Next Article in Journal
Effects of a 12-Week Functional Training Program on the Strength and Power of Chinese Adolescent Tennis Players
Next Article in Special Issue
Change of Sleep Stage during Gastroesophageal Reflux in Infants
Previous Article in Journal
Pediatric Bowel Management Options and Organizational Aspects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current Role of Monoclonal Antibody Therapy in Pediatric IBD: A Special Focus on Therapeutic Drug Monitoring and Treat-to-Target Strategies

1
Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-University-Erlangen-Nuremberg, 91054 Erlangen, Germany
2
German Center Immunotherapy, DZI, Ulmenweg 18, 91054 Erlangen, Germany
*
Author to whom correspondence should be addressed.
Children 2023, 10(4), 634; https://doi.org/10.3390/children10040634
Submission received: 13 February 2023 / Revised: 22 March 2023 / Accepted: 27 March 2023 / Published: 28 March 2023
(This article belongs to the Special Issue Cutting Edge Research on Pediatric Gastroenterology)

Abstract

:
In the last two decades, biologicals have become essential in treating children and adolescents with inflammatory bowel disease. TNF-α inhibitors (infliximab, adalimumab and golimumab) are preferentially used. Recent studies suggest that early application of TNF-α inhibitors is beneficial to inducing disease remission and preventing complications such as development of penetrating ulcers and fistulas. However, treatment failure occurs in about one third of pediatric patients. Particularly, children and adolescents differ in drug clearance, emphasizing the importance of pharmacokinetic drug monitoring in the pediatric setting. Here, current data on the choice and effectiveness of biologicals and therapeutic drug monitoring strategies are reviewed.

1. Introduction

The use of biologicals in pediatric patients with inflammatory bowel disease (IBD) has widely increased since their introduction [1]. Biologicals have substantially improved the disease course for many pediatric patients suffering from Crohn’s disease (CD) and ulcerative colitis (UC). Current guidelines recommend the use of TNF-α inhibitors in pediatric IBD patients with high disease activity or in those who do not respond to other therapeutic strategies [2,3]. Infliximab and adalimumab were approved for the treatment of pediatric Crohn’s disease in 2006 and 2012, respectively, and later for pediatric UC also. These compounds are thus the most recommended. As a result, the data on the use of biologics in pediatric and adolescent patients with IBD have increased in number significantly in recent years. Biologic agents are effective and safe, and they are one of the most used medication classes in pediatric IBD [4].
In a large German cohort (CEDATA), adalimumab was the most commonly used biologic when therapy with infliximab failed. According to Cozijnsen and colleagues, this approach was effective in a small retrospective study [5]. Infliximab, adalimumab and golimumab bind sTNF and mTNF, while etanercept only binds soluble TNFu [6]. The reason for the use of adalimumab after infliximab treatment failure may be that other biological agents lack official approval in childhood and are used off-label despite being available for treatment in adults. However, information on treatment success exists in children for alternative biologicals such as golimumab, certolizumab (both TNF-α inhibitors), vedolizumab, an α4β7-integrin blocker, and ustekinumab, an IL-12/IL-23 blocker (see Figure 1 for an overview of currently used biologicals in pediatric IBD). Approximately 54% of 42 pediatric IBD patients treated with vedolizumab entered clinical remission within 14 weeks, even when prior treatment with anti-TNF-α drugs had failed, according to a retrospective multicenter study [7]. Another large multicenter study demonstrated ustekinumab to be safe and effective in the treatment of Crohn’s disease [8]. Pediatric patients with UC achieved deep mucosal remission when treated with ustekinumab, even when a relapse occurred under therapy with infliximab and vedolizumab [9]. Similarly, golimumab resulted in clinical remission [10]. Ninety percent of TNF-α naïve patients remained steroid-free compared to 50% of patients who did not respond to other biologic therapies [10]. Therefore, European guidelines recommend treatment with infliximab or adalimumab as an effective option for pediatric patients with moderate-to-severe Crohn’s disease. After the failure of anti-TNF-α or other treatments, ustekinumab and vedolizumab are recommended [2,3].

2. Clinical Effect of Biologics

The large German–Austrian registry study of GPGE (CEDATA) showed that pediatric patients with high disease activity were significantly more likely to receive biologic agents, preferably infliximab [1]. Treatment is most likely to last approximately one year, but only because the surveillance ended [1,11]. The discontinuation rate was 3.2% per year due to a loss of response in a Canadian study [11]. Treatment outcomes suggest that patients with high disease activity in Crohn’s disease respond equally well to infliximab and adalimumab, although randomized controlled head-to-head studies are lacking [12,13]. After three months of treatment, infliximab induced significant mucosal healing and was associated with improvement in clinical disease scores in adults [14]. Similarly, patients with Crohn’s disease treated with adalimumab experienced mucosal healing [15]. Scarallo and colleagues also found that infliximab and adalimumab induced mucosal and histologic healing in about 40% of pediatric patients with CD and UC [16]. Inflammatory markers such as C-reactive protein (CRP) and white blood cell count decreased in children and adolescents with moderate-to-severe Crohn’s disease on TNF-α inhibitor therapy [17]. Compared with enteral nutrition therapy alone, biological therapy is similarly effective in inducing mucosal remission in patients with Crohn’s disease and significantly improves the quality of life [18,19]. A review of clinical trials demonstrated the long-term therapeutic benefit of infliximab in pediatric patients with Crohn’s disease on continuous therapy [20]. With anti-TNF-α therapy, approximately 60% of pediatric patients with perianal CD respond well to treatment, and 40% achieve sustained remission [21].
In moderate-to-severe ulcerative colitis, treatment with infliximab led to remission and was safe [22]. The treatment with infliximab in pediatric patients with ulcerative colitis is associated with a lower frequency of colectomy compared with other treatment options [23]. Adalimumab also showed good results in the double-blind ENVISION I trial to treat children with moderate-to-severe ulcerative colitis, with a higher induction dosage yielding better results [24]. However, treatment with biologics did not affect the number of hospitalizations in general [23]. Comparing the years before the introduction of biologics in children and adolescents with Crohn’s disease, the time thereafter showed less disease progression to stenosing disease and fewer surgeries but unaltered overall hospitalizations [25].
Pediatric patients with IBD also gained weight and, especially, grew up to the same height as healthy controls when treated with TNF-α blockers. This also led to a significant increase in physical activity, while the overall health-related quality of life remained unchanged when compared to pre-anti-TNF-α treatment [26].

3. Methods and Selection Criteria

A non-systematic literature search of PubMed was performed in January 2023, using the following search terms: (“Crohn Disease”[Mesh] OR “Inflammatory Bowel Diseases”[Mesh] OR “Colitis, Ulcerative”[Mesh] OR “Pediatric ulcerative colitis” [Supplementary Concept] OR “Pediatric Crohn’s disease” [Supplementary Concept]) AND (“Infliximab”[Mesh] OR “Adalimumab”[Mesh] OR “Tumor Necrosis Factor Inhibitors” [Pharmacological Action] OR “vedolizumab” [Supplementary Concept] OR “Ustekinumab”[Mesh] OR “golimumab” [Supplementary Concept] OR “tofacitinib” [Supplementary Concept]) AND (“Child”[Mesh] OR “Adolescent”[Mesh]). Additionally, the references of selected studies were screened for further studies. The inclusion criteria were a pediatric sample with IBD and the use of a biological agent such as infliximab, adalimumab, golimumab, ustekinumab and vedolizumab. The main aim was to include current literature, so mostly studies of retrospective or prospective nature, meta-analysis, and case reports since 2020 were considered, but we included earlier studies and adult studies if evidence was lacking (see Table 1 for all included pediatric studies).

4. Early and Effective Use of TNF-α Blockers Prevents Disease Progression and Disease Complications

Evidence suggests that early treatment with biological agents is favorable for pediatric CD patients [1,28,29]. Early application of biological agents significantly prevented treatment failure [1]. In 2020, the first randomized direct comparison of first-line infliximab with exclusive enteral nutrition or corticosteroids as first-line treatment in pediatric patients with moderate-to-severe Crohn’s disease was provided by Jongsma and colleagues [50]. Of the patients treated with first-line anti-TNF-α therapy, a significantly higher percentage accomplished clinical and endoscopic remission [50]. In addition, first-line TNF-α-blocker therapy needed less dose escalation while achieving mucosal healing [50]. Consistent with this, data from the CEDATA registry study showed that first-line infliximab therapy led to a higher rate of clinical remission in the short term compared to conventional therapy with biologics, which led to endoscopic remission in another study [1,50]. In addition, Jongsma and colleagues found that the probability of continued clinical remission at week 52 with monotherapy of azathioprine was higher in children who received infliximab as first-line therapy to induce remission [50]. Comparable results were shown for adalimumab [72]. In Crohn’s disease, early and effective use of TNF-α blockers also prevented the development of disease complications, for example, strictures or penetrating ulcerations and disease progression [28]. A Korean study revealed that early anti-TNF-α medication led to a lower risk of surgery during disease progression [30]. In another study, early admission of biologics significantly reduced the risk of penetrating complications but not stenosing complications [31].
A large cohort study showed that unrelated to the IBD subtype, the administration of biologicals a short time after diagnosis (<120 days) is connected to fewer glucocorticoids being needed [32]. Another large cohort study of pediatric patients with IBD demonstrated that the early treatment with TNF-α blockers was superior to immunomodulators in achieving remission within three months [29]. In a retrospective Canadian study, earlier initiation of anti-TNF-α treatment in patients with Crohn’s disease and ulcerative colitis was more common in adolescents and was associated with higher PCDAI/PUCAI and lower serum albumin levels at diagnosis [33].
The incidence of extraintestinal manifestation in IBD throughout treatment with biologicals ranges from 14% to 25% [1,78]. However, the study with the longer follow-up reported a higher incidence of extraintestinal manifestations [1]. In the large German multicenter cohort, first-line prescription significantly reduced the incidence of extraintestinal manifestations longitudinally [1]. These patients had the highest disease activity due to systemic inflammation before treatment, so the findings are promising. Moreover, the incidence of extraintestinal manifestations was reduced from about 27% to 25% immediately after treatment initiation, with a further reduction to 17% at six months [1].

5. Occurrence and Frequency of Adverse Treatment Events

In several studies evaluating different biologicals, adverse treatment events occurred in around 46% of patients within all IBD subtypes [1,79]. Immediate infusion reactions (11%) and a psoriasis-like rash (11%) were reported [34,79]. Symptoms of infusion reactions include dyspnea, coughing, cyanosis and vomiting [19]. Minor infections were reported in 15.4% of patients [34].
Several studies have reported varying rates of skin complications due to biological treatment ranging from 13% to 39%, with the most recent study reporting 17% [1,36,37]. Even for golimumab, severe skin reactions were the reason for discontinuation in a case study of adults with CD [80]. Dolinger et al. recommend switching to ustekinumab in the event of skin reactions on TNF-α inhibitor therapy. In the study by Nuti et al., a psoriasis-like rash was observed in 11% of patients treated with infliximab or adalimumab [34]. The risk of skin adverse events appears to be increased only with adalimumab and not with infliximab in patients with IBD and juvenile idiopathic arthritis [38]. There appears to be no association between higher drug concentrations and increased adverse event rates [39]. Vedolizumab and ustekinumab showed a good safety profile [79,81]. The most common adverse events were respiratory tract infections (33%) with vedolizumab [79] and infusion reactions with ustekinumab [10]. Additional adverse events related to vedolizumab were headaches (4%) and myalgia (3%), while only 1% of patients discontinued treatment due to adverse events in a multicenter cohort study [75]. More serious adverse events are rarely observed and are described in case reports. The currently available evidence suggests that treatment with TNF-α inhibitors is associated with a very low risk of developing malignancies. These almost exclusively occurred when combining TNF-α blockers with azathioprine in male patients [35].

