeks
Subcutaneous UC, CD 5 mg/kg at weeks 0, 2 (IV)
120 mg at week 6 (SC)
120 mg every 2 weeks
Remaloce (SB2) Intravenous UC, CD 5 mg/kg at weeks 0, 2, and 6 5 mg/kg every 8 weeks
Adalimumab Humira Subcutaneous (citrate-free formulation) UC, CD 160 mg at week 0
80 mg at week 2
40 mg at week 4
40 mg every other week
Adalloce (SB5) Subcutaneous UC, CD 160 mg at week 0
80 mg at week 2
40 mg at week 4
40 mg every other week
Yuflyma (CT-P17) Subcutaneous (citrate-free formulation) UC, CD 160 mg at week 0
80 mg at week 2
40 mg at week 4
40 mg every other week
Golimumab Simponi Subcutaneous UC 200 mg at week 0
100 mg at week 2
50 mg at week 6 (100 mg if weight > 80 kg)
50 mg (or 100 mg) every 4 weeks

TNF-α, tumor necrosis factor-α; UC, ulcerative colitis; CD, Crohn’s disease; IV, intravenous; SC, subcutaneous.

REFERENCES

1. Song EM, Yang SK. Natural history of inflammatory bowel disease: a comparison between the East and the West. Intest Res 2021. Dec. 2. [Epub]. https://doi.org/10.5217/ir.2021.00104 .
crossref pmid
2. Bryant RV, Brain O, Travis SP. Conventional drug therapy for inflammatory bowel disease. Scand J Gastroenterol 2015;50:90–112.
crossref pmid
3. Park J, Park S, Lee SA, Park SJ, Cheon JH. Improving the care of inflammatory bowel disease (IBD) patients: perspectives and strategies for IBD center management. Korean J Intern Med 2021;36:1040–1048.
crossref pmid pmc pdf
4. Yoon JS, Lee SJ, Kim ES, Kim SK, Jung MK, Lee HS, et al. Quality of information on the Internet for Korean patients with inflammatory bowel disease. Korean J Intern Med 2019;34:1215–1222.
crossref pmid pmc pdf
5. Turner D, Ricciuto A, Lewis A, et al. STRIDE-II: an update on the Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE) Initiative of the International Organization for the Study of IBD (IOIBD): determining therapeutic goals for treat-to-target strategies in IBD. Gastroenterology 2021;160:1570–1583.
crossref pmid
6. Im JP, Ye BD, Kim YS, Kim JS. Changing treatment paradigms for the management of inflammatory bowel disease. Korean J Intern Med 2018;33:28–35.
crossref pmid pmc pdf
7. Sood A, Ahuja V, Midha V, et al. Colitis and Crohn’s Foundation (India) consensus statements on use of 5-aminosalicylic acid in inflammatory bowel disease. Intest Res 2020;18:355–378.
crossref pmid pmc pdf
8. Plosker GL, Croom KF. Sulfasalazine: a review of its use in the management of rheumatoid arthritis. Drugs 2005;65:1825–1849.
crossref pmid
9. Di Paolo MC, Paoluzi OA, Pica R, et al. Sulphasalazine and 5-aminosalicylic acid in long-term treatment of ulcerative colitis: report on tolerance and side-effects. Dig Liver Dis 2001;33:563–569.
crossref pmid
10. Mansfield JC, Giaffer MH, Cann PA, McKenna D, Thornton PC, Holdsworth CD. A double-blind comparison of balsalazide, 6.75 g, and sulfasalazine, 3 g, as sole therapy in the management of ulcerative colitis. Aliment Pharmacol Ther 2002;16:69–77.
crossref pmid pdf
11. Nielsen OH, Munck LK. Drug insight: aminosalicylates for the treatment of IBD. Nat Clin Pract Gastroenterol Hepatol 2007;4:160–170.
crossref pmid pdf
12. Berends SE, Strik AS, Lowenberg M, D’Haens GR, Mathot RA. Clinical pharmacokinetic and pharmacodynamic considerations in the treatment of ulcerative colitis. Clin Pharmacokinet 2019;58:15–37.
