eISSN: 1644-4124
ISSN: 1426-3912
Central European Journal of Immunology
Current issue Archive Manuscripts accepted About the journal Special Issues Editorial board Abstracting and indexing Subscription Contact Instructions for authors
Editorial System
Submit your Manuscript
SCImago Journal & Country Rank
4/2023
vol. 48
 
Share:
Share:
Experimental immunology

Efficacy of IL-23 inhibitors and IL-12/23 inhibitors in the induction treatment of Crohn’s disease: A meta-analysis based on randomized controlled trials

Boyang Gao
1
,
Haojie Shentu
2
,
Suyong Sha
3
,
Dongying Wang
4
,
Xi Chen
2
,
Zhenwei Huang
2
,
Nan Dong
2
,
Haijia Lai
2
,
Jianying Xu
2
,
Xiaoshuai Zhou
5

1.
Wenzhou Medical University, Wenzhou, Zhejiang, China
2.
The Medical Imaging College, Hangzhou Medical College, Hangzhou, Zhejiang, China
3.
Emergency Medical Center, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, China
4.
The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
5.
Department of Anus & Intestine Surgery, Ningbo Yinzhou No. 2 Hospital, Ningbo, Zhejiang, China
Cent Eur J Immunol 2023; 48 (4): 301-310
Online publish date: 2024/01/09
Article files
- Efficacy of IL-23.pdf  [1.28 MB]
Get citation
 
PlumX metrics:
 

Introduction

Crohn’s disease (CD) is a type of inflammatory bowel disease (IBD) characterized by granulomatous, chronically recurrent gastrointestinal inflammation that tends to occur in the terminal ileum and colon [1, 2]. Over time, nearly half of patients develop complications (strictures, perianal disease, fistula or abscess), often resulting in surgical interventions [3, 4].

Crohn’s disease occurs more frequently in developed countries in North America and Western Europe [5]. Since the 21st century, the incidence of CD has gradually increased globally, especially in some emerging industrialized countries in Asia, South America, and Africa [6]. Currently, the goal of treating CD is to induce remission in the short term and maintain it in the long term. Pharmacological treatments include corticosteroids, thiopurine, methotrexate, and biological therapies. The introduction of biologics has brought considerable efficacy to the management of CD, along with a better safety profile than corticosteroids and immunosuppressants/immunosuppressive agents [2, 7]. However, some inhibitors, such as tumor necrosis factor (TNF) inhibitors, still have drawbacks in the treatment of CD, primarily non-response, loss of secondary reaction, potential serious adverse events (SAEs), and treatment-related costs [8-10].

The IL--23 pathway has been implicated in the pathogenesis of several immune-mediated chronic inflammatory diseases including psoriasis and CD [11, 12]. Also, biologics such as IL-23 inhibitors and IL-12/23 inhibitors have been shown to be effective in the treatment of patients with CD in randomized controlled trials (RCTs), both in the induction period and maintenance period. With the continuous emergence of the IL-23 inhibitors, there has not been a comprehensive analysis of the research in this area. In the present study, the efficacy of IL-23 and IL-12/23 inhibitors in the induction phase for the treatment of CD was further evaluated through a meta-analysis based on published randomized controlled trials.

Material and methods

Search strategy

This study was carried out according to the latest Preferred Reporting Items for Systematic review and Meta- Analysis (PRISMA) 2020 guidelines [13]. A comprehensive systematic search of the literature was conducted from inception until December, 2022, using Medline, Embase, Web of Science and the Cochrane Library. Any potential RCTs relevant to the efficacy of IL-23 inhibitors or IL-12/23 inhibitors in the treatment of CD would be adopted. The search algorithms were as the follows: ((Interleukin-23) OR (Interleukin 23) OR (IL-23) OR (IL 23) OR (Interleukin-12) OR (Interleukin 12) OR (IL-12) OR (IL 12)) AND ((Crohn’s Enteritis) OR (Crohn Disease) OR (Regional Enteritis) OR (Crohn’s Disease) OR (Crohns Disease) OR (Inflammatory Bowel Disease 1) OR (Enteritis, Granulomatous) OR (Granulomatous Enteritis) OR (Colitis, Granulomatous) OR (Granulomatous Colitis)). The literature search was conducted independently by two experienced researchers in systematic reviews.