6. Therapeutic Drug Monitoring to Optimize the Treatment Strategy and Maintain the Efficacy of Biological Agents

Primary and secondary treatment failures with anti-TNF-α drugs are common and challenging in daily clinical practice. Approximately 10% to 30% of adult patients experience primary non-response [82], while 20% to 50% develop secondary loss of response during biological therapy [82]. A large retrospective registry study of adults with ulcerative colitis showed that around 50% of the patients had a suboptimal response to anti-TNF-α agents, leading to dose escalation or treatment discontinuation [83]. The main causes of primary non-response or loss of response are low trough levels or anti-drug antibodies, respectively [2]. For preventing treatment failure and also for following a treat-to-target strategy, optimal dosing is important for achieving not only clinical remission but also mucosal healing as one of the most important long-term goals [84]. Short-term goals that reflect adequate therapy management include the normalization of inflammation markers in the serum and feces [84].
A meta-analysis by the ESPGHAN-IBD working group suggests that a higher dose per kilogram of body weight may be appropriate in younger IBD patients, as they often have lower trough levels in the early phase of therapy induction [51]. Indeed, a retrospective British study demonstrated that children with very early onset IBD received the increased dose of 10 mg/kg body weight [40]. The remission rate in these patients was 62%; otherwise, the course of very young children with IBD treated with biologics appears to be similar to that of older patients [40,41]. In particular, studies in pediatric patients with ulcerative colitis and Crohn’s disease have shown that higher infliximab trough levels after induction predict remission one year after infliximab administration [52,53] (see also Table 2).
A large UK prospective observational study (PANTS) of approximately 1000 children and adults with IBD tried to identify clinical and pharmacokinetic factors that might predict primary non-response at week 14, non-remission at week 54, and adverse events leading to drug discontinuation [42]. In the multivariable regression analysis, the only factor independently associated with a primary non-response was low drug trough levels of infliximab and adalimumab at week 14 [42]. Approximately 63% of the patients developed anti-drug antibodies to infliximab and 29% to adalimumab [42]. For both drugs, suboptimal drug concentrations at week 14 predicted immunogenicity and the development of neutralizing anti-drug antibodies predicted subsequent low drug concentrations [42]. A further important finding was that a combination immunomodulatory therapy (thiopurine or methotrexate) reduced the risk of anti-drug antibody development for infliximab and adalimumab [42].
Therapeutic drug monitoring (TDM) is advocated to assess trough levels and/or neutralizing anti-drug antibodies (ADA) to optimize the treatment strategy and maintain the efficacy of biological agents. TDM can either be reactive or proactive. In reactive TDM, drug concentrations and/or the occurrence of ADAs are assessed in the serum in case of persistent or recurrent flares of IBD. Reactive TDM can streamline the management of primary non-response and secondary loss of response. If the drug concentration appears to be subtherapeutic, the dose may either be increased or the interval between the doses reduced. If the ADA titer is low, adding an immunomodulator to the biologic treatment should be considered. In case of a high ADA titer, the biological agent may be switched to another biologic in the same class or a different class.
Proactive TDM signifies the assessment of drugs’ trough levels during remission to ensure effective therapy, prevent a disease relapse by maintaining adequate drug levels, and reduce the formation of ADA. Proactive TDM increases clinical remission and the durability of the response to a biological agent [69]. In recent years, guidelines and consensus statements have been published on the emerging topic of TDM for adults [85,86,87,88,89,90] and children [2,3]. These guidelines recommend that reactive TDM to guide treatment in patients with biologicals is more cost-effective than empiric dose escalation.
In recent years, randomized control trials such as the Pediatric Crohn’s Disease Adalimumab Level-based Optimization Treatment (PAILOT) trial [69] and the NOR-DRUM B study have suggested the utility of proactive TDM [91]. Proactive drug monitoring of adalimumab in the randomized PAILOT trial was associated with significantly higher rates of corticoid-free remission and lower inflammatory markers [69]. Infliximab trough levels greater than 10 mg/mL are generally associated with remission and higher rates of perianal fistula healing in pediatric IBD patients [54,92]. Yarur and colleagues recommend in adults a treat-to-target strategy until adequate infliximab levels are achieved [92]. Of note, current data indicate that an infliximab or adalimumab therapy should generally not be discontinued unless drug levels are greater than 10 μg/mL [92].
As higher infliximab levels after induction were associated with clinical remission [55], proactive drug monitoring in the induction phase of infliximab was associated with optimal trough levels at week 52 and clinical remission in pediatric IBD patients [56]. In particular, early response and drug monitoring during induction appear to predict response rates, possibly due to higher drug clearance in children and an association with higher cytokine levels at diagnosis [57,58]. Higher drug clearance was associated with hypoalbuminemia, high CRP, higher BMI, male sex and anti-drug antibodies [59,81,93]. A small Spanish study points out that proactive drug monitoring during maintenance is favorable in order to maintain long term clinical response and showed response rates of 92.8% after three years in pediatric patients with Crohn’s disease [43].
In addition, in adults, a high initial serum TNF-α and a severe inflammation with extensive mucosal involvement leads to increased drug consumption [94] and fecal loss [95], while younger age (<10 years) is attributed to different pharmacokinetics in children compared to adults [51,96]. This has led to the revised recommendation of an intensified infliximab treatment (10 mg/kg body weight at weeks 0, 1 and 4) to achieve remission in cases of an acute severe colitis by the ESPGHAN in 2018 [97].
In a small Spanish cohort of pediatric patients with Crohn’s disease, proactive drug monitoring (measurement of trough levels) prevented loss of response to infliximab and adalimumab due to antibodies [43]. Anti-drug antibodies are associated with loss of response to infliximab [60]. In patients who have already developed anti-drug antibodies, dose escalation of the biological drug suppressed anti-drug antibodies in the subsequent study [43,45]. Another approach to suppress anti-drug antibodies is to combine the biologic with an immunomodulator which is supported by the evidence for infliximab [46] but not for adalimumab [70] in pediatric patients with Crohn’s disease. Patients receiving infliximab as a second-line treatment for failed therapy benefit significantly from combination therapy with immunomodulators [20]. In general, combination therapy increased the likelihood of continuing infliximab at two years [20].
For several of the above-mentioned reasons, the dose of the biological drug does not necessarily correspond to the determined drug trough levels [54]. Therefore, a Bayesian calculation model applied to drug concentrations represents a new approach to optimize treatment response to biologics in IBD by incorporating several individual parameters that affect drug clearance, such as sex, hypoalbuminemia, and fecal loss [54]. It predicts the treatment response to optimize dosing [98]. With the implementation of three trough-level measurements, the model was able to predict drug concentrations and thus be helpful for therapy adjustments [54]. Precision dosing showed better remission and response rates in adults compared to traditional dosing regimens [61]. Sufficient models for children in clinical practice have yet to be determined due to the large amount of data needed to test the robustness and identify an appropriate computational model to predict individual drug concentrations. However, data on TDM in pediatric IBD are emerging and allow for recommendations for treatment monitoring (summarized in Table 2). Notably, to date, the superiority of proactive TDM has not been consistently demonstrated in randomized controlled trials [99].
Table 2. Target trough levels during induction and maintenance as reported from recent studies.
Table 2. Target trough levels during induction and maintenance as reported from recent studies.
InductionMaintenance
Infliximab>18 µg/mL before 3rd infusion to achieve clinical remission in CD [63]
>12.7 µg/mL before 4th infusion for fistula healing and >9.1 to prevent treatment failure in CD [64,65]
>13 ug/mL before 4th infusion for fistula healing (*) [66]
>7µg/mL to prevent treatment failure in CD [42]—>8.3 µg/mL for clinical remission in CD [67]
>10.1 µg/mL for fistula healing in CD [92]
Adalimumab>13.85 µg/mL at the end of induction for long term clinical remission in UC and CD [73]≥10.1 μg/mL–12 µg/mL (*) to prevent treatment failure [42]
In case of loss of response—>new induction dose or weekly application [71,100]
Golimumab >0.97 μg/mL at week 14 for clinical response in UC [101]
Ustekinumab ≈6.6 µg/mL at week 8 (associated with steroid-free remission week 52) in all IBD subtypes [10]
Vedolizumab>37 µg/mL before 3rd infusion and >20 µg/mL before 4th infusion to achieve steroid free-clinical remission in UC and CD [77]
>30 µg/mL in week 2 (*) for endoscopic remission, clinical remission in CD and UC [102]
<30 kg: >7 μg/mL for steroid free and EEN-free remission in all IBD subtypes [75]
>30 kg: ≥11.5 µg/mL for clinical and biochemical remission in CD and UC (*) [76]
Note that not all studies performed cut-off tests for trough levels and some studies did not find an association between trough levels and disease outcome (e.g., [9,103]), (*) = adult studies.
Surgical interventions and partial bowel resection for Crohn’s disease still represent a rescue option. However, compared to the beginning of the 2000’s, these procedures are less frequent, especially in patients responding to TNF-α inhibitors [68]. It is well known, especially in pediatric Crohn’s disease patients, that the postoperative recurrence risk after surgery is substantial. In a pediatric series, clinical recurrence rates after partial intestinal resection were 17% at 1 year, 38% at 3 years and 60% at 5 years [104]. Therefore, a postoperative remission-maintaining therapy should be used after surgically induced remission in children, as recommended by the ECCO/ESPGHAN expert committee [2,105]. Thiopurine is recommended as the first choice for postoperative relapse prophylaxis in IFX-naïve patients and anti-TNF-α antibodies in high-risk cases. While in pediatric IBD, randomized controlled trials on this topic are lacking, supporting data for the postoperative use of anti-TNF-α therapy to reduce the risk of recurrence at the anastomoses was reported by three RCTs conducted in adult patients with ileocolonic resections and primary anastomoses [106,107,108]. A recent German study reported a reduced endoscopic recurrence after ileocecal resection in children receiving preoperative TNF-α inhibitors [49].
In summary, the quality and efficacy of treatment in pediatric IBD appear to have improved, as children with Crohn’s disease suffer fewer relapses in the last five years than 10–15 years ago [47]. Early treatment with infliximab or adalimumab should be considered if patients are at high risk of a poor outcome, e.g., Crohn’s with persistently high disease activity despite adequate induction therapy, extensive or pan-enteric manifestation, deep colonic ulcerations, marked growth retardation, severe perianal involvement, radiologically or endoscopically proven structures, the occurrence of fistulas, intestinal perforations, inflammatory conglomerates and/or abscesses, and CMV infections [2]. Similar features in ulcerative colitis qualify for a TNF-α inhibitor as pancolitis, extensive and deep colonic ulcerations, the early need for (recurrent) steroid therapy, and recurrent infections with Clostridioides difficile or CMV [3,97].
Additional recommendations will soon further refine biological therapy strategies; for example, trials are comparing longer dosing intervals in children in remission on TNF-α blockers [109], which would further improve the quality of life of children. One study even showed that discontinuation of biologics could be considered if endoscopic and histologic remission occurs in children with ulcerative colitis on TNF-α blockers [48]. Individualized medicine, considering pharmacogenetic and pharmacogenomic aspects, is expected to lead to further advances in treatment. For example, a study of response to infliximab found that a variant in the FCGR3A gene was associated with a decreased response to infliximab with lower levels and higher anti-IFX antibody concentrations [62]. HLA polymorphisms (G allele of rs2395185 and the C allele of rs2097432) were associated with reduced long-term response in adults but not children with CD to anti-TNF-α medication [44]. So pharmacological models might have to take different polymorphisms in children and adults into account.