crossref pmid pmc pdf
13. Shimodaira Y, Onochi K, Watanabe K, et al. Effect of acid-reducing agents on clinical relapse in ulcerative colitis with pH-dependent-released 5-aminosalicylic acid: a multicenter retrospective study in Japan. Intest Res 2021;19:225–231.
crossref pmid pmc pdf
14. Thakral S, Thakral NK, Majumdar DK. Eudragit: a technology evaluation. Expert Opin Drug Deliv 2013;10:131–149.
crossref pmid
15. Varum F, Freire AC, Bravo R, Basit AW. OPTICORE™, an innovative and accurate colonic targeting technology. Int J Pharm 2020;583:119372.
crossref pmid
16. Nardelli S, Pisani LF, Tontini GE, Vecchi M, Pastorelli L. MMX® technology and its applications in gastrointestinal diseases. Therap Adv Gastroenterol 2017;10:545–552.
crossref pmid pmc pdf
17. Sonu I, Lin MV, Blonski W, Lichtenstein GR. Clinical pharmacology of 5-ASA compounds in inflammatory bowel disease. Gastroenterol Clin North Am 2010;39:559–599.
crossref pmid
18. Tamura S, Ishida N, Miyazu T, et al. Mesalazine granule formulation improves clinical data in Crohn’s disease compared with tablet formulation. Sci Rep 2020;10:21353.
crossref pmid pmc pdf
19. Veloso PM, Machado R, Nobre C. Mesalazine and inflammatory bowel disease: from well-established therapies to progress beyond the state of the art. Eur J Pharm Biopharm 2021;167:89–103.
crossref pmid
20. Peskar BM, Dreyling KW, May B, Schaarschmidt K, Goebell H. Possible mode of action of 5-aminosalicylic acid. Dig Dis Sci 1987;32(12 Suppl):51S–56S.
crossref pmid pdf
21. Wahl C, Liptay S, Adler G, Schmid RM. Sulfasalazine: a potent and specific inhibitor of nuclear factor kappa B. J Clin Invest 1998;101:1163–1174.
crossref pmid pmc
22. Rousseaux C, Lefebvre B, Dubuquoy L, et al. Intestinal anti-inflammatory effect of 5-aminosalicylic acid is dependent on peroxisome proliferator-activated receptor-gamma. J Exp Med 2005;201:1205–1215.
pmid pmc
23. Couto D, Ribeiro D, Freitas M, Gomes A, Lima JL, Fernandes E. Scavenging of reactive oxygen and nitrogen species by the prodrug sulfasalazine and its metabolites 5-aminosalicylic acid and sulfapyridine. Redox Rep 2010;15:259–267.
crossref pmid pmc
24. 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(Suppl 3):s1–s106.
crossref pmid pmc
25. Rubin DT, Ananthakrishnan AN, Siegel CA, Sauer BG, Long MD. ACG clinical guideline: ulcerative colitis in adults. Am J Gastroenterol 2019;114:384–413.
crossref pmid
26. Choi CH, Moon W, Kim YS, et al. Second Korean guideline for the management of ulcerative colitis. Korean J Gastroenterol 2017;69:1–28.
crossref pmid pdf
27. Sandborn WJ, Regula J, Feagan BG, et al. Delayed-release oral mesalamine 4.8 g/day (800-mg tablet) is effective for patients with moderately active ulcerative colitis. Gastroenterology 2009;137:1934–1943.
crossref pmid
28. Lichtenstein GR, Ramsey D, Rubin DT. Randomised clinical trial: delayed-release oral mesalazine 4.8 g/day vs. 2.4 g/day in endoscopic mucosal healing: ASCEND I and II combined analysis. Aliment Pharmacol Ther 2011;33:672–678.
crossref pmid
29. Orchard TR, van der Geest SA, Travis SP. Randomised clinical trial: early assessment after 2 weeks of high-dose mesalazine for moderately active ulcerative colitis: new light on a familiar question. Aliment Pharmacol Ther 2011;33:1028–1035.
crossref pmid
30. Ford AC, Khan KJ, Achkar JP, Moayyedi P. Efficacy of oral vs. topical, or combined oral and topical 5-aminosalicylates, in Ulcerative Colitis: systematic review and meta-analysis. Am J Gastroenterol 2012;107:167–176.