Study selection

The study selection was based on the PICOS (participants, intervention, comparison, outcomes and study design) principle. Studies that met the following criteria were considered eligible: 1) participants: patients aged 18 years or older who were suffering from CD; 2) intervention: IL-23 inhibitors or IL-12/23 inhibitors (alone or combined with other therapies) in the induction period; 3) comparison: placebo and/or other non-biological treatments; 4) outcomes: improvement/normalization of symptoms (Crohn’s Disease Activity Index – CDAI) and/or biochemical markers and/or endoscopic findings; 5) study design: phase II/III RCTs.

The exclusion criteria were as follows: 1) studies limited to maintenance therapy; 2) the data were unavailable or missing; 3) articles not published in English; 4) studies including pediatric patients. For updated duplicated articles, the latest published or the studies with largest sample sizes were included in the study.

Data extraction

Two investigators independently completed the extraction according to the pre-specified formats, and discrepancies were ironed out through discussion. The following study characteristics were extracted: author, year of publication, country, trial phase, participants’ characteristics (e.g., age, gender, disease duration, biological therapy experience), number of patients, treatment regimen, follow-up duration and reported outcomes.

The primary outcome of the induction therapy was achievement of clinical remission (defined by a CDAI < 150). Secondary endpoints included achievement of clinical response (defined by a reduction in the CDAI ≥ 100 points compared to baseline), endoscopic remission, endoscopic response and normalization of C-reactive protein (CRP).

Quality assessment

The Cochrane Collaboration’s tool [14] was used to assess the risk of bias inherent in the included RCTs. The inherent risk of bias included random sequence generation (selection bias), allocation concealment (selection bias), blinding of participants and personnel (performance bias), blinding of outcome assessment (detection bias), incomplete outcome data (attrition bias), selective reporting (reporting bias) and other biases. Each aspect was scored as “low risk”, “high risk”, or “unclear risk”, with low overall risk indicating that the study was less prone to bias.

Statistical analysis

RevMan version 5.4 software was used in the statistical analysis. For dichotomous variables, the effect size was calculated using relative risk (RR) and the corresponding 95% confidence interval (CI). The heterogeneity between studies was evaluated using the I2 statistic: a value of 0% indicates no observed heterogeneity, and a larger value denotes increased heterogeneity [15]. Given the complexity of patient baseline characteristics and the use of different biological inhibitors, a random-effects model was used to analyze the results of the included studies. In order to increase the credibility of the results, publication bias (Egger’s weighted regression test [16]) and sensitivity analyses were performed since more than 10 studies were included. Moreover, a two-sided p-value < 0.05 for all statistical outcomes described above was considered statistically significant.

Results

Study selection

Based on our literature search, a total of 944 studies were identified. After excluding duplicates, 524 studies remained, 15 of which were considered potentially relevant and were read in full text. Among the 15 studies, 1 was excluded because the study protocol did not meet the inclusion criteria, 4 were excluded because no outcomes of interest were reported and 3 were excluded because the dates were unavailable. Finally, 7 studies containing 3,990 patients were included in the meta-analysis. The flowchart for the search procedure is displayed in Figure 1.

Fig. 1

Flow diagram describing inclusion and exclusion criteria

/f/fulltexts/CEJOI/52222/CEJI-48-52222-g001_min.jpg

Study characteristics and quality assessment

The characteristics of the selected RCTs are shown in Table 1. Of these 7 studies, 5 RCTs evaluated the induction of IL-23 inhibitors (including risankizumab, guselkumab, mirikizumab and brazikumab) [17-21], and 3 RCTs evaluated the induction of IL-12/23 inhibitors (ustekinumab only) [18, 22, 23]. Notably, one of the studies evaluated both IL-23 inhibitors and IL-12/23 inhibitors. All intervention groups were treated with a single IL-23 inhibitor or IL-12/23 inhibitor, while control groups were placebo-controlled only. Clinical remission and clinical response were reported in all studies. In addition, among the research of IL-23 induced therapy, 3 studies reported endoscopic remission rates, and 4 studies reported endoscopic response rates. The IL-23 inhibitors were assessed at week 12, while IL-12/23 inhibitors were assessed at week 8.