7. Conclusions

TNF-α blockers are a safe and efficient way to treat IBD with high disease activity in children and adolescents. Infliximab and adalimumab are efficient in achieving clinical and mucosal remission. However, as treatment failure still occurs, therapeutic drug monitoring and exclusion of the formation of anti-drug antibodies are helpful for further treatment management. For both infliximab and adalimumab, drug concentrations to achieve different treatment goals are available. Therapeutic drug monitoring involves a proactive and a reactive strategy, yet further prospective RCTs are still needed to pose recommendations for which one to prefer. For other monoclonal antibodies, such as vedolizumab and ustekinumab, favorable drug concentrations are mostly derived from adult studies.

Author Contributions

Conceptualization, M.C. and A.H.; methodology, M.C.; validation, A.H., writing—original draft preparation, M.C.; writing—review and editing, M.C. and A.H.; visualization, M.C.; supervision, A.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded in part by a research grant from the Johannes und Frieda Marohn-Stiftung to A.H.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAAnti-drug Antibodies
BMIBody mass index
CD
CEDATA
CMV
Crohn’s disease
Registry of Pediatric Patients with IBD in German-speaking countries
Cytomegaly virus
CRPC-reactive Protein
EIMExtraintestinal manifestations
ESPGHANEuropean Society for Pediatric Gastroenterology Hepatology and Nutrition
GPGEGerman Association of Pediatric Gastroenterology
IBDInflammatory bowel disease
IFXInfliximab
PCDAIPediatric Crohn’s disease activity index
PUCAI
UC
Pediatric ulcerative colitis activity index
Ulcerative colitis
TNFTumor necrosis factor
TDMTherapeutic drug monitoring