crossref pmid pdf
31. Osterman MT, Aberra FN, Cross R, et al. Mesalamine dose escalation reduces fecal calprotectin in patients with quiescent ulcerative colitis. Clin Gastroenterol Hepatol 2014;12:1887–1893.
crossref pmid pmc
32. d’Albasio G, Pacini F, Camarri E, et al. Combined therapy with 5-aminosalicylic acid tablets and enemas for maintaining remission in ulcerative colitis: a randomized double-blind study. Am J Gastroenterol 1997;92:1143–1147.
pmid
33. Feagan BG, Macdonald JK. Oral 5-aminosalicylic acid for induction of remission in ulcerative colitis. Cochrane Database Syst Rev 2012;10:CD000543.
crossref pmid
34. Feagan BG, MacDonald JK. Once daily oral mesalamine compared to conventional dosing for induction and maintenance of remission in ulcerative colitis: a systematic review and meta-analysis. Inflamm Bowel Dis 2012;18:1785–1794.
crossref pmid
35. Kruis W, Kiudelis G, Racz I, et al. Once daily versus three times daily mesalazine granules in active ulcerative colitis: a double-blind, double-dummy, randomised, non-inferiority trial. Gut 2009;58:233–240.
crossref pmid pmc
36. Tripathi K, Dong J, Mishkin BF, Feuerstein JD. Patient preference and adherence to aminosalicylates for the treatment of ulcerative colitis. Clin Exp Gastroenterol 2021;14:343–351.
crossref pmid pmc pdf
37. Mizuno S, Ono K, Mikami Y, et al. 5-Aminosalicylic acid intolerance is associated with a risk of adverse clinical outcomes and dysbiosis in patients with ulcerative colitis. Intest Res 2020;18:69–78.
crossref pmid pmc pdf
38. Lim WC, Wang Y, MacDonald JK, Hanauer S. Aminosalicylates for induction of remission or response in Crohn’s disease. Cochrane Database Syst Rev 2016;7:CD008870.
crossref pmid pmc
39. Akobeng AK, Zhang D, Gordon M, MacDonald JK. Oral 5-aminosalicylic acid for maintenance of medically-induced remission in Crohn’s disease. Cochrane Database Syst Rev 2016;9:CD003715.
crossref pmid pmc
40. Honap S, Sharma E, Samaan MA. 5-ASAs in Crohn’s disease: time to stop the salicylate? Dig Dis Sci 2022;67:2699–2700.
crossref pmid pdf
41. Coward S, Kuenzig ME, Hazlewood G, et al. Comparative effectiveness of mesalamine, sulfasalazine, corticosteroids, and budesonide for the induction of remission in Crohn’s disease: a Bayesian network meta-analysis: republished. Inflamm Bowel Dis 2017;23:E26–E37.
pmid
42. Bernstein CN, Tenakoon A, Singh H, Targownik LE. Continued 5ASA use after initiation of anti-TNF or immunomodulator confers no benefit in IBD: a population-based study. Aliment Pharmacol Ther 2021;54:814–832.
crossref pmid pdf
43. Xie C, Quan R, Hong F, Zou K, Yan W, Fu Y. The culprit of mesalamine intolerance: case series and literature review. BMC Gastroenterol 2019;19:138.
crossref pmid pmc pdf
44. Tsujii Y, Nishida T, Osugi N, et al. Classification and clinical features of adverse drug reactions in patients with ulcerative colitis treated with 5-aminosalicylate acid: a single-center, observational study. Scand J Gastroenterol 2022;57:190–196.
crossref pmid
45. Wild GE, Waschke KA, Bitton A, Thomson AB. The mechanisms of prednisone inhibition of inflammation in Crohn’s disease involve changes in intestinal permeability, mucosal TNFalpha production and nuclear factor kappa B expression. Aliment Pharmacol Ther 2003;18:309–317.
crossref pmid pdf
46. Baron JH, Connell AM, Kanaghinis TG, Lennard-Jones JE, Jones AF. Out-patient treatment of ulcerative colitis: comparison between three doses of oral prednisone. Br Med J 1962;2:441–443.
pmid pmc
47. Waljee AK, Wiitala WL, Govani S, et al. Corticosteroid use and complications in a US inflammatory bowel disease cohort. PLoS One 2016;11:e0158017.