Table 1

Baseline characteristics of studies included in the meta-analysis

Author, yearTrial phaseNumber of patientsTherapeutic regimenInduction durationPrimary outcome definition
TreatmentControl
Feagan B.G., 201728239Risankizumab, 200 mg/600 mg at 0, 4, and 8 week12 weekClinical remission: CDAI < 150
Sands B.E., 201725960Brazikumab, 700 mg at 0 and 4 weeks12 weekClinical remission: CDAI
decrease of 100 points from baseline
or clinical remission [CDAI < 150]
D’Haens G., 20223675175Risankizumab, 600 mg/1200 mg at 0, 4, and 8 weeks12 weekClinical remission: CDAI < 150 and average daily liquid or very soft stool frequency ≤ 2.8, plus average daily abdominal pain score ≤ 1, and both not worse
than baseline
Endoscopic response: A greater than 50%
decrease
in SES-CD from baseline
D’Haens G., 20223382187Risankizumab, 600 mg/1200 mg at 0, 4, and 8 weeks12 week
Sands B.E., 2022212764Mirikizumab, 200 mg/600 mg/1000 mg
at 0, 4, and 8 weeks
12 weekEndoscopic response: 50% reduction from baseline
in SES-CD score
Sandborn W.J., 2022218561Guselkumab, 200 mg/600 mg/1200 mg
at 0, 4, and 8 weeks
12 weekThe change from baseline in
CDAI score: Undefined
Sandborn W.J., 20122394132Ustekinumab, 1/3/6 mg/kg at 0 weeks8 weekClinical response: ≤ 100-point decrease from the baseline CDAI
Feagan B.G., 20163494247Ustekinumab, 130 mg; 6 mg/kg at 0 weeks8 weekClinical response: Decrease from baseline in CDAI
of at least 100 points or a total CDAI
score less than 150
Feagan B.G., 20163419210Ustekinumab, 130 mg; 6 mg/kg at 0 weeks12 week
Sandborn W.J., 202226361Ustekinumab, 6 mg/kg at 0 weeks12 weekThe change from baseline in CDAI score: Undefined

[i] mg – milligrams, kg – kilograms, CDAI – Crohn’s Disease Activity Index, SES-CD – Simplified Endoscopic activity Score for Crohn’s Disease

The mean age of patients ranged from 35 to 40 years. The mean duration of CD history in patients across studies ranged from 8.8 to 13 years. Most patients had been treated with TNF inhibitors. For more information, see Supplementary Table 1.

Supplementary Figs. 1 and 2 show the results of the quality assessment. In the meta-analysis, the 7 studies were appraised for quality using the Cochrane Collaboration’s tool and all were judged to be of low overall risk and high quality.

Fig. 2

Forest plot of interleukin-23 and interleukin-12/23 inhibitors vs. placebo in clinical remission. A) Interleukin-23 inhibitor, B) interleukin-12/23 inhibitor

/f/fulltexts/CEJOI/52222/CEJI-48-52222-g002_min.jpg

Analysis of IL-23 or IL-12/23 inhibitors for the CD patients

For the primary outcome, patients receiving IL-23 inhibitors showed a higher clinical remission rate than patients with placebo therapy (RR = 2.11, 95% CI: 1.83-2.44, p < 0.001, I2 = 9%; Fig. 2A), and patients receiving IL-12/23 inhibitors also had a higher clinical remission rate than the placebo group (RR = 1.94, 95% CI: 1.64-2.29, p < 0.001; Fig. 2B). In terms of the second endpoints, both the IL-23 inhibitor group and the IL-12/23 inhibitor group showed significantly higher clinical response rates compared to the placebo group (RR = 1.92, 95% CI: 1.74-2.11; RR = 1.83, 95% CI: 1.61-2.09; respectively, Fig. 3). In addition, the IL-23 inhibitor group had a higher endoscopic remission rate and endoscopic response rate than placebo group, with a corresponding pooled RR of 3.40 (95% CI: 2.57-4.50, p < 0.001; Fig. 4) and 2.65 (95% CI: 2.65-3.12, p < 0.001; Fig. 5), respectively. Moreover, a higher proportion of patients in the IL-23 inhibitor group and IL-12/23 inhibitor group achieved CRP normalization compared to the placebo group (RR = 5.76, 95% CI: 4.10-8.11; RR = 2.38, 95% CI: 1.86-3.04; respectively, Fig. 6).