References

  1. Claßen, M.; de Laffolie, J.; Claßen, M.; Schnell, A.; Sohrabi, K.; Hoerning, A. Significant Advantages for First Line Treatment with TNF-Alpha Inhibitors in Pediatric Patients with Inflammatory Bowel Disease—Data from the Multicenter CEDATA-GPGE Registry Study. Front. Pediatr. 2022, 10, 903677. [Google Scholar] [CrossRef] [PubMed]
  2. van Rheenen, P.F.; Aloi, M.; Assa, A.; Bronsky, J.; Escher, J.C.; Fagerberg, U.L.; Gasparetto, M.; Gerasimidis, K.; Griffiths, A.; Henderson, P.; et al. The Medical Management of Paediatric Crohn’s Disease: An ECCO-ESPGHAN Guideline Update. J Crohns Colitis 2021, 15, 171–194. [Google Scholar] [CrossRef] [PubMed]
  3. Turner, D.; Ruemmele, F.M.; Orlanski-Meyer, E.; Griffiths, A.M.; de Carpi, J.M.; Bronsky, J.; Veres, G.; Aloi, M.; Strisciuglio, C.; Braegger, C.P.; et al. Management of Paediatric Ulcerative Colitis, Part 1: Ambulatory Care—An Evidence-Based Guideline from European Crohn’s and Colitis Organization and European Society of Paediatric Gastroenterology, Hepatology and Nutrition. J. Pediatr. Gastroenterol. Nutr. 2018, 67, 257–291. [Google Scholar] [CrossRef] [PubMed]
  4. Kaplan, J.L.; Liu, C.; King, E.C.; Bass, J.A.; Patel, A.S.; Tung, J.; Chen, S.; Lissoos, T.; Candela, N.; Saeed, S.; et al. Use, Durability, and Risks for Discontinuation of Initial and Subsequent Biologics in a Large Pediatric-Onset IBD Cohort. J. Pediatr. Gastroenterol. Nutr. 2023, e003734. [Google Scholar] [CrossRef] [PubMed]
  5. Cozijnsen, M.; Duif, V.; Kokke, F.; Kindermann, A.; van Rheenen, P.; de Meij, T.; Schaart, M.; Damen, G.; Norbruis, O.; Pelleboer, R.; et al. Adalimumab Therapy in Children with Crohn Disease Previously Treated With Infliximab. J. Pediatr. Gastroenterol. Nutr. 2015, 60, 205–210. [Google Scholar] [CrossRef]
  6. Danese, S.; Vuitton, L.; Peyrin-Biroulet, L. Biologic Agents for IBD: Practical Insights. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 537–545. [Google Scholar] [CrossRef]
  7. Garcia-Romero, R.; Martinez de Zabarte Fernandez, J.M.; Pujol-Muncunill, G.; Donat-Aliaga, E.; Segarra-Cantón, O.; Irastorza-Terradillos, I.; Medina-Benitez, E.; Ruiz-Hernández, C.J.; Carrillo-Palau, M.; Ros-Arnal, I.; et al. Safety and Effectiveness of Vedolizumab in Paediatric Patients with Inflammatory Bowel Disease: An Observational Multicentre Spanish Study. Eur. J. Pediatr. 2021, 180, 3029–3038. [Google Scholar] [CrossRef]
  8. Yerushalmy-Feler, A.; Pujol-Muncunill, G.; Martin-de-Carpi, J.; Kolho, K.-L.; Levine, A.; Olbjørn, C.; Granot, M.; Bramuzzo, M.; Rolandsdotter, H.; Mouratidou, N.; et al. Safety and Potential Efficacy of Escalating Dose of Ustekinumab in Pediatric Crohn Disease (the Speed-up Study): A Multicenter Study from the Pediatric IBD Porto Group of ESPGHAN. J. Pediatr. Gastroenterol. Nutr. 2022, 75, 717–723. [Google Scholar] [CrossRef]
  9. Dhaliwal, J.; McKay, H.E.; Deslandres, C.; Debruyn, J.; Wine, E.; Wu, A.; Huynh, H.; Carman, N.; Crowley, E.; Church, P.C.; et al. One-Year Outcomes with Ustekinumab Therapy in Infliximab-Refractory Paediatric Ulcerative Colitis: A Multicentre Prospective Study. Aliment. Pharmacol. Ther. 2021, 53, 1300–1308. [Google Scholar] [CrossRef]
  10. Dayan, J.R.; Dolinger, M.; Benkov, K.; Dunkin, D.; Jossen, J.; Lai, J.; Phan, B.L.; Pittman, N.; Dubinsky, M.C. Real World Experience with Ustekinumab in Children and Young Adults at a Tertiary Care Pediatric Inflammatory Bowel Disease Center. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 61–67. [Google Scholar] [CrossRef]
  11. deBruyn, J.C.; Jacobson, K.; El-Matary, W.; Carroll, M.; Wine, E.; Wrobel, I.; Van Woudenberg, M.; Huynh, H.Q. Long-Term Outcomes of Infliximab Use for Pediatric Crohn Disease: A Canadian Multicenter Clinical Practice Experience. J. Pediatr. Gastroenterol. Nutr. 2018, 66, 268–273. [Google Scholar] [CrossRef] [PubMed]
  12. Aardoom, M.A.; Veereman, G.; de Ridder, L. A Review on the Use of Anti-TNF in Children and Adolescents with Inflammatory Bowel Disease. Int. J. Mol. Sci. 2019, 20, 2529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Bronsky, J.; Copova, I.; Kazeka, D.; Lerchova, T.; Mitrova, K.; Pospisilova, K.; Sulovcova, M.; Zarubova, K.; Hradsky, O. Adalimumab vs Infliximab in Pediatric Patients with Crohn’s Disease: A Propensity Score Analysis and Predictors of Treatment Escalation. Clin. Transl. Gastroenterol. 2022, 13, e00490. [Google Scholar] [CrossRef] [PubMed]
  14. Björkesten, C.-G.a.; Nieminen, U.; Turunen, U.; Arkkila, P.E.; Sipponen, T.; Färkkilä, M.A. Endoscopic Monitoring of Infliximab Therapy in Crohn’s Disease. Inflamm. Bowel Dis. 2011, 17, 947–953. [Google Scholar] [CrossRef] [PubMed]
  15. Ribaldone, D.G.; Caviglia, G.P.; Abdulle, A.; Pellicano, R.; Ditto, M.C.; Morino, M.; Fusaro, E.; Saracco, G.M.; Bugianesi, E.; Astegiano, M. Adalimumab Therapy Improves Intestinal Dysbiosis in Crohn’s Disease. J. Clin. Med. 2019, 8, 1646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Scarallo, L.; Alvisi, P.; Bolasco, G.; Di Toma, M.; Lanari, M.; Cangiari, A.; Paci, M.; Naldini, S.; Renzo, S.; Barp, J.; et al. Mucosal and Histologic Healing in Children with Inflammatory Bowel Disease Treated With Antitumor Necrosis Factor-Alpha. J. Pediatr. Gastroenterol. Nutr. 2021, 72, 728–735. [Google Scholar] [CrossRef]
  17. Kierkus, J.; Dadalski, M.; Szymanska, E.; Oracz, G.; Wegner, A.; Gorczewska, M.; Szymanska, S.; Woynarowski, M.; Ryzko, J. The Impact of Infliximab Induction Therapy on Mucosal Healing and Clinical Remission in Polish Pediatric Patients with Moderate-to-Severe Crohn’s Disease. Eur. J. Gastroenterol. Hepatol. 2012, 24, 495–500. [Google Scholar] [CrossRef]
  18. Lee, D.; Baldassano, R.N.; Otley, A.R.; Albenberg, L.; Griffiths, A.M.; Compher, C.; Chen, E.Z.; Li, H.; Gilroy, E.; Nessel, L.; et al. Comparative Effectiveness of Nutritional and Biological Therapy in North American Children with Active Crohn’s Disease. Inflamm. Bowel Dis. 2015, 21, 1786–1793. [Google Scholar] [CrossRef]
  19. Luo, Y.; Yu, J.; Lou, J.; Fang, Y.; Chen, J. Exclusive Enteral Nutrition versus Infliximab in Inducing Therapy of Pediatric Crohn’s Disease. Gastroenterol. Res. Pract. 2017, 2017, e6595048. [Google Scholar] [CrossRef] [Green Version]
  20. van Rheenen, H.; van Rheenen, P.F. Long-Term Efficacy of Anti-Tumor Necrosis Factor Agents in Pediatric Luminal Crohn’s Disease: A Systematic Review of Real-World Evidence Studies. Pediatr. Gastroenterol. Hepatol. Nutr. 2020, 23, 121–131. [Google Scholar] [CrossRef]
  21. Carnovale, C.; Maffioli, A.; Zaffaroni, G.; Mazhar, F.; Battini, V.; Mosini, G.; Pozzi, M.; Radice, S.; Clementi, E.; Danelli, P. Efficacy of Tumour Necrosis Factor-Alpha Therapy in Paediatric Crohn’s Disease Patients with Perianal Lesions: A Systematic Review. Expert Opin. Biol. Ther. 2020, 20, 239–251. [Google Scholar] [CrossRef] [PubMed]
  22. Hyams, J.; Damaraju, L.; Blank, M.; Johanns, J.; Guzzo, C.; Winter, H.S.; Kugathasan, S.; Cohen, S.; Markowitz, J.; Escher, J.C.; et al. Induction and Maintenance Therapy with Infliximab for Children with Moderate to Severe Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 2012, 10, 391–399.e1. [Google Scholar] [CrossRef] [PubMed]
  23. Bolia, R.; Rajanayagam, J.; Hardikar, W.; Alex, G. Impact of Changing Treatment Strategies on Outcomes in Pediatric Ulcerative Colitis. Inflamm. Bowel Dis. 2019, 25, 1838–1844. [Google Scholar] [CrossRef] [PubMed]
  24. Croft, N.M.; Faubion, W.A.; Kugathasan, S.; Kierkus, J.; Ruemmele, F.M.; Shimizu, T.; Mostafa, N.M.; Venetucci, M.; Finney-Hayward, T.; Gonzalez, Y.S.; et al. Efficacy and Safety of Adalimumab in Paediatric Patients with Moderate-to-Severe Ulcerative Colitis (ENVISION I): A Randomised, Controlled, Phase 3 Study. Lancet Gastroenterol. Hepatol. 2021, 6, 616–627. [Google Scholar] [CrossRef] [PubMed]
  25. Ley, D.; Leroyer, A.; Dupont, C.; Sarter, H.; Bertrand, V.; Spyckerelle, C.; Guillon, N.; Wils, P.; Savoye, G.; Turck, D.; et al. New Therapeutic Strategies Have Changed the Natural History of Pediatric Crohn’s Disease: A Two-Decade Population-Based Study. Clin. Gastroenterol. Hepatol. 2022, 20, 2588–2597.e1. [Google Scholar] [CrossRef] [PubMed]
  26. Boros, K.K.; Veres, G.; Cseprekál, O.; Pintér, H.K.; Richter, É.; Cseh, Á.; Dezsőfi-Gottl, A.; Arató, A.; Reusz, G.; Dohos, D.; et al. Body Composition, Physical Activity, and Quality of Life in Pediatric Patients with Inflammatory Bowel Disease on Anti-TNF Therapy—An Observational Follow-up Study. Eur. J. Clin. Nutr. 2023, 77, 380–385. [Google Scholar] [CrossRef]
  27. D’Arcangelo, G.; Abi Nader, E.; Charbit-Henrion, F.; Talbotec, C.; Goulet, O.; Ruemmele, F.M.; Pigneur, B. Increased Use of Anti-Tumor Necrosis Factor Following the Implementation of the ECCO-ESPGHAN Guidelines and Its Impact on the Outcome of Pediatric Crohn’s Disease: A Retrospective Single-Center Study. J. Pediatr. Gastroenterol. Nutr. 2022, 74, 79–84. [Google Scholar] [CrossRef]
  28. Kim, H.J.; Oh, S.H.; Lee, S.H.; Kim, Y.-B.; Kim, D.Y.; Park, S.H.; Ye, B.D.; Yang, S.-K.; Kim, K.M. Risk Factors for Disease Behavior Evolution and Efficacy of Biologics in Reducing Progression in Pediatric Patients with Nonstricturing, Nonpenetrating Crohn’s Disease at Diagnosis: A Single-Center Experience in Korea. Gut Liver 2021, 15, 851–857. [Google Scholar] [CrossRef]
  29. Walters, T.D.; Kim, M.-O.; Denson, L.A.; Griffiths, A.M.; Dubinsky, M.; Markowitz, J.; Baldassano, R.; Crandall, W.; Rosh, J.; Pfefferkorn, M.; et al. Increased Effectiveness of Early Therapy with Anti-Tumor Necrosis Factor-α vs an Immunomodulator in Children with Crohn’s Disease. Gastroenterology 2014, 146, 383–391. [Google Scholar] [CrossRef]
  30. Choe, Y.J.; Han, K.; Shim, J.O. Treatment Patterns of Anti-Tumour Necrosis Factor-Alpha and Prognosis of Paediatric and Adult-Onset Inflammatory Bowel Disease in Korea: A Nationwide Population-Based Study. Aliment. Pharmacol. Ther. 2022, 56, 980–988. [Google Scholar] [CrossRef]
  31. Kugathasan, S.; Denson, L.A.; Walters, T.D.; Kim, M.-O.; Marigorta, U.M.; Schirmer, M.; Mondal, K.; Liu, C.; Griffiths, A.; Noe, J.D.; et al. Prediction of Complicated Disease Course for Children Newly Diagnosed with Crohn’s Disease: A Multicentre Inception Cohort Study. Lancet 2017, 389, 1710–1718. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kandavel, P.; Eder, S.J.; Adler, J.; The ImproveCareNow Network Pediatric IBD Learning Health. Reduced Systemic Corticosteroid Use among Pediatric Patients with Inflammatory Bowel Disease in a Large Learning Health System. J. Pediatr. Gastroenterol. Nutr. 2021, 73, 345–351. [Google Scholar] [CrossRef] [PubMed]
  33. Sherlock, M.E.; Zachos, M.; Issenman, R.M.; Mulder, D.J. Clinical and Laboratory Characteristics Are Associated with Biologic Therapy Use in Pediatric Inflammatory Bowel Disease: A Retrospective Cohort Study. J. Can. Assoc. Gastroenterol. 2021, 4, e92–e100. [Google Scholar] [CrossRef] [PubMed]
  34. Nuti, F.; Viola, F.; Civitelli, F.; Alessandri, C.; Aloi, M.; Dilillo, A.; Del Giudice, E.; Cucchiara, S. Biological Therapy in a Pediatric Crohn Disease Population at a Referral Center. J. Pediatr. Gastroenterol. Nutr. 2014, 58, 582–587. [Google Scholar] [CrossRef]