crossref pmid pmc
48. Buchman AL. Side effects of corticosteroid therapy. J Clin Gastroenterol 2001;33:289–294.
crossref pmid
49. Maconi G, Camatta D, Cannatelli R, et al. Budesonide MMX in the treatment of ulcerative colitis: current perspectives on efficacy and safety. Ther Clin Risk Manag 2021;17:285–292.
crossref pmid pmc pdf
50. Hibi T, Naganuma M, Oda E, et al. Predictive factors for achievement of mucosal healing by budesonide 2-mg foam in ulcerative colitis: a pooled analysis of data from two clinical trials. Intest Res 2020;18:56–68.
crossref pmid pmc pdf
51. Van Assche G, Manguso F, Zibellini M, et al. Oral prolonged release beclomethasone dipropionate and prednisone in the treatment of active ulcerative colitis: results from a double-blind, randomized, parallel group study. Am J Gastroenterol 2015;110:708–715.
crossref pmid pdf
52. Lee YJ, Cheon JH, Kim JH, et al. Clinical efficacy of beclomethasone dipropionate in Korean patients with ulcerative colitis. Yonsei Med J 2017;58:144–149.
crossref pmid pmc pdf
53. Lie MR, Kreijne JE, Dijkstra G, et al. No superiority of tacrolimus suppositories vs beclomethasone suppositories in a randomized trial of patients with refractory ulcerative proctitis. Clin Gastroenterol Hepatol 2020;18:1777–1784.
crossref pmid
54. Caron B, Sandborn WJ, Panaccione R, et al. Efficacy of pharmacological agents for ulcerative proctitis: a systematic literature review. J Crohns Colitis 2022;16:922–930.
crossref pmid pdf
55. Nunes T, Barreiro-de Acosta M, Marin-Jimenez I, Nos P, Sans M. Oral locally active steroids in inflammatory bowel disease. J Crohns Colitis 2013;7:183–191.
crossref pmid
56. Laine L, Hanauer SB. Considerations in the management of steroid-dependent Crohn’s disease. Gastroenterology 2003;125:906–910.
crossref pmid
57. Velayos F, Mahadevan U. Management of steroid-dependent ulcerative colitis: immunomodulatory agents, biologics, or surgery? Clin Gastroenterol Hepatol 2007;5:668–671.
crossref pmid
58. Singh A, Mahajan R, Kedia S, et al. Use of thiopurines in inflammatory bowel disease: an update. Intest Res 2022;20:11–30.
crossref pmid pmc pdf
59. Fraser AG, Orchard TR, Jewell DP. The efficacy of azathioprine for the treatment of inflammatory bowel disease: a 30 year review. Gut 2002;50:485–489.
crossref pmid pmc
60. Stournaras E, Qian W, Pappas A, et al. Thiopurine monotherapy is effective in ulcerative colitis but significantly less so in Crohn’s disease: long-term outcomes for 11 928 patients in the UK inflammatory bowel disease bioresource. Gut 2021;70:677–686.
crossref pmid pmc
61. Xu Z, Davis HM, Zhou H. Clinical impact of concomitant immunomodulators on biologic therapy: pharmacokinetics, immunogenicity, efficacy and safety. J Clin Pharmacol 2015;55:Suppl 3. S60–S74.
crossref pmid
62. Smith MA, Blaker P, Marinaki AM, Anderson SH, Irving PM, Sanderson JD. Optimising outcome on thiopurines in inflammatory bowel disease by co-prescription of allopurinol. J Crohns Colitis 2012;6:905–912.
crossref pmid
63. Louis E, Irving P, Beaugerie L. Use of azathioprine in IBD: modern aspects of an old drug. Gut 2014;63:1695–1699.
crossref pmid
64. Gilissen LP, Wong DR, Engels LG, et al. Therapeutic drug monitoring of thiopurine metabolites in adult thiopurine tolerant IBD patients on maintenance therapy. J Crohns Colitis 2012;6:698–707.
crossref pmid
65. Crouwel F, Buiter HJC, de Boer NK. There is still a place for optimised thiopurine therapy in IBD. Gut 2021;70:2207.
crossref pmid
66. Chaparro M, Ordas I, Cabre E, et al. Safety of thiopurine therapy in inflammatory bowel disease: long-term follow-up study of 3931 patients. Inflamm Bowel Dis 2013;19:1404–1410.