Fig. 3

Forest plot of interleukin-23 and interleukin-12/23 inhibitors vs. placebo in clinical response. A) Interleukin-23 inhibitor, B) interleukin-12/23 inhibitor

/f/fulltexts/CEJOI/52222/CEJI-48-52222-g003_min.jpg
Fig. 4

Forest plot of interleukin-23 inhibitors vs. placebo in endoscopic remission

/f/fulltexts/CEJOI/52222/CEJI-48-52222-g004_min.jpg
Fig. 5

Forest plot of interleukin-23 inhibitors vs. placebo in endoscopic response

/f/fulltexts/CEJOI/52222/CEJI-48-52222-g005_min.jpg
Fig. 6

Forest plot of interleukin-23 and interleukin-12/23 inhibitors vs. placebo in C-reactive protein. A) Interleukin-23 inhibitor, B) interleukin-12/23 inhibitor

/f/fulltexts/CEJOI/52222/CEJI-48-52222-g006_min.jpg

Discussion

In this study, we analyzed the efficacy of IL-23 inhibitors and IL-12/23 inhibitors in the induction phase for the treatment of CD. To our knowledge, this is the first systematic review and meta-analysis evaluating the effectiveness of IL-23 inhibitors in the induction phase of CD treatment. Although there have been previous meta-analyses on IL-12/23 inhibitors [24], this review was updated based on high-quality RCTs. Overall, our analysis validates the good performance of IL-23 inhibitors and IL-12/23 inhibitors in the induction phase of treating CD in patients with or without TNF experience. Compared with placebo, more patients in the IL-23 and IL-12/23 inhibitor groups achieved clinical remission, clinical response and CRP normalization. Additionally, more patients treated with IL-23 inhibitors achieved endoscopic remission and an endoscopic response compared to those who received placebo intervention.

Interleukin 23 plays an important role in a variety of inflammation-mediated autoimmune diseases such as psoriasis, psoriatic arthritis, ankylosing spondylitis, systemic lupus erythematosus, and IBD [25-29]. Biologics targeting IL-23p19 and IL-12/23p40 subunits have been confirmed to be effective in the treatment of psoriasis and psoriatic arthritis [30-32]. IL-23 is a member of the IL-12 cytokine family and consists of the IL-23p19 subunit and the IL-12/23p40 subunit, which is shared with IL-12 [33]. Ustekinumab specifically blocks the IL-12/23 subunit p40 in CD patients whereas risankizumab, brazikumab, guselkumab and mirikizumab selectively block the unique subunit p19. Ultimately, they all lead to the blockade of the IL-23 signaling pathway and suppress the inflammatory response [12, 34].

In previous studies, results from animal experiments and genetic studies have highlighted the role of the IL-23/IL-17 axis in the pathogenesis of IBD. A significant and selective increase of IL-23-responsive innate lymphoid cells (ILCs) was found in the inflamed intestine in CD patients but not in ulcerative colitis (UC) patients, where ILCs may promote intestinal inflammation through cytokine production and lymphocyte recruitment [35-37]. In addition, there are T-helper-cell-17 cells, γδT cell subsets, and natural killer T cells that respond to IL-23, and an increase in these cells also promotes the intestinal inflammatory response in CD patients [38-40]. In conclusion, various lines of evidence have suggested that IL-23 is an effector cytokine in the innate gut immune system, which may be a more specific target for the treatment of CD. Additionally, preclinical studies have shown no benefit of selective blocking of IL-12 in IBD [41, 42], and other studies in animal models have demonstrated that blocking IL-23 is more important than IL-12 in suppressing intestinal inflammation [43-45]. Research by Chapuy et al. found that IL-12 and mucosal CD14 monocyte-like cells induce IL-8 in colonic memory CD4 T cells in UC patients, but not in CD patients. This may be relevant to the pathogenesis of UC and CD [46].