  35. Beukelman, T.; Xie, F.; Chen, L.; Horton, D.B.; Lewis, J.D.; Mamtani, R.; Mannion, M.M.; Saag, K.G.; Curtis, J.R. Risk of Malignancy Associated with Paediatric Use of Tumour Necrosis Factor Inhibitors. Anna. Rheum. Dis. 2018, 77, 1012–1016. [Google Scholar] [CrossRef]
  36. Hradsky, O.; Kazeka, D.; Copova, I.; Lerchova, T.; Mitrova, K.; Pospisilova, K.; Sulovcova, M.; Zarubova, K.; Bronsky, J. Risk Factors for Dermatological Complications of Anti-TNF Therapy in a Cohort of Children with Crohn’s Disease. Eur. J. Pediatr. 2021, 180, 3001–3008. [Google Scholar] [CrossRef]
  37. Dolinger, M.T.; Rolfes, P.; Spencer, E.; Stoffels, G.; Dunkin, D.; Dubinsky, M.C. Outcomes of Children with Inflammatory Bowel Disease Who Develop Anti-Tumour Necrosis Factor-Induced Skin Reactions. J. Crohns Colitis 2022, 16, 1420–1427. [Google Scholar] [CrossRef]
  38. Baggett, K.; Brandon, T.G.; Xiao, R.; Valenzuela, Z.; Buckley, L.H.; Weiss, P.F. Incidence Rates of Psoriasis in Children with Inflammatory Bowel Disease and Juvenile Arthritis Treated with Tumor Necrosis Factor Inhibitors and Disease-Modifying Antirheumatic Drugs. J. Rheumatol. 2022, 49, 935–941. [Google Scholar] [CrossRef]
  39. Zvuloni, M.; Matar, M.; Levi, R.; Shouval, D.S.; Shamir, R.; Assa, A. High Anti-TNFα Concentrations Are Not Associated with More Adverse Events in Pediatric Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2021, 73, 717–721. [Google Scholar] [CrossRef]
  40. Eindor-Abarbanel, A.; Meleady, L.; Lawrence, S.; Hamilton, Z.; Krikler, G.; Lakhani, A.; Zhang, Q.; Jacobson, K. Progression to Anti-TNF Treatment in Very Early Onset Inflammatory Bowel Disease Patients. J. Pediatr. Gastroenterol. Nutr. 2022, 75, 473–479. [Google Scholar] [CrossRef]
  41. Kerur, B.; Fiedler, K.; Stahl, M.; Hyams, J.; Stephens, M.; Lu, Y.; Pfefferkorn, M.; Alkhouri, R.; Strople, J.; Kelsen, J.; et al. Utilization of Antitumor Necrosis Factor Biologics in Very Early Onset Inflammatory Bowel Disease: A Multicenter Retrospective Cohort Study from North America. J. Pediatr. Gastroenterol. Nutr. 2022, 75, 64–69. [Google Scholar] [CrossRef] [PubMed]
  42. Kennedy, N.A.; Heap, G.A.; Green, H.D.; Hamilton, B.; Bewshea, C.; Walker, G.J.; Thomas, A.; Nice, R.; Perry, M.H.; Bouri, S.; et al. Predictors of Anti-TNF Treatment Failure in Anti-TNF-Naive Patients with Active Luminal Crohn’s Disease: A Prospective, Multicentre, Cohort Study. Lancet Gastroenterol. Hepatol. 2019, 4, 341–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Rodríguez Azor, B.; Martín-Masot, R.; Dayaldasani Khialani, A.; Fernández-Martín, J.M.; Gallego Fernández, C.; Navas-López, V.M. Proactive Monitoring of Anti-TNF Agents Improves Follow-up of Paediatric Patients with Crohn Disease. Anales de Pediatría (Engl. Ed.) 2023, 98, 165–174. [Google Scholar] [CrossRef]
  44. Salvador-Martín, S.; Zapata-Cobo, P.; Velasco, M.; Palomino, L.M.; Clemente, S.; Segarra, O.; Sánchez, C.; Tolín, M.; Moreno-Álvarez, A.; Fernández-Lorenzo, A.; et al. Association between HLA DNA Variants and Long-Term Response to Anti-TNF Drugs in a Spanish Pediatric Inflammatory Bowel Disease Cohort. Int. J. Mol. Sci. 2023, 24, 1797. [Google Scholar] [CrossRef] [PubMed]
  45. Cohen, R.Z.; Schoen, B.T.; Kugathasan, S.; Sauer, C.G. Management of Anti-Drug Antibodies to Biologic Medications in Children With Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 551. [Google Scholar] [CrossRef]
  46. Colman, R.J.; Portocarrero-Castillo, A.; Chona, D.; Hellmann, J.; Minar, P.; Rosen, M.J. Favorable Outcomes and Anti-TNF Durability After Addition of an Immunomodulator for Anti-Drug Antibodies in Pediatric IBD Patients. Inflamm. Bowel Dis. 2021, 27, 507–515. [Google Scholar] [CrossRef]
  47. Sassine, S.; Djani, L.; Cambron-Asselin, C.; Savoie, M.; Lin, Y.F.; Qaddouri, M.; Zekhnine, S.; Grzywacz, K.; Groleau, V.; Dirks, M.; et al. Risk Factors of Clinical Relapses in Pediatric Luminal Crohn’s Disease: A Retrospective Cohort Study. Am. J. Gastroenterol. 2022, 117, 637–646. [Google Scholar] [CrossRef]
  48. Scarallo, L.; Bolasco, G.; Barp, J.; Bianconi, M.; di Paola, M.; Di Toma, M.; Naldini, S.; Paci, M.; Renzo, S.; Labriola, F.; et al. Anti-Tumor Necrosis Factor-Alpha Withdrawal in Children with Inflammatory Bowel Disease in Endoscopic and Histologic Remission. Inflamm. Bowel Dis. 2022, 28, 183–191. [Google Scholar] [CrossRef]
  49. Weigl, E.; Schwerd, T.; Lurz, E.; Häberle, B.; Koletzko, S.; Hubertus, J. Children with Localized Crohn’s Disease Benefit from Early Ileocecal Resection and Perioperative Anti-Tumor Necrosis Factor Therapy. Eur. J. Pediatr. Surg. 2023. [Google Scholar] [CrossRef]
  50. Jongsma, M.M.E.; Aardoom, M.A.; Cozijnsen, M.A.; van Pieterson, M.; de Meij, T.; Groeneweg, M.; Norbruis, O.F.; Wolters, V.M.; van Wering, H.M.; Hojsak, I.; et al. First-Line Treatment with Infliximab versus Conventional Treatment in Children with Newly Diagnosed Moderate-to-Severe Crohn’s Disease: An Open-Label Multicentre Randomised Controlled Trial. Gut 2020, 71, 34–42. [Google Scholar] [CrossRef]
  51. Jongsma, M.M.E.; Winter, D.A.; Huynh, H.Q.; Norsa, L.; Hussey, S.; Kolho, K.-L.; Bronsky, J.; Assa, A.; Cohen, S.; Lev-Tzion, R.; et al. Infliximab in Young Paediatric IBD Patients: It Is All about the Dosing. Eur. J. Pediatr. 2020, 179, 1935–1944. [Google Scholar] [CrossRef] [PubMed]
  52. Church, P.C.; Ho, S.; Sharma, A.; Tomalty, D.; Frost, K.; Muise, A.; Walters, T.D.; Griffiths, A.M. Intensified Infliximab Induction Is Associated with Improved Response and Decreased Colectomy in Steroid-Refractory Paediatric Ulcerative Colitis. J. Crohn’s Colitis 2019, 13, 982–989. [Google Scholar] [CrossRef] [PubMed]
  53. van Hoeve, K.; Dreesen, E.; Hoffman, I.; Van Assche, G.; Ferrante, M.; Gils, A.; Vermeire, S. Adequate Infliximab Exposure During Induction Predicts Remission in Paediatric Patients with Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2019, 68, 847–853. [Google Scholar] [CrossRef] [PubMed]
  54. Schnell, A.; Schwarz, B.; Wahlbuhl, M.; Allabauer, I.; Hess, M.; Weber, S.; Werner, F.; Schmidt, H.; Rechenauer, T.; Siebenlist, G.; et al. Distribution and Cytokine Profile of Peripheral B Cell Subsets Is Perturbed in Pediatric IBD and Partially Restored During a Successful IFX Therapy. Inflamm. Bowel Dis. 2021, 27, 224–235. [Google Scholar] [CrossRef]
  55. Cheifetz, A.S.; Vande Casteele, N.; Wang, Z.; Dubinsky, M.C.; Papamichael, K. Higher Postinduction Infliximab Concentrations Are Associated with Favorable Clinical Outcomes in Pediatric Crohn’s Disease: A Post Hoc Analysis of the REACH Trial. AJG Am. J. Gastroenterol. 2023, 118, 485–490. [Google Scholar] [CrossRef]
  56. Lawrence, S.; Faytrouni, F.; Harris, R.E.; Irvine, M.; Carrion, E.; Scott, G.; Clarke, B.; Garrick, V.; Curtis, L.; Gervais, L.; et al. Optimized Infliximab Induction Predicts Better Long-Term Clinical and Biomarker Outcomes Compared to Standard Induction Dosing. J. Pediatr. Gastroenterol. Nutr. 2022, 75, 601–607. [Google Scholar] [CrossRef]
  57. Chung, A.; Carroll, M.; Almeida, P.; Petrova, A.; Isaac, D.; Mould, D.; Wine, E.; Huynh, H. Early Infliximab Clearance Predicts Remission in Children with Crohn’s Disease. Dig. Dis. Sci. 2022. [Google Scholar] [CrossRef]
  58. Kwon, Y.; Kim, E.-S.; Kim, Y.-Z.; Choe, Y.-H.; Kim, M.-J. Cytokine Profile at Diagnosis Affecting Trough Concentration of Infliximab in Pediatric Crohn’s Disease. Biomedicines 2022, 10, 2372. [Google Scholar] [CrossRef]
  59. Constant, B.D.; Khushal, S.; Jiang, J.; Bost, J.E.; Chaisson, E.; Conklin, L.S. Early Inflammatory Markers Are Associated with Inadequate Post-Induction Infliximab Trough in Pediatric Crohn’s Disease. J. Pediatr. Gastroenterol. Nutr. 2021, 72, 410–416. [Google Scholar] [CrossRef]
  60. Merras-Salmio, L.; Kolho, K.-L. Clinical Use of Infliximab Trough Levels and Antibodies to Infliximab in Pediatric Patients with Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 272. [Google Scholar] [CrossRef]
  61. Dave, M.B.; Dherai, A.J.; Desai, D.C.; Mould, D.R.; Ashavaid, T.F. Optimization of Infliximab Therapy in Inflammatory Bowel Disease Using a Dashboard Approach-an Indian Experience. Eur. J. Clin. Pharmacol. 2021, 77, 55–62. [Google Scholar] [CrossRef] [PubMed]
  62. Curci, D.; Lucafò, M.; Cifù, A.; Fabris, M.; Bramuzzo, M.; Martelossi, S.; Franca, R.; Decorti, G.; Stocco, G. Pharmacogenetic Variants of Infliximab Response in Young Patients with Inflammatory Bowel Disease. Clin. Transl. Sci. 2021, 14, 2184–2192. [Google Scholar] [CrossRef] [PubMed]
  63. Clarkston, K.; Tsai, Y.-T.; Jackson, K.; Rosen, M.J.; Denson, L.A.; Minar, P. Development of Infliximab Target Concentrations During Induction in Pediatric Crohn Disease Patients. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 68–74. [Google Scholar] [CrossRef] [PubMed]
  64. El-Matary, W.; Walters, T.D.; Huynh, H.Q.; deBruyn, J.; Mack, D.R.; Jacobson, K.; Sherlock, M.E.; Church, P.; Wine, E.; Carroll, M.W.; et al. Higher Postinduction Infliximab Serum Trough Levels Are Associated with Healing of Fistulizing Perianal Crohn’s Disease in Children. Inflamm. Bowel Dis. 2019, 25, 150–155. [Google Scholar] [CrossRef] [PubMed]
  65. Stein, R.; Lee, D.; Leonard, M.B.; Thayu, M.; Denson, L.A.; Chuang, E.; Herskovitz, R.; Kerbowski, T.; Baldassano, R.N. Serum Infliximab, Antidrug Antibodies, and Tumor Necrosis Factor Predict Sustained Response in Pediatric Crohn’s Disease. Inflamm. Bowel Dis. 2016, 22, 1370–1377. [Google Scholar] [CrossRef]
  66. Drobne, D.; Kurent, T.; Golob, S.; Svegl, P.; Rajar, P.; Terzic, S.; Kozelj, M.; Novak, G.; Smrekar, N.; Plut, S.; et al. Success and Safety of High Infliximab Trough Levels in Inflammatory Bowel Disease. Scand. J. Gastroenterol. 2018, 53, 940–946. [Google Scholar] [CrossRef]
  67. Courbette, O.; Aupiais, C.; Viala, J.; Hugot, J.-P.; Roblin, X.; Candon, S.; Louveau, B.; Chatenoud, L.; Martinez-Vinson, C. Trough Levels of Infliximab at Week 6 Are Predictive of Remission at Week 14 in Pediatric Crohn’s Disease. J. Pediatr. Gastroenterol. Nutr. 2020, 70, 310–317. [Google Scholar] [CrossRef]
  68. Crombé, V.; Salleron, J.; Savoye, G.; Dupas, J.-L.; Vernier-Massouille, G.; Lerebours, E.; Cortot, A.; Merle, V.; Vasseur, F.; Turck, D.; et al. Long-Term Outcome of Treatment with Infliximab in Pediatric-Onset Crohn’s Disease: A Population-Based Study. Inflamm. Bowel Dis. 2011, 17, 2144–2152. [Google Scholar] [CrossRef]
  69. Assa, A.; Matar, M.; Turner, D.; Broide, E.; Weiss, B.; Ledder, O.; Guz-Mark, A.; Rinawi, F.; Cohen, S.; Topf-Olivestone, C.; et al. Proactive Monitoring of Adalimumab Trough Concentration Associated with Increased Clinical Remission in Children with Crohn’s Disease Compared With Reactive Monitoring. Gastroenterology 2019, 157, 985–996.e2. [Google Scholar] [CrossRef]