pmid
67. Nagy F, Molnar T, Szepes Z, Farkas K, Nyari T, Lonovics J. Efficacy of 6-mercaptopurine treatment after azathioprine hypersensitivity in inflammatory bowel disease. World J Gastroenterol 2008;14:4342–4346.
crossref pmid pmc
68. Lees CW, Maan AK, Hansoti B, Satsangi J, Arnott ID. Tolerability and safety of mercaptopurine in azathioprine-intolerant patients with inflammatory bowel disease. Aliment Pharmacol Ther 2008;27:220–227.
crossref pmid
69. Matsuoka K. NUDT15 gene variants and thiopurine-induced leukopenia in patients with inflammatory bowel disease. Intest Res 2020;18:275–281.
crossref pmid pmc pdf
70. Chang JY, Park SJ, Jung ES, et al. Genotype-based treatment with thiopurine reduces incidence of myelosuppression in patients with inflammatory bowel diseases. Clin Gastroenterol Hepatol 2020;18:2010–2018.
crossref pmid
71. Derijks LJ, Gilissen LP, Hooymans PM, Hommes DW. Review article: thiopurines in inflammatory bowel disease. Aliment Pharmacol Ther 2006;24:715–729.
crossref pmid
72. Maeda T, Sakuraba H, Hiraga H, et al. Long-term efficacy and tolerability of dose-adjusted thiopurine treatment in maintaining remission in inflammatory bowel disease patients with NUDT15 heterozygosity. Intest Res 2022;20:90–100.
crossref pmid pmc pdf
73. Haisma SM, Lijftogt T, Kindermann A, et al. Methotrexate for maintaining remission in paediatric Crohn’s patients with prior failure or intolerance to thiopurines: a multicenter cohort study. J Crohns Colitis 2015;9:305–311.
crossref pmid
74. Huang Z, Chao K, Li M, et al. Methotrexate for refractory Crohn’s disease compared with thiopurines: a retrospective non-head-to-head controlled study. Inflamm Bowel Dis 2017;23:440–447.
pmid
75. Feuerstein JD, Isaacs KL, Schneider Y, et al. AGA clinical practice guidelines on the management of moderate to severe ulcerative colitis. Gastroenterology 2020;158:1450–1461.
crossref pmid pmc
76. Feagan BG, McDonald JW, Panaccione R, et al. Methotrexate in combination with infliximab is no more effective than infliximab alone in patients with Crohn’s disease. Gastroenterology 2014;146:681–688.
crossref pmid
77. Lichtenstein GR, Loftus EV, Isaacs KL, Regueiro MD, Gerson LB, Sands BE. ACG clinical guideline: management of Crohn’s disease in adults. Am J Gastroenterol 2018;113:481–517.
crossref pmid
78. Vulliemoz M, Brand S, Juillerat P, et al. TNF-alpha blockers in inflammatory bowel diseases: practical recommendations and a user’s guide: an update. Digestion 2020;101:Suppl 1. 16–26.
crossref pmid pdf
79. Gecse K, Khanna R, Stoker J, et al. Fistulizing Crohn’s disease: diagnosis and management. United European Gastroenterol J 2013;1:206–213.
crossref pmid pmc pdf
80. Vaughn BP, Moss AC. Prevention of post-operative recurrence of Crohn’s disease. World J Gastroenterol 2014;20:1147–1154.
crossref pmid pmc
81. Oh SJ, Shin GY, Soh H, et al. Long-term outcomes of infliximab in a real-world multicenter cohort of patients with acute severe ulcerative colitis. Intest Res 2021;19:323–331.
crossref pmid pmc pdf
82. Jung YS, Han M, Park S, Cheon JH. Impact of early anti-TNF use on clinical outcomes in Crohn’s disease: a nationwide population-based study. Korean J Intern Med 2020;35:1104–1113.
crossref pmid pmc pdf
83. Schreiber S, Ben-Horin S, Leszczyszyn J, et al. Randomized controlled trial: subcutaneous vs intravenous infliximab CT-P13 maintenance in inflammatory bowel disease. Gastroenterology 2021;160:2340–2353.
crossref pmid
84. Ogata H, Hagiwara T, Kawaberi T, Kobayashi M, Hibi T. Safety and effectiveness of adalimumab in the treatment of ulcerative colitis: results from a large-scale, prospective, multicenter, observational study. Intest Res 2021;19:419–429.
crossref pmid pmc pdf
85. Hisamatsu T, Suzuki Y, Kobayashi M, et al. Long-term safety and effectiveness of adalimumab in Japanese patients with Crohn’s disease: 3-year results from a real-world study. Intest Res 2021;19:408–418.
crossref pmid pmc pdf
86. Yu J, Park SJ, Kim HW, et al. Effectiveness and safety of golimumab in patients with ulcerative colitis: a multicenter, prospective, postmarketing surveillance study. Gut Liver 2021. Dec. 27. [Epub]. https://doi.org/10.5009/gnl210335 .
crossref pmid
87. Murdaca G, Spano F, Contatore M, et al. Infection risk associated with anti-TNF-α agents: a review. Expert Opin Drug Saf 2015;14:571–582.
crossref pmid
88. Alrajhi S, Germain P, Martel M, et al. Concordance between tuberculin skin test and interferon-gamma release assay for latent tuberculosis screening in inflammatory bowel disease. Intest Res 2020;18:306–314.
crossref pmid pmc pdf
89. Hong HS, Jung J, Park SH, et al. Seroprevalence of viral infectious diseases and associated factors in Korean patients with inflammatory bowel diseases. Korean J Intern Med 2022;37:73–84.
crossref pmid pmc pdf
90. Annese V, Beaugerie L, Egan L, et al. European Evidence-based Consensus: inflammatory bowel disease and malignancies. J Crohns Colitis 2015;9:945–965.
crossref pmid
91. Phan H, Weideman RA, Cipher DJ, Feagins LA. Safety of tumor necrosis factor inhibitor use in patients with concomitant malignancy. Intest Res 2020;18:282–288.
crossref pmid pmc pdf
92. Jacobstein DA, Markowitz JE, Kirschner BS, et al. Premedication and infusion reactions with infliximab: results from a pediatric inflammatory bowel disease consortium. Inflamm Bowel Dis 2005;11:442–446.
crossref pmid
93. Roda G, Jharap B, Neeraj N, Colombel JF. Loss of response to anti-TNFs: definition, epidemiology, and management. Clin Transl Gastroenterol 2016;7:e135.
crossref pmid pmc pdf
94. Hong SW, Park J, Yoon H, et al. Comparison of loss of response between anti-tumor necrosis factor alone and combined use with immunomodulators in patients with inflammatory bowel disease. Korean J Intern Med 2021;36(Suppl 1):S9–S17.
crossref pmid pmc pdf
95. Wu JF. Therapeutic drug monitoring of biologics for patients with inflammatory bowel diseases: how, when, and for whom? Gut Liver 2022;16:515–524.
crossref pmid pmc
96. Papamichael K, Cheifetz AS, Melmed GY, et al. Appropriate therapeutic drug monitoring of biologic agents for patients with inflammatory bowel diseases. Clin Gastroenterol Hepatol 2019;17:1655–1668.
crossref pmid pmc
97. Papamichael K, Cheifetz AS. Therapeutic drug monitoring in inflammatory bowel disease: for every patient and every drug? Curr Opin Gastroenterol 2019;35:302–310.
pmid pmc
98. Argollo M, Kotze PG, Kakkadasam P, D’Haens G. Optimizing biologic therapy in IBD: how essential is therapeutic drug monitoring? Nat Rev Gastroenterol Hepatol 2020;17:702–710.
crossref pmid pdf
TOOLS
METRICS Graph View
  • 3 Web of Science
  • 5 Crossref
  • 4 Scopus
  • 2,023 View
  • 283 Download
Related articles

Editorial Office
101-2501, Lotte Castle President, 109 Mapo-daero, Mapo-gu, Seoul 04146, Korea
Tel: +82-2-2271-6792    Fax: +82-2-790-0993    E-mail: kaim@kams.or.kr                

Copyright © 2024 by Korean Association of Internal Medicine.

Close layer
prev next