The natural history of CD is characterized by periods of remission and relapse, and the probability of gastrointestinal complications increases when intestinal inflammation has not been well controlled [3]. The risk of developing stenosis or penetrating disease within 5 years of diagnosis of CD was 33.7%. The cumulative incidence of perianal or rectovaginal fistula within 10 years of diagnosis is 18% [47, 48]. Two RCTs reported improved fistula closure rates with TNF inhibitor treatment compared to placebo. However, due to the high recurrence rate and side effects, the long-term effect of TNF inhibitors was less favorable [49-51]. It is therefore necessary to find better treatments for CD and its complications.

In this study, the efficacy of IL-12/23 and IL-23 inhibitors in the induction phase of CD was confirmed in terms of clinical manifestations, endoscopic response and inflammatory markers. Clinical symptoms of CD correlate poorly with the degree of mucosal inflammation, and it is common to find significant mucosal inflammation during complete clinical remission [52]. Endoscopic healing is considered to be a better indicator of the severity of inflammation of the intestinal mucosa. Therefore, researchers consider symptom relief (clinical response, clinical remission) to be a short-term and intermediate treatment goal, while treatment aimed at endoscopic healing is more reflective of long-term outcomes and may reduce the risk of bowel injury [53, 54]. Fecal calprotectin and CRP were the two most widely used biomarkers in IBD, fecal calprotectin has high sensitivity and low specificity in identifying mucosal inflammation, whereas CRP has the opposite profile. Therefore, a normal CRP after initiating treatment should be considered as a minimal obligatory short- to medium-term target [53].

Treatment of CD includes induction therapy during an acute phase of the attack and long-term maintenance therapy. Therapeutic agents during the induction phase include higher doses of corticosteroids and biological therapies. However, with corticosteroids, adverse effects (especially metabolic bone diseases, infectious complications and growth disorders [55]) and complications associated with long-term use limit their use during the maintenance phase. In the maintenance phase, treatment options include immunomodulators (azathioprine, 6-mercaptopurine, and methotrexate) and biological therapies (TNF-α, IL-12/23, IL-23 and integrin α4β7).

At present, the “step-up” strategy (the introduction of biological therapies when multiple traditional therapeutic agents have failed) is still widely used in clinical practice [7, 56]. However, recent studies have shown that early biological therapy (mainly anti-TNF therapy) may improve the prognosis of patients and prevent progression to irreversible bowel damage [2, 57-59]. Berg et al. stated that the benefits of early biologics for CD were beneficial to control inflammation and avoid progression to irreversible stricture and penetrating disease. In contrast, patients with UC have predominantly mucosal rather than transmural inflammation and complications such as strictures are rare [7]. According to the British Society of Gastroenterology consensus guidelines, patients with jejunum or extensive small bowel disease have a poor prognosis, and early use of biological therapies may be considered. It also recommended that ustekinumab could be used in the induction and maintenance period of CD, both in anti-TNF naive patients and in patients who have failed anti-TNF treatment [60, 61]. This is despite growing evidence that IL-23 inhibitors are effective and safe for the treatment of CD. However, the use of IL-23 inhibitors was not explicitly recommended in recently published guidelines [2, 57, 60]. For the treatment of advanced severe CD, a head-to-head RCT showed that laparoscopic ileocecal resection is more medically effective and more cost-effective in treating end-stage ileitis in CD compared with biological therapies [62, 63].

There are some limitations to the present study. Firstly, IL-12/23 inhibitors other than ustekinumab were not included as data could not be extracted. Secondly, because the study focused on evaluating the efficacy of IL-23 and IL-12/23 inhibitors in the induction phase for the treatment of CD, the efficacy of long-term maintenance therapy is unknown. Finally, due to differences in study design among the included RCTs, the efficacy of IL-23 and IL-12/23 inhibitors in the induction period was not directly compared.

Conclusions

In this systematic review and meta-analysis, IL-12/23 inhibitors and IL-23 inhibitors were shown to be effective in the induction phase in the treatment of patients with CD, as evidenced by three aspects: clinical symptoms, endoscopic symptoms, and inflammatory markers. The IL-23 and IL-12/23 inhibitors have the potential to be a promising therapeutic option for CD patients who have had insufficient responses to conventional and TNF therapies.

Notes

[2] Conflicts of interest The authors declare no conflict of interest.

References

1 

Torres J, Mehandru S, Colombel JF, Peyrin-Biroulet L (2017): Crohn’s disease. Lancet 389: 1741-1755.