  70. Matar, M.; Shamir, R.; Turner, D.; Broide, E.; Weiss, B.; Ledder, O.; Guz-Mark, A.; Rinawi, F.; Cohen, S.; Topf-Olivestone, C.; et al. Combination Therapy of Adalimumab with an Immunomodulator Is Not More Effective Than Adalimumab Monotherapy in Children with Crohn’s Disease: A Post Hoc Analysis of the PAILOT Randomized Controlled Trial. Inflamm. Bowel Dis. 2020, 26, 1627–1635. [Google Scholar] [CrossRef]
  71. Dubinsky, M.C.; Rosh, J.; Faubion, W.A.; Kierkus, J.; Ruemmele, F.; Hyams, J.S.; Eichner, S.; Li, Y.; Huang, B.; Mostafa, N.M.; et al. Efficacy and Safety of Escalation of Adalimumab Therapy to Weekly Dosing in Pediatric Patients with Crohn’s Disease. Inflamm. Bowel Dis. 2016, 22, 886–893. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Payen, E.; Neuraz, A.; Zenzeri, L.; Talbotec, C.; Abi Nader, E.; Chatenoud, L.; Chhun, S.; Goulet, O.; Ruemmele, F.M.; Pigneur, B. Adalimumab Therapy in Pediatric Crohn Disease: A 2-Year Follow-Up Comparing “Top-Down” and “Step-Up” Strategies. J. Pediatr. Gastroenterol. Nutr. 2023, 76, 166. [Google Scholar] [CrossRef] [PubMed]
  73. Lucafò, M.; Curci, D.; Bramuzzo, M.; Alvisi, P.; Martelossi, S.; Silvestri, T.; Guastalla, V.; Labriola, F.; Stocco, G.; Decorti, G. Serum Adalimumab Levels After Induction Are Associated with Long-Term Remission in Children with Inflammatory Bowel Disease. Front. Pediatr. 2021, 9, 244. [Google Scholar] [CrossRef] [PubMed]
  74. Hyams, J.S.; Dubinsky, M.C.; Baldassano, R.N.; Colletti, R.B.; Cucchiara, S.; Escher, J.; Faubion, W.; Fell, J.; Gold, B.D.; Griffiths, A.; et al. Infliximab Is Not Associated with Increased Risk of Malignancy or Hemophagocytic Lymphohistiocytosis in Pediatric Patients with Inflammatory Bowel Disease. Gastroenterology 2017, 152, 1901–1914.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Atia, O.; Shavit-Brunschwig, Z.; Mould, D.R.; Stein, R.; Matar, M.; Aloi, M.; Ledder, O.; Focht, G.; Urlep, D.; Hyams, J.; et al. Outcomes, Dosing, and Predictors of Vedolizumab Treatment in Children with Inflammatory Bowel Disease (VEDOKIDS): A Prospective, Multicentre Cohort Study. Lancet Gastroenterol. Hepatol. 2023, 8, 31–42. [Google Scholar] [CrossRef] [PubMed]
  76. Ungaro, R.C.; Yarur, A.; Jossen, J.; Phan, B.L.; Chefitz, E.; Sehgal, P.; Kamal, K.; Bruss, A.; Beniwal-Patel, P.; Fox, C.; et al. Higher Trough Vedolizumab Concentrations During Maintenance Therapy Are Associated with Corticosteroid-Free Remission in Inflammatory Bowel Disease. J. Crohn’s Colitis 2019, 13, 963–969. [Google Scholar] [CrossRef]
  77. Colman, R.J.; Mizuno, T.; Fukushima, K.; Haslam, D.B.; Hyams, J.S.; Boyle, B.; Noe, J.D.; D’Haens, G.R.; Van Limbergen, J.; Chun, K.; et al. Real World Population Pharmacokinetic Study in Children and Young Adults with Inflammatory Bowel Disease Discovers Novel Blood and Stool Microbial Predictors of Vedolizumab Clearance. Aliment. Pharmacol. Ther. 2023, 57, 524–539. [Google Scholar] [CrossRef]
  78. Jansson, S.; Malham, M.; Paerregaard, A.; Jakobsen, C.; Wewer, V. Extraintestinal Manifestations Are Associated with Disease Severity in Pediatric Onset Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2020, 71, 40–45. [Google Scholar] [CrossRef]
  79. D’Arcangelo, G.; Distante, M.; Raso, T.; Rossetti, D.; Catassi, G.; Aloi, M. Safety of Biological Therapy in Children with Inflammatory Bowel Disease. J. Pediatr. Gastroenterol. Nutr. 2021, 72, 736–741. [Google Scholar] [CrossRef]
  80. Russi, L.; Scharl, M.; Rogler, G.; Biedermann, L. The Efficacy and Safety of Golimumab as Third- or Fourth-Line Anti-TNF Therapy in Patients with Refractory Crohn’s Disease: A Case Series. IID 2017, 2, 131–138. [Google Scholar] [CrossRef] [Green Version]
  81. Conrad, M.A.; Kelsen, J.R. The Treatment of Pediatric Inflammatory Bowel Disease with Biologic Therapies. Curr. Gastroenterol. Rep. 2020, 22, 36. [Google Scholar] [CrossRef] [PubMed]
  82. Roda, G.; Jharap, B.; Neeraj, N.; Colombel, J.-F. Loss of Response to Anti-TNFs: Definition, Epidemiology, and Management. Clin. Transl. Gastroenterol. 2016, 7, e135. [Google Scholar] [CrossRef] [PubMed]
  83. Shin, J.-Y.; Park, H.-M.; Lee, M.-Y.; Jeon, J.-Y.; Yoo, H.-J.; Ye, B.D. Real-World Incidence of Suboptimal Response to Anti-Tumor Necrosis Factor Therapy for Ulcerative Colitis: A Nationwide Population-Based Study. Gut Liver 2021, 15, 867–877. [Google Scholar] [CrossRef] [PubMed]
  84. Turner, D.; Ricciuto, A.; Lewis, A.; D’Amico, F.; Dhaliwal, J.; Griffiths, A.M.; Bettenworth, D.; Sandborn, W.J.; Sands, B.E.; Reinisch, W.; 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. [Google Scholar] [CrossRef] [PubMed]
  85. Feuerstein, J.D.; Nguyen, G.C.; Kupfer, S.S.; Falck-Ytter, Y.; Singh, S.; American Gastroenterological Association Institute Clinical Guidelines Committee. American Gastroenterological Association Institute Guideline on Therapeutic Drug Monitoring in Inflammatory Bowel Disease. Gastroenterology 2017, 153, 827–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Gomollón, F.; Dignass, A.; Annese, V.; Tilg, H.; Van Assche, G.; Lindsay, J.O.; Peyrin-Biroulet, L.; Cullen, G.J.; Daperno, M.; Kucharzik, T.; et al. 3rd European Evidence-Based Consensus on the Diagnosis and Management of Crohn’s Disease 2016: Part 1: Diagnosis and Medical Management. J. Crohns Colitis 2017, 11, 3–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Lichtenstein, G.R.; Loftus, E.V.; Isaacs, K.L.; Regueiro, M.D.; Gerson, L.B.; Sands, B.E. ACG Clinical Guideline: Management of Crohn’s Disease in Adults. Am. J. Gastroenterol. 2018, 113, 481–517. [Google Scholar] [CrossRef]
  88. Mitrev, N.; Vande Casteele, N.; Seow, C.H.; Andrews, J.M.; Connor, S.J.; Moore, G.T.; Barclay, M.; Begun, J.; Bryant, R.; Chan, W.; et al. Review Article: Consensus Statements on Therapeutic Drug Monitoring of Anti-Tumour Necrosis Factor Therapy in Inflammatory Bowel Diseases. Aliment. Pharmacol. Ther. 2017, 46, 1037–1053. [Google Scholar] [CrossRef] [Green Version]
  89. Steinhart, A.H.; Panaccione, R.; Targownik, L.; Bressler, B.; Khanna, R.; Marshall, J.K.; Afif, W.; Bernstein, C.N.; Bitton, A.; Borgaonkar, M.; et al. Clinical Practice Guideline for the Medical Management of Perianal Fistulizing Crohn’s Disease: The Toronto Consensus. Inflamm. Bowel Dis. 2019, 25, 1–13. [Google Scholar] [CrossRef] [Green Version]
  90. Cheifetz, A.S.; Abreu, M.T.; Afif, W.; Cross, R.K.; Dubinsky, M.C.; Loftus, E.V.; Osterman, M.T.; Saroufim, A.; Siegel, C.A.; Yarur, A.J.; et al. A Comprehensive Literature Review and Expert Consensus Statement on Therapeutic Drug Monitoring of Biologics in Inflammatory Bowel Disease. Am. J. Gastroenterol. 2021, 116, 2014–2025. [Google Scholar] [CrossRef]
  91. Syversen, S.W.; Jørgensen, K.K.; Goll, G.L.; Brun, M.K.; Sandanger, Ø.; Bjørlykke, K.H.; Sexton, J.; Olsen, I.C.; Gehin, J.E.; Warren, D.J.; et al. Effect of Therapeutic Drug Monitoring vs Standard Therapy During Maintenance Infliximab Therapy on Disease Control in Patients with Immune-Mediated Inflammatory Diseases: A Randomized Clinical Trial. JAMA 2021, 326, 2375–2384. [Google Scholar] [CrossRef]
  92. Yarur, A.J.; Kanagala, V.; Stein, D.J.; Czul, F.; Quintero, M.A.; Agrawal, D.; Patel, A.; Best, K.; Fox, C.; Idstein, K.; et al. Higher Infliximab Trough Levels Are Associated with Perianal Fistula Healing in Patients with Crohn’s Disease. Aliment. Pharmacol. Ther. 2017, 45, 933–940. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Ordás, I.; Mould, D.R.; Feagan, B.G.; Sandborn, W.J. Anti-TNF Monoclonal Antibodies in Inflammatory Bowel Disease: Pharmacokinetics-Based Dosing Paradigms. Clin. Pharmacol. Ther. 2012, 91, 635–646. [Google Scholar] [CrossRef] [PubMed]
  94. Yarur, A.J.; Jain, A.; Sussman, D.A.; Barkin, J.S.; Quintero, M.A.; Princen, F.; Kirkland, R.; Deshpande, A.R.; Singh, S.; Abreu, M.T. The Association of Tissue Anti-TNF Drug Levels with Serological and Endoscopic Disease Activity in Inflammatory Bowel Disease: The ATLAS Study. Gut 2016, 65, 249–255. [Google Scholar] [CrossRef] [PubMed]
  95. Brandse, J.F.; van den Brink, G.R.; Wildenberg, M.E.; van der Kleij, D.; Rispens, T.; Jansen, J.M.; Mathôt, R.A.; Ponsioen, C.Y.; Löwenberg, M.; D’Haens, G.R.A.M. Loss of Infliximab into Feces Is Associated With Lack of Response to Therapy in Patients With Severe Ulcerative Colitis. Gastroenterology 2015, 149, 350–355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Winter, D.A.; Joosse, M.E.; de Wildt, S.N.; Taminiau, J.; de Ridder, L.; Escher, J.C. Pharmacokinetics, Pharmacodynamics, and Immunogenicity of Infliximab in Pediatric Inflammatory Bowel Disease: A Systematic Review and Revised Dosing Considerations. J. Pediatr. Gastroenterol. Nutr. 2020, 70, 763–776. [Google Scholar] [CrossRef]
  97. Turner, D.; Ruemmele, F.M.; Orlanski-Meyer, E.; Griffiths, A.M.; De Carpi, J.M.; Bronsky, J.; Veres, G.; Aloi, M.; Strisciuglio, C.; Braegger, C.P.; et al. Management of Paediatric Ulcerative Colitis, Part 2: Acute Severe Colitis—An Evidence-Based Consensus Guideline from the European Crohn’s and Colitis Organization and the European Society of Paediatric Gastroenterology, Hepatology and Nutrition. J. Pediatr. Gastroenterol. Nutr. 2018, 67, 292–310. [Google Scholar] [CrossRef]
  98. Desai, D.C.; Dherai, A.J.; Strik, A.; Mould, D.R. Personalized Dosing of Infliximab in Patients with Inflammatory Bowel Disease Using a Bayesian Approach: A Next Step in Therapeutic Drug Monitoring. J. Clin. Pharmacol. 2023, 63, 480–489. [Google Scholar] [CrossRef]
  99. Irving, P.M.; Gecse, K.B. Optimizing Therapies Using Therapeutic Drug Monitoring: Current Strategies and Future Perspectives. Gastroenterology 2022, 162, 1512–1524. [Google Scholar] [CrossRef]
  100. Bodini, G.; Giannini, E.G.; Savarino, V.; Del Nero, L.; Pellegatta, G.; De Maria, C.; Baldissarro, I.; Savarino, E. Adalimumab Trough Serum Levels and Anti-Adalimumab Antibodies in the Long-Term Clinical Outcome of Patients with Crohn’s Disease. Scand. J. Gastroenterol. 2016, 51, 1081–1086. [Google Scholar] [CrossRef]
  101. Hyams, J.S.; Chan, D.; Adedokun, O.J.; Padgett, L.; Turner, D.; Griffiths, A.; Veereman, G.; Heyman, M.B.; Rosh, J.R.; Wahbeh, G.; et al. Subcutaneous Golimumab in Pediatric Ulcerative Colitis: Pharmacokinetics and Clinical Benefit. Inflamm. Bowel Dis. 2017, 23, 2227–2237. [Google Scholar] [CrossRef] [PubMed]
  102. Dreesen, E.; Verstockt, B.; Bian, S.; de Bruyn, M.; Compernolle, G.; Tops, S.; Noman, M.; Van Assche, G.; Ferrante, M.; Gils, A.; et al. Evidence to Support Monitoring of Vedolizumab Trough Concentrations in Patients with Inflammatory Bowel Diseases. Clin. Gastroenterol. Hepatol. 2018, 16, 1937–1946.e8. [Google Scholar] [CrossRef] [PubMed]
  103. Mechie, N.-C.; Burmester, M.; Mavropoulou, E.; Pilavakis, Y.; Kunsch, S.; Ellenrieder, V.; Amanzada, A. Evaluation of Ustekinumab Trough Levels during Induction and Maintenance Therapy with Regard to Disease Activity Status in Difficult to Treat Crohn Disease Patients. Medicine 2021, 100, e25111. [Google Scholar] [CrossRef]
  104. Baldassano, R.N.; Han, P.D.; Jeshion, W.C.; Berlin, J.A.; Piccoli, D.A.; Lautenbach, E.; Mick, R.; Lichtenstein, G.R. Pediatric Crohn’s Disease: Risk Factors for Postoperative Recurrence. Am. J. Gastroenterol. 2001, 96, 2169–2176. [Google Scholar] [CrossRef]