2 

Torres J, Bonovas S, Doherty G, et al. (2020): ECCO Guidelines on therapeutics in Crohn’s disease: Medical treatment. J Crohns Colitis 14: 4-22.

3 

Peyrin-Biroulet L, Loftus EV, Jr., Colombel JF, Sandborn WJ (2010): The natural history of adult Crohn’s disease in population-based cohorts. Am J Gastroenterol 105: 289-297.

4 

Meima-van Praag EM, Buskens CJ, Hompes R, Bemelman WA (2021): Surgical management of Crohn’s disease: a state of the art review. Int J Colorectal Dis 36: 1133-1145.

5 

Feuerstein JD, Cheifetz AS (2017): Crohn disease: Epidemiology, diagnosis, and management. Mayo Clin Proc 92: 1088-1103.

6 

Ng SC, Shi HY, Hamidi N, et al. (2017): Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet 390: 2769-2778.

7 

Berg DR, Colombel JF, Ungaro R (2019): The role of early biologic therapy in inflammatory bowel disease. Inflamm Bowel Dis 25: 1896-1905.

8 

Ben-Horin S, Chowers Y (2011): Review article: loss of response to anti-TNF treatments in Crohn’s disease. Aliment Pharmacol Ther 33: 987-995.

9 

Roda G, Jharap B, Neeraj N, Colombel JF (2016): Loss of response to anti-TNFs: Definition, epidemiology, and management. Clin Transl Gastroenterol 7: e135.

10 

Ben-Horin S, Novack L, Mao R, et al. (2022): Efficacy of biologic drugs in short-duration versus long-duration inflammatory bowel disease: A systematic review and an individual-patient data meta-analysis of randomized controlled trials. Gastroenterology 162: 482-494.

11 

Uhlig HH, McKenzie BS, Hue S, et al. (2006): Differential activity of IL-12 and IL-23 in mucosal and systemic innate immune pathology. Immunity 25: 309-318.

12 

Schmitt H, Neurath MF, Atreya R (2021): Role of the IL23/IL17 pathway in Crohn’s disease. Front Immunol 12: 622934.

13 

Page MJ, McKenzie JE, Bossuyt PM, et al. (2021): The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ 372: n71.

14 

Higgins JPT, Altman DG, Gøtzsche PC, et al. (2011): The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ 343: d5928.

15 

Higgins JPT, Thompson SG, Deeks JJ, Altman DG (2003): Measuring inconsistency in meta-analyses. BMJ 327: 557-560.

16 

Egger M, Davey Smith G, Schneider M, Minder C (1997): Bias in meta-analysis detected by a simple, graphical test. BMJ 315: 629-634.

17 

D’Haens G, Panaccione R, Baert F, et al. (2022): Risankizumab as induction therapy for Crohn’s disease: results from the phase 3 ADVANCE and MOTIVATE induction trials. Lancet 399: 2015-2030.

18 

Sandborn WJ, D’Haens GR, Reinisch W, et al. (2022): Guselkumab for the treatment of Crohn’s disease: Induction results from the phase 2 GALAXI-1 study. Gastroenterology 162: 1650-1664.e1658.

19 

Sands BE, Peyrin-Biroulet L, Kierkus J, et al. (2022): Efficacy and safety of mirikizumab in a randomized phase 2 study of patients with Crohn’s disease. Gastroenterology 162: 495-508.

20 

Sands BE, Chen J, Feagan BG, et al. (2017): Efficacy and safety of MEDI2070, an antibody against interleukin 23, in patients with moderate to severe Crohn’s disease: A phase 2a study. Gastroenterology 153: 77-86.e76.

21 

Feagan BG, Sandborn WJ, D’Haens G, et al. (2017): Induction therapy with the selective interleukin-23 inhibitor risankizumab in patients with moderate-to-severe Crohn’s disease: a randomised, double-blind, placebo-controlled phase 2 study. Lancet 389: 1699-1709.

22 

Feagan BG, Sandborn WJ, Gasink C, et al. (2016): Ustekinumab as induction and maintenance therapy for Crohn’s disease. N Engl J Med 375: 1946-1960.

23 

Sandborn WJ, Gasink C, Gao LL, et al. (2012): Ustekinumab induction and maintenance therapy in refractory Crohn’s disease. N Engl J Med 367: 1519-1528.

24 

Honap S, Meade S, Ibraheim H, et al. (2022): Effectiveness and safety of ustekinumab in inflammatory bowel disease: A systematic review and meta-analysis. Dig Dis Sci 67: 1018-1035.

25 

Lubberts E (2015): The IL-23-IL-17 axis in inflammatory arthritis. Nat Rev Rheumatol 11: 415-429.

26 

Vecellio M, Hake VX, Davidson C, et al. (2020): The IL-17/IL-23 axis and its genetic contribution to psoriatic arthritis. Front Immunol 11: 596086.

27 

Bunte K, Beikler T (2019): Th17 cells and the IL-23/IL-17 axis in the pathogenesis of periodontitis and immune-mediated inflammatory diseases. Int J Mol Sci 20: 3394.

28 

Gaffen SL, Jain R, Garg AV, Cua DJ (2014): The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol 14: 585-600.

29 

Almradi A, Hanzel J, Sedano R, et al. (2020): Clinical trials of IL-12/IL-23 inhibitors in inflammatory bowel disease. BioDrugs 34: 713-721.

30 

Huang X, Shentu H, He Y, et al. (2023): Efficacy and safety of IL-23 inhibitors in the treatment of psoriatic arthritis: a meta-analysis based on randomized controlled trials. Immunol Res 71: 505-515.

31 

Yang K, Oak ASW, Elewski BE (2021): Use of IL-23 inhibitors for the treatment of plaque psoriasis and psoriatic arthritis: A comprehensive review. Am J Clin Dermatol 22: 173-192.

32 

van Vollenhoven RF, Hahn BH, Tsokos GC, et al. (2018): Efficacy and safety of ustekinumab, an IL-12 and IL-23 inhibitor, in patients with active systemic lupus erythematosus: results of a multicentre, double-blind, phase 2, randomised, controlled study. Lancet 392: 1330-1339.

33 

Oppmann B, Lesley R, Blom B, et al. (2000): Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity 13: 715-725.

34 

Fiorino G, Allocca M, Correale C, et al. (2020): Positioning ustekinumab in moderate-to-severe ulcerative colitis: new kid on the block. Expert Opin Biol Ther 20: 421-427.

35 

Duerr RH, Taylor KD, Brant SR, et al. (2006): A genome-wide association study identifies IL23R as an inflammatory bowel disease gene. Science 314: 1461-1463.

36 

Geremia A, Arancibia-Carcamo CV, Fleming MP, et al. (2011): IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J Exp Med 208: 1127-1133.

37 

Hanžel J, D’Haens GR (2020): Anti-interleukin-23 agents for the treatment of ulcerative colitis. Expert Opin Biol Ther 20: 399-406.

38 

Doisne JM, Becourt C, Amniai L, et al. (2009): Skin and peripheral lymph node invariant NKT cells are mainly retinoic acid receptor-related orphan receptor (gamma)t+ and respond preferentially under inflammatory conditions. J Immunol 183: 2142-2149.

39 

McVay LD, Li B, Biancaniello R, et al. (1997): Changes in human mucosal gamma delta T cell repertoire and function associated with the disease process in inflammatory bowel disease. Mol Med 3: 183-203.

40 

Nanno M, Kanari Y, Naito T, et al. (2008): Exacerbating role of gammadelta T cells in chronic colitis of T-cell receptor alpha mutant mice. Gastroenterology 134: 481-490.

41 

Cua DJ, Sherlock J, Chen Y, et al. (2003): Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature 421: 744-748.

42 

Davies SC, Nguyen TM, Parker CE, et al. (2019): Anti-IL-12/23p40 antibodies for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 12: Cd012804.

43 

Yen D, Cheung J, Scheerens H, et al. (2006): IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6. J Clin Invest 116: 1310-1316.

44 

Iwakura Y, Ishigame H (2006): The IL-23/IL-17 axis in inflammation. J Clin Invest 116: 1218-1222.

45 

Parigi TL, Iacucci M, Ghosh S (2022): Blockade of IL-23: What is in the Pipeline? J Crohns Colitis 16: ii64-ii72.