  105. Amil-Dias, J.; Kolacek, S.; Turner, D.; Pærregaard, A.; Rintala, R.; Afzal, N.A.; Karolewska-Bochenek, K.; Bronsky, J.; Chong, S.; Fell, J.; et al. Surgical Management of Crohn Disease in Children: Guidelines from the Paediatric IBD Porto Group of ESPGHAN. J. Pediatr. Gastroenterol. Nutr. 2017, 64, 818–835. [Google Scholar] [CrossRef] [PubMed]
  106. Regueiro, M.; Feagan, B.G.; Zou, B.; Johanns, J.; Blank, M.A.; Chevrier, M.; Plevy, S.; Popp, J.; Cornillie, F.J.; Lukas, M.; et al. Infliximab Reduces Endoscopic, but Not Clinical, Recurrence of Crohn’s Disease After Ileocolonic Resection. Gastroenterology 2016, 150, 1568–1578. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Regueiro, M.; Schraut, W.; Baidoo, L.; Kip, K.E.; Sepulveda, A.R.; Pesci, M.; Harrison, J.; Plevy, S.E. Infliximab Prevents Crohn’s Disease Recurrence after Ileal Resection. Gastroenterology 2009, 136, 441–450.e1. [Google Scholar] [CrossRef] [Green Version]
  108. De Cruz, P.; Kamm, M.A.; Hamilton, A.L.; Ritchie, K.J.; Krejany, E.O.; Gorelik, A.; Liew, D.; Prideaux, L.; Lawrance, I.C.; Andrews, J.M.; et al. Efficacy of Thiopurines and Adalimumab in Preventing Crohn’s Disease Recurrence in High-Risk Patients—A POCER Study Analysis. Aliment. Pharmacol. Ther. 2015, 42, 867–879. [Google Scholar] [CrossRef] [Green Version]
  109. Bouhuys, M.; Lexmond, W.S.; Dijkstra, G.; Lobatón, T.; Louis, E.; van Biervliet, S.; Groen, H.; Guardiola, J.; Rheenen, P. van Efficacy of Anti-TNF Dosing Interval Lengthening in Adolescents and Young Adults with Inflammatory Bowel Disease in Sustained Remission (FREE-Study): Protocol for a Partially Randomised Patient Preference Trial. BMJ Open 2021, 11, e054154. [Google Scholar] [CrossRef]
Figure 1. Overview of pathophysiological mechanisms and the effects of biologic agents currently used in pediatric IBD.
Figure 1. Overview of pathophysiological mechanisms and the effects of biologic agents currently used in pediatric IBD.
Children 10 00634 g001
Table 1. All included studies with pediatric patients and biologics.
Table 1. All included studies with pediatric patients and biologics.
StudyN TNFAge (Yrs)
(CD, UC)
m % (CD, UC)Study TypeObservation PeriodCountryIBD TypeBiologicTime from Diagnosis to BiologicComparison-GroupPost-AssessmentsOutcomesResults
Claßen et al., 2022 [1]48711.959.1Retrospective registry2004–2020Germany, Austria, SwitzerlandCD, CU, IBDuall19 monthsFirst-line vs. Second line Laboratory markers, clinical scores, side effects, treatment failurePatients with CD significantly benefitted from early treatment, with lower clinical scores, fewer EIMs and lower risk for treatment failure
D’Arcangelo et al., 2021 [27]1851358Retrospective, observational cohort
Single-center
2012–2020ItalyCD, UC, IBDuIFX, ADL, UST, VEDO2 yrs Immediate and delayed AEs32.8% biologic-related Aes
10% immediate reactions, 45% delayed
14% treatment discontinuation because of AEs
Kaplan et al., 2023 [4]17,649 Retrospective, observational cohort2006–2016USACD, UCall Use, discontinuation43% of pediatric IBD patients treated with biologic, more likely for CD, discontinuation significantly higher in UC
TNF-α inhibitors
Bronsky et al., 2022 [13]6211.64–16.2755–68Prospective observational cohort2013–2017Czech RepublicCDIFX, ADL0.6–1.04 yrsIFX vs. ADLUp to 24 monthsTreatment escalation
Non-response
Serious AEs
No difference between IFX and ADL in efficacy and safety
Lee et al., 2015 [18]5213.946Observational cohort Canada, USACDIFX (1xADL)0.7 yrEEN (n = 22), PEN (n = 16)8 weeksPCDAI, QoL, mucosal healing via FCPClinical response: 64% PEN, 88% EEN, 84% TNFi
Mucosal healing: PEN 14%, EEN 45%, TNFi 62%
QoL not statistically significant
Scarallo et al., 2021 [16]13410.9, 10.365.4,
50
retrospective, observational
(two centers)
2008–2018Italy78 (CD) 56 (UC)IFX, ADL Endoscopically assessed mucosal remissionMucosal remission in 41% of CD patients and 53.6% of UC patients, histological remission in 33.3% of CD patients and 39.3% of UC patients
Boros et al., 2023 [26]3215.2, 16.449Prospective, observational follow up
Single-center
2016–2018HungaryCD, UCTNFi1.4 yrs, 3 yrsHealthy controls2 & 6 monthsBody composition, health-related quality of life, physical activityBody composition and physical activity significantly improved after 6 months and caught up to healthy controls, no change in health-related quality of life
58% of CD 37.5% of UC patients in remission
Kim et al., 2021 [28]841574.1Retrospective single-center2000–2013KoreaCDTNFi Thiopurine treatment (N = 287)Up to 13 yrsDisease behavior evolutionEarly treatment (within 3 months after diagnosis) was associated with lower risk of disease behavior progression
Walters et al., 2014 [29]6811.861Retrospective multicenter2008–2012North AmericaCDIFX, ADLWithin 3 monthsImmunomodulator (IM) (N = 68), no IM (N 68) Steroid-free and surgery-free remission, growth85.3% in remission with TNFi, significantly more than other groups, growth improved in biologic group only
Ley et al., 2022 [25]1007 Retrospective
multicenter
1988–2011FranceCDTNFi M = 8.8 yrsIntestinal resection, disease progression,
hospitalizations
Reduction in intestinal resection and disease progression, no change in hospitalization over time
Choe et al., 2022 [30] Pediatric + adult Population-based2006–2015KoreaCD, UCTNFi TNFi prescription, fistulectomy, surgeryLower odds of surgery in CD patients under TNFi therapy
Kugathasan et al., 2017 [31]913 Prospective inception cohort2008–2012USA, Canada (28 sites)CDTNFi Disease complicationsEarly TNFi admission reduced risk for penetrating complications but not stricturing complications
Kandavel et al., 2021 [32]27,321 Retrospective cohort, multicenter2007–2018US, UK, QatarCD, UC, IBDuTNFi Use of corticosteroidsAppliance of TNFi within the first 120 days after diagnosis reduces risk for need of steroids later in CD not in UC
Sherlock et al., 2021 [33]19810.559.1Retrospective cohort,
single-center
2001–2015CanadaCD, UC, IBDu 21.5 months M = 47.8 Biologic therapy associated with older age, higher PCDAI/ PUCAI hypoalbuminemia in UC and CD
Nuti et al., 2014 [34]781563Single-center cohort2001–2011ItalyCDIFX, ADL40.6 months 1, 2, 3 yrsClinical activity (PCDAI), discontinuation, AEs81% continuation yr 1, 54% yr 2, 33% yr 3, no serious AEs
Beukelmann et al., 2018 [35]6808 43Retrospective, cohort USIBD, JIA, PsATNFi No TNFi use (N = 20,049) MalignanciesTNFi use in combination with thiopurines increased the risk for malignancies
Hradsky et al., 2021 [36]1001557–65Retrospective CDTNFi Skin complicationsAfter 2 yrs of treatment 35% of patients developed at least one skin complication
Dolinger et al., 2022 [37]638 Retrospective IFX, ADL 6 monthsSkin reactions21% infliximab patients, 11% adalimumab patients
Baggett et al., 2022 [38]3794 Retrospective2008–2020 IFX, ADL, etanercept Non-TNFi exposure Incidence of psoriasisHigher risk of psoriasis in patients treated with TNFi (highest in adalimumab)
Zvuloni et al., 2021 [39]13512.956.3Retrospective, cohort
single-center
2015–2020IsraelCD, UCIFX, ADL MD = 1.7 yrsIncidence of AEs37% of patients had AEs, psoriatiform rashes (45%), elevated transaminases (32%) and infusion reactions (13%)
Eindor-Abarbanel et al., 2022 [40]893.862.8Retrospective2005–2019United statesVEO IBDTNFi TNFi-naive1 yrDisease course, dose, and dose interval of IFX39.5% of VEO IBD patients received TNFi, higher disease activity was associated with TNFi-treatment, clinical remission on first biologic agent in 61,8%
Kerur et al., 2022 [41]294 Retrospective, cohort
Multicenter
2008–2013North AmericaVEO IBDIFX, ADL 1, 3, 5 yrsUtilization and durability of TNFi55% of patients treated with TNFi between 0–6 yrs old, durability 90% after one yr, 55% after 5 yrs, lower durability in UC und IBDu
Kennedy et al., 2019 [42]219 pediatricAdult + pediatric49Prospective, observational cohort, multicenter2013–2016UK, Korea, USACDIFX, ADL2.3–3.3 yrs 12 monthsDisease activity, AEs, discontinuation, treatment failure, anti-drug antibodiesLow drug concentration the only predictor for primary non-response in week 14, and remission by week 54
62.8% ADAs in IFX, 28.5% in ADL predicted by suboptimal drug concentration in week 14
Rodriguez Azor et al., 2023 [43]3011.370Prospective observational2015–2020 CDIFX, ADL9.9 months M = 27.1 monthsClinical remission, mucosal healing, laboratory markers87.1% in clinical remission (wPCDAI), 83% achieved mucosal healing (MINI)
Salvador-Martín et al., 2023 [44]34011.260.3Observational, multicenter SpainCD, UC, IBDuIFX, ADL6.1 monthsResponders vs. non-responders9 yrsTreatmtent failureOnly in adults association of HLA polymorphisms and treatment failure
Cohen et al., 2019 [45]2341354.2Retrospective, single-center USACD, UCIFX, ADL With and without ADAs ADAs24.8% developed ADAs, 48% of those underwent dose optimization and of those 54% had undetectable ADAs on follow-up,
Patients switching to another agent were not more likely to develop ADAs
Colman et al., 2021 [46]8912.2–17.758.7Retrospective cohort, single-center2014–2018USACD, UC, IBDuIFX, ADL With and without immunomodulator (IM)6, 12 monthsClinical and biochemical remission, discontinuation, ADAsSignificantly more patients in combination therapy with TNFi and IM were in remission after one yr than without IM (53.9% vs. 26.8%)
Without IM ADAs were unlikely to reverse if titer > 329 ng/ml
Sassine et al., 2022 [47]6391456Retrospective cohort study2009–2019 lCDTNFi Clinical relapseUse of TNFi reduced risk for relapse compared to immunomodulators
Scarallo et al., 2021b [48]1701265.6, 46.7Retrospective
Two centers
2008–2018ItalyCD, UCIFX, ADL1–1.5 Endoscopic (mucosal and histological) remissionMH was achieved by 32 patients with CD (41%) and 30 patients with UC (53.6%); 26 patients with CD (33.3%) and 22 patients with UC (39.3%) achieved HH
Withdrawal of TNFi associated with relapse
Weigl et al., 2023 [49]13 52Retrospective GermanyCDTNFi No perioperative TNFi (N = 16) Weight, height, disease activity, infectionsImprovement of weight, height after ileocecal resection, significantly more improvement in disease activity in TNFi group, no increase in infections
Infliximab
deBruyn et al., 2018 [11]18014.354.4Retrospective, multicenter2008–2012CanadaCDIFX1.5 yrs Discontinuation, dose optimizationDose escalation occurred in 57.3% primarily due to loss of response
Annual discontinuation 3.2% per yr
Kierkus et al., 2012 [17]6614.143.9Prospective cohort PolandCDIFX5.6 yrs 2, 6, 10 weeksDisease activity (clinical, laboratory & endoscopic)33% reached clinical remission, 28% non-responders, endoscopic improvement in week 10
Luo et al., 2017 [19]1311.746.2Prospective ChinaCDIFX12 monthsEEN (n = 13)8 weeksPCDAI, growth, AEsSignificantly higher percentage of clinical response, growth, and AEs in IFX group
Hyams et al., 2012 [22]6014.553.3Randomized2006–2010USA, CanadaUCIFX1.4 yrsDosing interval 8 vs. 12 weeks8, 54 weeksClinical remission, AEsResponse at week 8 73.3%, overall remission rate at week 54 was 28.6%, no serious AEs