46 

Chapuy L, Bsat M, Rubio M, et al. (2020): IL-12 and mucosal CD14+ monocyte-like cells induce IL-8 in colonic memory CD4+ T cells of patients with ulcerative colitis but not Crohn’s disease. J Crohns Colitis 14: 79-95.

47 

Thia KT, Sandborn WJ, Harmsen WS, et al. (2010): Risk factors associated with progression to intestinal complications of Crohn’s disease in a population-based cohort. Gastroenterology 139: 1147-1155.

48 

Park SH, Aniwan S, Scott Harmsen W, et al. (2019): Update on the natural course of fistulizing perianal Crohn’s disease in a population-based cohort. Inflamm Bowel Dis 25: 1054-1060.

49 

Lichtiger S, Binion DG, Wolf DC, et al. (2010): The CHOICE trial: adalimumab demonstrates safety, fistula healing, improved quality of life and increased work productivity in patients with Crohn’s disease who failed prior infliximab therapy. Aliment Pharmacol Ther 32: 1228-1239.

50 

Present DH, Rutgeerts P, Targan S, et al. (1999): Infliximab for the treatment of fistulas in patients with Crohn’s disease. N Engl J Med 340: 1398-1405.

51 

Sands BE, Anderson FH, Bernstein CN, et al. (2004): Infliximab maintenance therapy for fistulizing Crohn’s disease. N Engl J Med 350: 876-885.

52 

Laterza L, Piscaglia AC, Minordi LM, et al. (2018): Multiparametric evaluation predicts different mid-term outcomes in Crohn’s disease. Dig Dis 36: 184-193.

53 

Turner D, Ricciuto A, Lewis A, et al. (2021): 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 160: 1570-1583.

54 

Mumolo MG, Bertani L, Ceccarelli L, et al. (2018): From bench to bedside: Fecal calprotectin in inflammatory bowel diseases clinical setting. World J Gastroenterol 24: 3681-3694.

55 

Irving PM, Gearry RB, Sparrow MP, Gibson PR (2007): Review article: appropriate use of corticosteroids in Crohn’s disease. Aliment Pharmacol Ther 26: 313-329.

56 

Yadav A, Foromera J, Feuerstein I, et al. (2017): Variations in health insurance policies regarding biologic therapy use in inflammatory bowel disease. Inflamm Bowel Dis 23: 853-857.

57 

Lichtenstein GR, Loftus EV, Jr., Isaacs KL, et al. (2018): Correction: ACG Clinical Guideline: Management of Crohn’s disease in adults. Am J Gastroenterol 113: 1101.

58 

Sriranganathan D, Segal JP, Garg M (2022): Biologics recommendations in the ECCO guidelines on therapeutics in Crohn’s disease: medical treatment. Frontline Gastroenterol 13: 168-170.

59 

Schreiber S, Reinisch W, Colombel JF, et al. (2013): Subgroup analysis of the placebo-controlled CHARM trial: increased remission rates through 3 years for adalimumab-treated patients with early Crohn’s disease. J Crohns Colitis 7: 213-221.

60 

Lamb CA, Kennedy NA, Raine T, et al. (2019): British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut 68: s1-s106.

61 

Deepak P, Loftus EV, Jr. (2016): Ustekinumab in treatment of Crohn’s disease: design, development, and potential place in therapy. Drug Des Devel Ther 10: 3685-3698.

62 

Ponsioen CY, de Groof EJ, Eshuis EJ, et al. (2017): Laparoscopic ileocaecal resection versus infliximab for terminal ileitis in Crohn’s disease: a randomised controlled, open-label, multicentre trial. Lancet Gastroenterol Hepatol 2: 785-792.

63 

Stevens TW, Haasnoot ML, D’Haens GR, et al. (2020): Laparoscopic ileocaecal resection versus infliximab for terminal ileitis in Crohn’s disease: retrospective long-term follow-up of the LIR!C trial. Lancet Gastroenterol Hepatol 5: 900-907.

Copyright: © 2024 Polish Society of Experimental and Clinical Immunology This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0) License (http://creativecommons.org/licenses/by-nc-sa/4.0/), allowing third parties to copy and redistribute the material in any medium or format and to remix, transform, and build upon the material, provided the original work is properly cited and states its license.
 
Quick links
© 2024 Termedia Sp. z o.o.
Developed by Bentus.