Bolia et al., 2019 [23]2041250Retrospective2005–2016AustraliaUCIFX Colectomy ratesReduction in colectomy rates after introduction of IFX
Jongsma et al., 2020 [50]50 Multicenter open label randomized controlled trial CDIFX Conventional treatment (N = 50) Steroids/EEN10, 52 weeksClinical and endoscopic remissionHigher percentage of patients in TNFi group achieved clinical (59%) and endoscopic remission (59%) at week 10, no significant difference in week 52, less treatment escalation needed in TNFi group at week 52
Jongsma et al., 2020b [51]20159.2257Retrospective, case–control, multicenter2015–2019Europe, CanadaCD, UC, IBDuIFX Start IFX < 10 yrs of age vs. >10 yrs1 yrDosing, treatment intervals, trough levels, discontinuation, clinical remissionEqual amount of patients maintained therapy with IFX, younger patients on significantly higher dosage per kg, no effect of proactive drug monitoring
Church et al., 2019 [52]1251454–70Retrospective
Single-center
2000–2015CanadaSR UCIFX0.7 yrsStandard vs. intensified inductionM = 1.4 yrsColectomy, remission, mucosal healing, AEsLower chance of colectomy in intensified regimen, other long-term outcomes are similar, 66% mucosal healing, AEs were rare
van Hoeve et al., 2019 [53]35 retrospective2012–2018 CD, UCIFX Remission at week 52 vs. non remission52 weeksClinical, biological remission, trough levelsTrough levels just before maintenance were the only predictors for clinical and biological remission
Schnell et al., 2021 [54]4213.3, 14.2764.3Prospective, controlled, single-center GermanyCD, UCIFX Healthy matched controls2, 6, 12 monthsBiological remission, trough levels, cytokinesHigher trough levels in patients responding to treatment after 6 months, no effect of comedication with azathioprine
Before treatment different cytokine profiles in IBD patients and healthy controls
Cheifetz et al., 2022 [55]103 Post hoc REACH trial CDIFX 10, 30, 54 weeksClinical remissionHigher infliximab concentration at week 10 was associated with clinical remission at week 10, and 30
Lawrence et al., 2022 [56]14014,154%trial2016–2018Canada, Scotland IFX Standard induction vs. Optimization-based induction52 weeksClinical remissionHigher rates of clinical remission in optimized induction
Chung et al., 2022 [57]85 Single-center retrospective CDIFX Pharmacokinetic model of infliximab clearance, clinical remissionCRP and Albumin predict trough levels, induction trough levels predict remission
Kwon et al., 2022 [58]3013.780Prospective2020–2021KoreaCDIFX Cytokines, trough levels, clinical and biochemical remissionHigher cytokine profiles in patients not achieving remission than in patients in remission, Cut-off for higher IFX doses TNFi concentration > 27.6 pg/ml
Constant et al., 2021 [59]5513.169Retrospective single-center2013–2019USACDIFX 2, 8 weeksLaboratory markers, IFX trough levelsBaseline laboratory markers (CRP, hypoalbuminemia, ESR) significantly associated with inadequate post-induction IFX trough concentration
Merras-Salmio et al., 2017 [60]14614.857Retrospective,
Single-center
2003–2014NorwayCD, UC, IBDuIFX1.8 IFX trough levels, IFX ADAs63% of patients had loss of response, trough level significantly higher in patients in remission or ongoing therapy
Dave et al., 2021 [61]3014.3–33.560Part prospective, part retrospective2017–2019IndiaCD, UCIFX5 IFX trough level, ADAs, evaluation of iDose softwareiDose predicted 70% of patients’ trough concentrations correctly
Of 11 patients managed with iDose, 8 achieved clinical remission, 2 showed partial response, one developed antibody
Curci et al., 2021 [62]7614.747.4Prospective,
two centers
ItalyCD, UCIFX 8, 22, 52 weeksClinical responsesingle-nucleotide polymorphisms (SNPs) rs396991 in FCGR3A variant had significantly lower trough levels, higher chance of ADAs and reduced clinical response
Clarkston et al., 2019 [63]7213.665Prospective cohort,
Single-center
2014–2018USCDIFX51 days 1 yrClinical response (wPCDAI), biological response, maintenance concentrationsClinical response 64%, fecal calprotectin improvement in 54%
El-Matary et al., 2019 [64]5213.560.8Cohort,
multicenter
2014–2017CanadafCDIFX 24 weeksFistula healing, trough levelsCorrelation between pre-fourth infusion trough levels and fistula healing
Stein et al., 2016 [65]7714.863Prospective
single-center
2006–2011USCDIFX1.66 yrs 1 yrOngoing treatment with IFX
CRP, ADAs, trough levels
78% remained on IFX associated higher week 10 trough levels
Drobne et al., 2018 [66]18315.4–4057Cohort,
single-center
2010–2015SloveniaCD, UC, IBDuIFX7.3, 5.7 Trough level, CRP, fecal calprotectinHigher trough levels were associated with lower levels of CRP and fecal calprotectin, no higher number of infections in higher trough levels
Courbette et al., 2020 [67]11111.659Retrospective
single-center
2002–2014FranceCDIFX 14 weeksClinical response, predictors for response, through levels38.7% in clinical remission plus 36% partial response
Normal growth and normal albumin levels at first application associated with clinical response
Crombé et al., 2011 [68]12014.545Retrospective registry1988–2004FranceCDIFX41 months Short- and long-term efficacy, rate of resection surgery, AEs58% response rate, reduced risk for surgery in responder group, 13% of AEs that led to discontinuation
Adalimumab
Cozijnsen et al., 2015 [5]531149.1Observational cohort NetherlandsCDADL3 yrs MD = 12 monthsCategorized cPCDAI, discontinuation/treatment failure64% remission after three months, maintained by 50% for two yrs, more IFX primary non-responders failed ADL than Patients with loss of response
Croft et al., 2021 [24]93 Double blind
nulticenter
2014–201810 countriesUCADL High dose induction vs. standard dose8, 52 weeksClinical remission, mucosal healing, AEsRemission rates in ADL group better than in placebo groups, high dose induction had higher rate of remission in week 8 and week 52
Assa et al., 2019 [69]7814.371Randomized controlled trial2015–2018IsraelCDADL Proactive vs. reactive drug monitoring8–72 weeksSteroid-free remission, biologic remission, discontinuationSignificantly higher proportion of patients achieved steroid-free remission in the proactive group than in the reactive group (82% vs. 48%), as well as drug intensification (87% vs. 60%)
Matar et al., 2020 [70]7814.371Randomized controlled trial
multicenter
2015–2018IsraelCDADL With and without immunomodulator (IM) Sustained steroid-free remission, laboratory markers, trough levels, ADAs, AEsNo difference in steroid-free remission between groups with and without IM (73% vs. 63%), or laboratory markers, trough levels, ADAs, occurrence of AEs
Dubinsky et al., 2016 [71]188 51, 57Randomized controlled trial
multicenter
8 countriesCDADL3 yrsHigh dose, low dose weekly4, 26, 52 weeksRemission, response rate, AEsSignificantly higher proportion of patients in high dose group responded (31.4% vs. 18.8%) and achieved remission (57.1% vs. 47.9%), same rate of AEs
Payen et al., 2023 [72]120 2008–2019 CDADL Top-down vs. step-up12, 24 monthsSteroid -, EEN-free remission, clinical remissionTop-down strategy more effective, higher serum levels of ADL, no serious AEs
Lucafò et al., 2021 [73]3214.962.5Retrospective cross sectional
multicenter
2013–2019ItalyCD, UCADL41.73 4, 52, 82 weeksDisease activity (PUCAI, PCDAI), trough levelsAround 50% remission rate, higher trough levels in patients with sustained clinical remission
Golimumab
Hyams et al., 2017 [74]3515 Open-label
multicenter
2014–2015North America, Europe, IsraelUCGOL15 yrs 2, 4, 6 weeksSerum concentration, clinical outcomes, AEs60% clinical response, 34% clinical remission, and 54% mucosal healing, no safety concerns
Vedolizumab
Garcia Romero et al., 2021 [7]4212.652.4Retrospective, multicenter2017–2019SpainCD UCVEDO2.6 yrs (CD),
4.1 yrs (UC)
14, 30, 52 weeksLaboratory markers, activity indices, AEs52.4% overall remission rate at week 14, more in UC, 84.5% remained remission in week 52
Atia et al., 2023 [75]14213.646%Multicenter, prospective cohort, multicenter2016–20226 countriesCD, UC, IBDuVEDO 14 weeksSteroid-free -/EEN-free remission42% UC in remission under vedolizumab, 32% CD, optimal drug concentration at week 14—> 7µg/ml
Ungaro et al., 2019 [76]22 pediatricadult + pediatric Cross-sectional,
two centers
USACD, UCVEDO Clinical -, steroid-free -, biochemical remission, drug concentrationVedolizumab concentration > 11.5 µg/mL was associated with steroid free and biochemical remission
Colman et al., 2023 [77]741651Prospective observational2014–2019USACD, UC, IBDuVEDO33 months Pharmacokinetic model, clinical remission, through levelsFinal model includes weight, erythrocyte sedimentation rate, and hypoalbuminemia
Ustekinumab
Yerushalmy-Feler et al., 2022 [8]6915.8 Retrospective,
Multicenter
EuropeCDUST4.3 yrs 3 monthsClinical remission, CRP, fecal Calprotectin, endoscopic, histological healingReduction in inflammatory markers, 16% endoscopic, 13% histological mucosal healing
Dhaliwal et al., 2021 [9]2514.828Prospective,
multicenter
2018–2019CanadaUCUST2.3 yrs 26, 52 weeksSteroid-free remission, PUCAI, endoscopic remission, AEs69% steroid free remission, significantly more of whom only failed TNFi treatment before (instead of TNFi and VEDO also)
Dayan et al., 2019 [10]5216.850, 20Observational cohort2014–2018USACD, UC, IBDuUST4.9 yrs (CD) and 1.8 yrs (UC/IBDu) 52 weeksSteroid-free remission, clinical and biomarker remission75% maintained on UST after one yr, 50% of bio-exposed and 90% of bio-naïve in steroid free remission
Note: Only studies including pediatric patients receiving TNFi are included in the table. ADAs = anti-drug antibodies, ADL = adalimumab, AEs = adverse events, CD = Crohn’s disease, EEN = exclusive enteral nutrition, fCD = fistulizing Crohn’s disease, GOL = golimumab, IBDu = unclassified IBD, IFX = infliximab, IM = immunomodulator JIA = juvenile idiopathic arthritis, lCD = luminal Crohn’s disease, MINI = Mucosal Inflammation Non-invasive Index, (w)PCDAI = (weighted) pediatric Crohn’s disease activity index, PEN = partial enteral nutrition, PsA = psoriasis arthritis, PUCAI = pediatric ulcerative colitis activity index, SR UC = steroid refractory, TNFi = TNF-inhibitors, UC = ulcerative colitis, UST = ustekinumab, VEDO = vedolizumab, VEO IBD = very early onset inflammatory bowel disease, yrs = years.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Claßen, M.; Hoerning, A. Current Role of Monoclonal Antibody Therapy in Pediatric IBD: A Special Focus on Therapeutic Drug Monitoring and Treat-to-Target Strategies. Children 2023, 10, 634. https://doi.org/10.3390/children10040634

AMA Style

Claßen M, Hoerning A. Current Role of Monoclonal Antibody Therapy in Pediatric IBD: A Special Focus on Therapeutic Drug Monitoring and Treat-to-Target Strategies. Children. 2023; 10(4):634. https://doi.org/10.3390/children10040634

Chicago/Turabian Style

Claßen, Merle, and André Hoerning. 2023. "Current Role of Monoclonal Antibody Therapy in Pediatric IBD: A Special Focus on Therapeutic Drug Monitoring and Treat-to-Target Strategies" Children 10, no. 4: 634. https://doi.org/10.3390/children10040634

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop