Next Article in Journal
Is Intermittent Abdominal Pressure Ventilation Still Relevant? A Multicenter Retrospective Pilot Study
Next Article in Special Issue
The Inflammatory Bowel Disease—Disk Tool for Assessing Disability in Inflammatory Bowel Disease Patients: Validation of the Greek Version
Previous Article in Journal
Efficacy and Safety of Roxadustat in Chinese Hemodialysis Patients: A Systematic Review and Meta-Analysis
Previous Article in Special Issue
Contemporary Management of Postoperative Crohn’s Disease after Ileocolonic Resection
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recent Advances in the Optimization of Anti-TNF Treatment in Patients with Inflammatory Bowel Disease

by
Eleni Orfanoudaki
*,
Kalliopi Foteinogiannopoulou
,
Eirini Theodoraki
and
Ioannis E. Koutroubakis
Department of Gastroenterology, University Hospital of Heraklion, Medical School, University of Crete, 71003 Heraklion, Greece
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(7), 2452; https://doi.org/10.3390/jcm12072452
Submission received: 11 February 2023 / Revised: 11 March 2023 / Accepted: 16 March 2023 / Published: 23 March 2023
(This article belongs to the Special Issue New Proactive Approaches and Precision Medicine in Crohn's Disease)

Abstract

:
Despite the evolution in inflammatory bowel disease (IBD) management during the last 20 years owing to the advent of new advanced therapies, anti-TNF agents still remain the cornerstone of therapy for both Crohn’s disease and ulcerative colitis. However, this does not only secure favorable outcomes for patients considering the progressive disease character and the high likelihood of primary or secondary loss of response. Therefore, trying to reach a better treatment approach and maximize the benefits anti-TNF agents offer, optimization strategies should be examined. It has been indicated that optimizing treatment with anti-TNF enhances drug efficacy and has been associated with improved disease outcomes and a complication-free disease course. From this perspective, we aim to provide an overview of currently available data and recent advances in the practices of anti-TNF treatment optimization. Special focus has been given to the role of therapeutic drug monitoring (TDM), as well as the utility of combining anti-TNF with an immunomodulator and the treat-to-target approach.

1. Introduction

The introduction of anti-TNFs (tumor necrosis factors) in the therapeutic algorithm of inflammatory bowel diseases (IBD) in the late 1990s has revolutionized patient management [1]. Since then, several other promising advanced therapies, including other biologics and small molecules targeting different inflammatory pathways, have gained approval for IBD treatment and many more emerging therapies are still under investigation [2]. Despite the advances in treatment, recent network meta-analysis, in the absence of head-to-head comparative studies, still suggest that anti-TNFs are a cornerstone of therapy for both moderate-to-severe Crohn’s disease (CD) and ulcerative colitis (UC) [3,4].
As treatment goals evolve, initiating anti-TNF IBD specialists aim not only for a clinical response, steroid free remission and endoscopic healing, but also for more strict endpoints, including deep remission (histologic and transmural healing) and, somehow, modification of the natural course of the disease [5]. For some patients these are realistic and feasible targets to achieve, but as with all advanced therapies, anti-TNFs reach a far from perfect plateau as regards their efficacy (60–70%), meaning that there is still a proportion of patients who either initially lack (refractory to treatment) or in the long-term lose response [6,7]. This variability in responses is attributed to differences in pharmacokinetics and pharmacodynamics, and is defined as primary non-response and secondary loss of response, respectively. Additionally, there is also a considerable percentage of patients who will develop adverse reactions or an intolerance necessitating treatment modification or discontinuation.
Given all the above, it is considered that anti-TNFs and their optimal use are still powerful weapons in the treatment of IBD. It is of crucial importance that the best choice is made for each patient, especially when there is high risk of worse outcomes and complications. It matters even more in the first-line choice for a biologic-naïve patient, since experience has shown that in subsequent agents upon a first failure the effectiveness rates decrease [8,9].
Apart from acting early in the disease course and aggressively with a top-down approach, once anti-TNF therapy is commenced, optimization of the therapy should be strongly considered. The importance of such a practice is becoming increasingly acknowledged, as it is a way to enhance drug efficacy, improve treatment persistence, reduce side effects, manage costs and, therefore, improve outcomes.
In this review, we provide current data on strategies to optimize anti-TNF therapy with a focus on therapeutic drug monitoring (TDM), along with the role of combination therapy of anti-TNF with immunomodulators and the tight control approach.

2. Therapeutic Drug Monitoring (TDM)

2.1. Anti-TNF Trough Concentrations and Antibodies

Measuring trough concentrations (TCs) and targeting a concentration within a therapeutic range has been implemented in several medications other than anti-TNFs. It is a way to avoid under- and overtreatment that could lead to impaired efficacy and safety issues, respectively. As for anti-TNFs, there are plenty of studies on the value of TC measurement, primarily for infliximab (IFX), to a lesser extent for adalimumab (ADA), and scarcely for golimumab and certolizumab pegol. This may be attributed to the former’s broader use, availability, indications, effectiveness and structure (chimeric antibody), rendering it more susceptible to immunogenicity issues.
The whole practice is based on the theory of the dose–response relationship [10]. Increased, within predefined therapeutic ranges, drug concentrations are associated with positive disease outcomes. Notably, there is a positive correlation between higher TCs and the clinical and/or endoscopic response and remission, mucοsal healing, lower relapse rates, fewer complications, fewer hospitalizations and fewer disease-related surgeries [11,12,13,14,15,16]. Moreover, optimal TCs are associated with a remarkable improvement in the patients’ quality of life, which is one of the long-term targets set out in the recent STRIDE (selecting therapeutic targets in inflammatory bowel disease) consensus [17].
Correspondingly, suboptimal TCs correlate with unfavorable results. In a large prospective observational study [PANTS (personalized anti-TNF therapy in Crohn’s disease (CD))], which included 1610 patients under both IFX and ADA treatment, low week (w) 14 TCs were independently associated with primary treatment failure (w14), and were highly predictive of w54 non-remission and immunogenicity [18]. This association of serum TCs with therapeutic outcomes has been confirmed in both induction and maintenance periods. As a matter of fact, achieving optimal induction or post-induction concentrations might be of most importance, due to the high inflammatory burden that has to be regulated and these concentrations may prejudge the agents’ survival thereafter. However, not all patients will show the same response to an administered anti-TNF, even with IFX whose dosage is determined by the patient’s weight. There are certain factors affecting the pharmacokinetics (PK) of anti-TNFs, meaning the drugs’ absorption, metabolism, distribution into the tissues and, lastly, its elimination from the body [10]. The key determinants of anti-TNF PK, associated with increased drug clearance, lower drug concentrations and, therefore, worse outcomes, are displayed in Table 1. Sometimes, despite even achieving an adequate drug concentration, the response may still not be satisfactory indicating a possible non-TNF driven inflammatory pathway.
Immunogenicity, characterized by the formation of anti-drug antibodies (Abs), constitutes one of the main causes of loss of response (LOR) due to an anti-TNF agent interfering with the PKs [10]. Of note, patients with positive Abs were found to run a 3-fold higher risk of LOR (relative risk = 3.2; 95% CI, 1.9–5.5; p < 0.0001) [21], the majority of which are formed as early as the first 12 months of anti-TNF initiation (90%) [22].
However, not all types of Abs impact on drug clearance. Apart from total neutralizing Abs that bind to the drug rendering it ineffective, there are also reports about the low titer of transient Abs, which lack neutralizing potential and are, therefore, clinically insignificant [23,24]. As for the technical aspects, there is a variability in the assays used for Abs detection and quantification, but a standard methodology has not been widely adopted yet. As a consequence, there is problem when interpreting Abs titles from different assays. Comparative studies have shown that drug tolerant assays designed to detect Abs, despite the presence of a circulating drug, are more accurate and should be preferred over drug sensitive ones [24,25]. Additionally, they enable earlier identification of potential positive cases, offering time for the clinician to act and optimize the therapy preventing an unlikely event of LOR [26]. In detail, the detection of transient or low-concentration Abs can be managed by treatment optimization (dose escalation, dose interval shortening and/or the addition of an immunomodulator), whereas usually the presence of higher titer Abs can lead, inevitably, to undetectable or low drug concentrations, infusion reactions and treatment failure [16].

2.2. Therapeutic Drug Monitoring

Based on the significant evidence in favor of the value of TCs of anti-TNFs in IBD management, the therapeutic drug monitoring (TDM) approach has gained attention during the last decade in clinical practice. According to this practice, TCs and Abs are measured and treatment is adjusted with the aim of reaching the TCs within a therapeutic range [16]. Evidently, it is a way to maintain an adequate dose and, therefore, enhance drug efficacy, prolong response durability and avoid complications. It is a valuable tool for treatment optimization, used widely for anti-TNF agents.
Acknowledging that TCs are positively correlated with clinical outcomes, it is necessary to determine the appropriate cut-off levels to apply in TDM algorithms. However, the optimal TC threshold concentrations to achieve the maximal efficacy of anti-TNFs cannot be clearly predefined. Recent data extracted from retrospective observational reports or post hoc analyses were found to be heterogeneous for the patient sample, outcome and assay used.
From the interpretation of all the available studies in this field, it has become clear that target TCs may differ depending on the administered anti-TNF agent, the measurement time point, the IBD phenotype and severity and, lastly, but foremost, the desired outcome of interest. Regarding the time point, TCs differ as expected between the induction, post-induction or maintenance periods, with the former requiring higher concentrations justified by the higher inflammatory burden at initiation and, consequently, the higher anti-TNF fecal loses and serum needs [27]. Likewise, in the same sense, higher TCs are required for more aggressive disease phenotypes (for example penetrating or perianal CD, acute severe ulcerative colitis) and when more stringent endpoints are being considered (endoscopic or histologic remission) [27].
Table 2 shows a summary of the most relevant and representative studies regarding TCs in the maintenance phase and their association with different outcomes. A maintenance phase target TC of ≥3–7 μg/mL for IFX and ≥5–8 μg/mL for ADA have been suggested in order to achieve favorable outcomes [28].
Undeniably, determining the best thresholds for anti-TNF agents is not an “one size fits all” situation. When awaiting prospective data, a more personalized approach should be followed since certain populations may benefit from higher concentrations and in these cases optimal management should be offered.
To date there are two different approaches to the application of TDM, the reactive and proactive approach, respectively. Reactive TDM is performed in the setting of loss of response in a patient at induction or maintenance treatment with an anti-TNF agent, basically aiming to guide the next steps in the management, whereas proactive TDM is used prophylactically in asymptomatic patients at certain intervals and situations, so as to prevent a disease flare [27]. There is evidence that both proactive and reactive TDM, when added to routine investigations, altered the management decisions in almost half of the adult CD patients receiving anti-TNF therapy [29]. The implementation of TDM is supported by many IBD experts and societies. In official guidelines it stands more as a suggestion or a low-quality evidence recommendation owing to the conflicting results in the current literature and the absence of prospective studies and RCTs (randomized controlled trials) [30,31,32,33].
Table 2. Summary of most relevant and representative studies regarding TCs in the maintenance phase and their association with different outcomes.
Table 2. Summary of most relevant and representative studies regarding TCs in the maintenance phase and their association with different outcomes.
Author, Year [Ref.]Disease Type TCs (mg/mL)OutcomeAssay
IFX
Roblin, 2017 [34]CD>2.1Clinical remissionELISA
Vande Casteele, 2015 [35]CD>2.8Normal CRP (≤5 mg/L)HMSA
Ward, 2017 [36]CD>3.4Normal CRP (≤5 mg/L)ELISA
Ward, 2017 [36]CD>5.7Normal FC (<59 μg/g)ELISA
Papamichael, 2018 [11]CD≥9.7Endoscopic remissionELISA/HMSA
Papamichael, 2018 [11]CD≥9.8Histologic remissionELISA/HMSA
Yarur, 2017 [37]CD>10.1Mucosal healingHMSA
Yarur, 2017 [37]CD>10.1Fistula healingHMSA
Adedokun, 2014 [38]UC>2.4Clinical response w54ELISA
Margo, 2017 [39]UC>3Normal FC (<250 μg/g)ELISA
Papamichael, 2017 [12]UC≥7.5Endoscopic healing HMSA/ELISA
Papamichael, 2017 [12]UC≥10.5Histologic healingHMSA/ELISA
ADA
Nakase, 2017 [40]CD>5Clinical remissionELISA
Mazor, 2014 [41]CD>5.9Normal CRP (≤5 mg/L)ELISA
Morita, 2016 [42]CD>7.9Mucosal healingELISA
Juncadella, 2018 [43]CD≥12Endoscopic remissionHMSA
Juncadella, 2018 [43]CD≥12.2Histologic remissionHMSA
Paul, 2014 [44]CD/UC>4.8 Clinical remissionELISA/Radioimmunoassay
Ungar, 2016 [45]CD/UC>6.6Normal CRP (≤5 mg/L)ELISA
Yarur, 2016 [46]UC/CD>7.5Endoscopic healingHMSA
Yarur, 2016 [46]UC/CD>7.8Histologic healingHMSA
Morita, 2016 [47]UC>10.3Mucosal healingELISA
ELISA: enzyme-linked immunosorbent assay; HMSA: homogenous mobility shift assay; CD: Crohn’s disease; UC: ulcerative colitis; IFX: infliximab; ADA: adalimumab; FC: fecal calprotectin; CRP: C-reactive protein; TCs: trough concentrations.

2.2.1. Reactive TDM vs. Empiric Use of Anti-TNF

The utility of reactive TDM has been investigated in several studies and is more established in contrast to that of proactive TDM. In the setting of LOR, assessing TCs and Abs helps to identify the possible cause and manage it in a more efficient and documented way. It can be precisely identified whether failure is attributed to pharmacokinetic (immunogenic or not) or mechanistic reasons, which makes it more likely that successful management decisions are taken (optimize the current anti-TNF or change the medication). On the other hand, an empiric approach alone has been proved to be suboptimal, costly and may put the patient in jeopardy.
Reactive TDM-based treatment was found to be significantly superior in terms of clinical response, endoscopic remission, cost savings and hospitalization rates, than empiric treatment of IBD patients under IFX [48]. Regarding ADA, Restellini et al. reported that reactive TDM-guided dosing in CD patients with LOR had better outcomes than empiric dose escalation [49].
On the contrary, the GETAID (Groupe d’Etude Therapeutique des Affections Inflammatoires du Tube Digestif) team, following symptom-based treatment optimization in IBD patients under maintenance with IFX, showed similar rates of clinical, endoscopic and steroid-free remission at week 54, as the biomarker and TC-driven-based therapy [50]. Steenholdt et al., in their study, also verified the lack of superiority in clinical outcomes between conventional and reactive management, despite the significant advantage in costs in favor of the latter [51].
A systematic review and a meta-analysis of the literature concluded that reactive TDM is not better in terms of clinical efficacy versus the standard of care, however it offers a cost advantage. The authors highlighted that the data are insufficient and mostly of low quality to evidently support, for or against, this approach [15,52].
In practice, reactive TDM, if available, should be considered along with clinical evaluation, biomarkers and endoscopy to optimize treatment in the advent of LOR, gaining in costs, time and agents. In case of immunogenicity with high Ab concentrations, by following the suggested reactive TDM algorithm, time is not wasted waiting for improvement after the empirically applied dose escalation. Moreover, an agent is not easily abandoned before being completely sure that the optimal concentrations have been reached, getting the best of it.

2.2.2. Proactive TDM versus Empiric Use of Anti-TNF

Proactive TDM is defined as a periodically performed measurement of TCs and ADAs in patients in clinical remission, followed by an appropriate adjustment in the drug dosing or timing intervals with the aim of reaching certain concentrations associated with better outcomes. Apart from efficacy reasons, it has been suggested as a method to promote safety and cost effectiveness. However, the benefits of such a practice when compared to standard practice have not been convincingly proven. Maybe, the implementation of proactive TDM during or shortly after induction is theoretically of more principal importance. High inflammatory burden, usually present during induction, may cause low drug concentrations. Low week 14 TCs, both for IFX and ADA, were independently associated with primary non-response and a lack of remission at week 54 [18]. Moreover, low TCs are predisposed to antibody formation and further drug clearance, rendering it ineffective (secondary LOR); a condition not easily reversed. Retrospective analysis of the ACCENT 1 (a Crohn’s disease clinical trial evaluating infliximab in a new long-term treatment regimen) trial has also demonstrated durable sustained response through to week 54 in CD patients under IFX, achieving both satisfactory TCs at week 14 (≥3.5 µg/m) and a ≥60% decrease in CRP (C-reactive protein) [53]. Furthermore, in perianal CD IFX TCs > 9.2 μg/mL at week 2 of IFX induction were associated with fistula closure at week 14 and 30 [54]. Similar short- and long-term beneficial outcomes have been observed in studies concerning both disease types and all anti-TNF agents with variable TC limits, according to the week tested and targeted outcome (Table 3).
An example, underscoring the value of proactive TDM during induction is that of the recognized need for accelerated IFX dosing during induction in severe acute ulcerative colitis. It was performed empirically at first, based on the understanding of underlying pharmacokinetic mechanisms influencing drug clearance [71], but eventually accelerated IFX therapy was deemed to be associated with reduced colectomy rates and less LOR episodes. Boosting effective serum concentrations during induction has proved to be beneficial in other at-risk patients, apart from those with high disease activity, like obese patients or smokers. One can guess how advantageous it could be if the accelerated dosing was performed more precisely using proactive TDM, quantifying exactly the needs caused by this inevitable medicine loss. To sum up, the TDM practice instituted early after treatment initiation could help avoid the 30% of described cases or primary non-response, and actually limit its causes to just mechanistic failure and predict better outcomes in the long term [72].
On the contrary, in the recently published Norwegian NOR-DRUM (NORwegian DRUg Monitoring) study, an RCT designed to assess the value of proactive TDM during the induction of IFX in patients with various chronic immune mediated inflammatory diseases, including IBD, found no benefit in the use of TDM when compared with standard care for clinical remission rates over week 30 [73].
The maintenance phase data on proactive TDM remain conflicting. TAXIT (trough level adapted infliximab treatment) and TAILORIX (a study investigating tailored treatment with infliximab in active Crohn’s disease), two out of the limited existing RCTs in this field, failed to demonstrate a significant benefit of proactive TDM in achieving remission at year one. However, fewer flares were noted in the TDM group implying a more efficient use of the anti-TNF [50,74]. On the other side, the maintenance sub-study of the aforementioned NOR-DRUM study did find significant superiority in the proactive vs. the standard approach in sustaining disease control without worsening, during the 52 week study period [75]. This was confirmed by the pediatric PAILOT (pediatric Crohn’s disease adalimumab level-based optimization treatment) study regarding ADA in CD and 1-year steroid-free clinical remission, as well as more composite outcomes (steroid-free clinical remission + normal CRP + normal FC (fecal calprotectin)) [76], and became further validated when applied for a longer time period, far from one year. Indicatively, in a 3-year Spanish prospective trial during a median follow-up at 84 [34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118] weeks, longer treatment durability, less disease-related surgeries or hospital admissions and less serious infusion or other adverse reactions, were noted in the proactive TDM group [77]. Moreover, even stronger target outcomes, like mucosal healing, were accomplished in higher rates than with standard management [78].
Similarly, conflicting results on proactive TDM during maintenance are displayed in retrospective observational studies. TDM-driven anti-TNF treatment (IFX or ADA) demonstrated favorable outcomes in terms of efficacy, health care utilization, surgeries, drug survival and durable drug effect not only in the short term, but also in the long term, regarding both disease types [79,80,81,82,83]. Even if applied at least once during the maintenance in patients receiving ADA, it was found to be associated with less 3-year treatment failure [83]. On the other hand, Bernardo et al. failed to show any difference between proactive TDM and the empiric arm for clinical remission, relapse and surgery [84], although time to relapse was found to be notably longer in the former. In the 3-year follow-up of the TAXIT trial patients that were assigned to either arm during the study (TDM or clinical driven dosing), the same rates of mucosal healing and treatment persistence were found [74]. However, both arms were initially optimized using the proactive TDM algorithm before randomization, which might have influenced the results.
Two meta-analyses by Ricciuoto et al. and Nguyen et al. did not identify a significant difference in the clinical remission rates using proactive TDM versus the normal standard of care, although its implementation might provide potential advantages to the length of anti-TNF survival/persistence [15,85]. However, the authors conclude that a possible positive effect in patients with certain disease characteristics (high risk of worse outcomes) or at a certain phase (ex. induction) cannot be ruled out (possible underrepresentation at RCTs). On the contrary, Sethi et al., including both RCTs and observational studies in their meta-analysis, did find a significant association of proactive TDM with reduced treatment failure and surgical rates, as well as superiority regarding endoscopic remission. Considering all the available data and before they become further validated and substantiated, the individualized use of proactive TDM in certain cases could be recommended.

2.2.3. Other Applications of Proactive TDM

The utility of proactive TDM is not restricted to exclusively guiding the treatment aimed at preventing LOR, but it has also displayed value in other clinical scenarios.
One of these scenarios has to do with treatment de-escalation of anti-TNFs for safety and cost issues when the disease is well controlled. This could be conducted either by decreasing the dose or by lengthening the time interval. TDM implementation in such a case helps to better determine the patients that can be safely and successfully de-escalated, rather than using clinical and/or biochemical parameters alone. Amiot et al., based on its association with relapse-free survival, suggested that TDM should be used prior to de-escalation in preference to symptoms and CRP [87]. Additionally, using IFX-TCs > 7 mg/L as a trough cut-off to perform de-escalation, patients were found to run a decreased relapse risk (HR: 0.45, p = 0.024) [88], whereas for ADA the cut-off value for successful dose reduction was 12.2 μg/mL, as described in an observational prospective study by Peris et al. [89]. Even after deintensification, continuing TDM can be beneficial for the follow-up [90]. Maintaining IFX–TCs > 2.4 μg/mL, thereafter, is critical according to Pettitcolin et al. since lower concentrations turned out to be predictive of relapse (p = 0.0001) [90].
Similarly, TDM could be considered to guide immunosuppressants (IMM) cessation in patients receiving combination treatments. Drobne et al. demonstrated that withdrawing IMM after at least 6 months of combination treatment did not significantly affect TCs [91]. Detectable > 0.3 μg/mL TCs at IMM cessation were associated with long-term response. Interestingly, in the same study, none of the patients with IFX–TCs > 5 μg/mL at the time of IMM withdrawal lost response during the 29-month (IQR, 15–45) follow-up period.
In cases where combination treatment cannot be used (because of a history of severe infections, malignancies etc.), there is evidence that optimized monotherapy using proactive TDM could be used. A post hoc analysis from the SONIC (study of biologic and immunomodulator naïve patients in Crohn’s disease) trial showed that after stratifying patients into quartiles based on IFX–TCs, outcomes at w26 within a quartile were comparable irrespective of IMM use [92]. Likewise, no difference in either the efficacy or drug persistence was noted when proactively optimized IFX monotherapy and combination therapy were compared [93,94]. However, the former was associated with increased drug consumption [93].
Another scenario fitting for proactive TDM, is when considering resuming an anti-TNF agent after a drug holiday. Applied early after re-exposure (before second dose), proactive TDM may help in predicting the outcomes. At that time, positive Abs predispose to severe infusion reactions and, apparently, there is a need for treatment discontinuation, whereas high TCs are related to the long-term response [95].
Lastly, and not well-established, is the role of proactive TDM peri-operatively. In the era of advanced therapies most patients at the time of surgery have been exposed or are currently being exposed to biologic treatment. Lau et al. reported a positive relationship between TCs and adverse post-operative outcomes in CD patients [96]. Unlike, in the PUCCINI (prospective cohort of ulcerative colitis and Crohn’s disease patients undergoing surgery to identify risk factors for post-operative infection) trial, the biggest prospective observational study in this field, which included 947 patients undergoing surgery (382 patients were exposed to anti-TNF within 12 weeks of surgery), no association was found between detectable TCs and post-operative infections (surgical site or not) [97]. The outcomes were comparable in smaller cohort studies, demonstrating no difference in post-operative complications irrespective of anti-TNF exposure, the titer of TCs, or the time interval from surgery to the last dose [98,99]. The current practice is not to delay surgery for reasons like recent anti-TNF exposure.

2.3. Limitations and Challenges Implementing TDM

Based on the current evidence, TDM could not be recommended for wide use in clinical practice. Although the additional guidance it offers in the management of IBD is quite established, there are several aspects that have not been fully clarified.
There are still knowledge gaps regarding the optimal application of TDM, result interpretation and determination of the optimal thresholds to target. This can be attributed partially to the lack of standardization, the heterogeneity in populations and outcomes across most studies, and the high variability of testing kits and assays, especially for antibody detection, causing confusion in result interpretation.
There is also an evident difficulty in obtaining the results in a timely fashion, so as to drive early therapy decisions. The commonly used ELISA (enzyme-linked immunosorbent assay), has a turnaround time of 2–4 weeks causing a delay in the implementation of changes. Moreover, most experience refers to IFX rather than other anti-TNF agents, mostly the maintenance phase rather than the induction and, last but not least, the cost issue owing to the lack of reimbursement from insurance companies.
Another issue that has yet to be addressed, is that of the best timing within a treatment circle for the serum concentrations to be measured. Conventionally, serum anti-TNF concentrations are measured at the trough, meaning at their expected lowest point just before the scheduled re-administration. However, better understanding of the underlying pharmacokinetics has led to the assessment of concentrations other than at the trough, like at the peak or intermediate point. There is emerging evidence on the utility and value of these concentrations in predicting remission, particularly if implemented at the induction [100]. This is usually the case for IFX, since for ADA concentrations measured any time in between a single treatment circle seem to be comparable (POETIC study (prospective observational evaluation of time-dependency of adalimumab immunogenicity and drug concentration)) [65]. In this context, tissue drug concentrations are also being considered and their concentrations in noninflamed tissue are associated with higher sustained response and remission [101].

2.4. Current Perspectives

In order to overcome the time lag between TCs measurement and result availability, point of care (POC) methods have been developed for the evaluation of TCs and Abs. They are rapid assays carried out at the point where care is provided, making accurate results feasible within minutes. Regarding TCs measurement, POC assays exhibit high correlation with standard ELISA, with an overall agreement of up to 89.4% [102,103]. Moreover, POC tests can detect Abs accurately and reliably enough, with 100% specificity and 76% sensitivity, and their results are highly correlated with those from the ELISA assays too [104]. Despite the proven non-inferiority of POC tests, a revision of the currently suggested TC thresholds should be considered, as in POC tests TC values seem to be higher [105]. In clinical practice ultra-proactive TDM applied using POC tests was no different in rates of IFX failure or sustained clinical remission after 1-year when compared with a once-applied reactive TDM [106].
Another emerging attempt toward a more personalized approach, is the use of dashboard-driven TDM. By entering patients’ data, including pharmacokinetic parameters like CRP and albumin that influence drug clearance, dashboards offer guidance on optimal dosing and treatment intervals by indirectly forecasting TCs. The PRECISION trial demonstrated lower LOR rates and FC values 1-year after application of dashboard-driven dosing in patients with IBD in remission under maintenance treatment with IFX in comparison with the normal standard of care [107]. If applied earlier to guide accelerated IFX induction dosing, a PK dashboard may improve drug durability and immunogenicity [108]. In the meantime, an RCT is ongoing (the OPTIMIZE trial) also focusing on the induction phase, with the aim of assessing remission rates and the need for rescue therapy from week 14 through to week 52 in patients with moderate-to-severe CD initiating IFX and comparing the proactive TDM dashboard-driven dosing with the normal standard of care [109].
The combination of these innovations, point-of-care testing and dashboard-driven TDM, introduced early at anti-TNF initiation could potentially further maximize the outcomes (drug survival, effectiveness, costs, safety).

3. Combination Therapy

Several studies suggest that combination therapy of an anti-TNF with an immunomodulator (thiopurine or methotrexate (MTX)) has a superior effect over anti-TNF monotherapy in IBD [110,111] (Table 4). The main mechanism explaining combination therapy superiority is the effect of the combination therapy on the immunogenicity of the anti-TNF. The risk of Abs development in patients under combination therapy seems to be lower than in those under monotherapy. The presence of Abs leads to lower serum TCs, less probabilities of therapy success and higher rates of loss of response. IFX use is more likely to develop Abs because of its structure, and this may explain the fact that most trials supporting combination therapy refer to IFX [112]. ADA antibody formation rates are lower than for IFX, however, there is still a benefit from using it in combination with an immunomodulator over monotherapy [113]. Another possible mechanism explaining the benefits of combination therapy, regarding thiopurines, is their separate ability to manage disease activity or even, mucosal healing [114,115].

3.1. IFX Combination Therapy

There is strong evidence, coming from large clinical trials, supporting the use of combination therapy of IFX with an immunomodulator in IBD. The SONIC trial showed that combination therapy of IFX with AZA (azathioprine) displays better outcomes compared with monotherapy [116]. Correspondingly, regarding UC, the SUCCESS study highlighted the superiority of the combination of IFX with AZA against IFX monotherapy in naïve UC patients [117].
The ECCO (European Crohn’s and Colitis Organization) recommends the combination of IFX with a thiopurine for the induction of remission in patients with moderate-to-severe Crohn’s disease after a failure in response to conventional therapy [30]. The AGA (American Gastroenterological Association), as well, suggests the use of IFX combined with a thiopurine for the induction and maintenance therapy of naïve to both biologics and thiopurine patients, and comments that the combination of IFX with methotrexate may as well be preferred over monotherapy [32].
Notably, concerning combination therapy with MTX, there is no clear evidence to support its use to improve IFX efficacy, since the data remain rather conflicting.

3.2. Adalimumab Combination Therapy

In contrast to what applies for IFX, there are differences in the guidelines regarding the necessity of concomitant immunosuppression regarding treatment with ADA. The ECCO does not recommend the combination to achieve clinical response and remission [30], whereas the AGA guidelines favor the use of combination therapy with thiopurines for the induction and maintenance of remission over ADA monotherapy in naïve CD patients. Furthermore, according to the AGA recommendations, the combination therapy of ADA with MTX may be also considered [32].
The Diamond study, evaluating the utility of ADA in combination or not with a thiopurine showed no superiority of the former over monotherapy in the achievement of clinical remission in week 52. However, combination therapy was associated with higher endoscopic remission rates in week 26 [118].
Targownik et al., in a retrospective analysis of more than 70,000 patients with CD and UC, compared the use of IFX or ADA in combination with an immunosuppressant, either thiopurine or methotrexate.
Any combination therapy was found to be associated with a significant decrease in treatment failure for both disease types. ADA and IFX were equally effective (in combination with an immunomodulator) in CD, but regarding UC, thiopurines performed better over methotrexate in combination with an anti-TNF [122].

3.3. Anti-TNF Experienced Patients

An important issue is that most of the available data come from clinical trials with patients naïve to biologics. This seems to be only a part of the real-world management. In fact, a respectable proportion of patients previously exposed to anti-TNF need to change therapy to another anti-TNF and this turns out to be rather challenging. However, in a recent study the use of combination therapy versus monotherapy in patients with previous immunogenicity related failure, changing to another anti-TNF was evaluated. Patients previously exposed to IFX changed to either ADA or ADA combined with AZA, and patients previously exposed to ADA changed to either IFX or IFX combined with AZA, respectively. Combination therapy was the only factor associated with better outcomes at 24 months [120].

3.4. Combination Treatment with Immunomodulator: Dosage, Timing and Safety

Although the value of combination treatment has been clearly demonstrated, there are certain other issues that have not been fully clarified yet.
The recommended therapeutic thiopurine dose in IMM monotherapy is 2–2.5 mg/kg for azathioprine and 1–1.5 mg/kg for 6-MP (6-mercaptopurine). It seems that when applied as combination therapy, not as high doses are needed. In a recent study by Ramos et al., remission, response or failure rates using anti-TNF (IFX or ADA) in combination treatment with either a standard (2–2.5 mg/kg) or decreased (<2 mg/kg) azathioprine dose, were similar in the two groups. Anti-TNF (IFX or ADA) serum concentrations and Abs formation, as well as endoscopic or biochemical remission, in the two groups were similar [124,125]. Even if thiopurine was started in full dose and patients were in durable remission under combination treatment, dose reduction shows equal efficacy performance as its maintenance at full dose [126].
Considering the duration of the combination treatment, data are rather conflicting. Lambrescak et al. in a retrospective cohort study, which included a group of 139 patients with CD or UC, showed that a shorter duration of IFX combination therapy with an immunomodulator (thiopurine or MTX) was not associated with a higher risk of treatment failure. Moreover, in patients where the immunosuppressant was discontinued, a combination therapy of 6–11 months was not associated with a higher risk of treatment failure compared with those with a duration of combination therapy >12 months [127].
On the other hand, in patients with CD in clinical remission, a duration of combination therapy with IFX and AZA of less than 27 months was associated with higher rates of relapse over the withdrawal of AZA, after a period of 27 months [128]. Additionally, Mahmoud et al., in a retrospective cohort study, showed that the immunomodulator withdrawal was not associated with a higher risk of loss of response, but the presence of Abs was more frequent in the serum of these patients over those with uninterrupted use of combination therapy. Notably, a longer period of immunomodulator use before withdrawal and higher TCs at the time of withdrawal were associated with a lower risk of Abs formation. Furthermore, higher TCs and clinical remission at the time of withdrawal were also associated with lower risk of loss of response [123]. Apparently, there is no optimal duration for combination treatment and an individualized approach should be considered.
As for the timing of the immunomodulator administration, the simultaneous initiation of both anti-TNF and IMM is preferable. Chen et al. in a retrospective study, which included more than 9000 patients with UC and CD, showed that biologic therapy (mainly anti-TNF) in combination with an immunosuppressant is related to a lower risk of biologic discontinuation and of note, when the immunomodulator therapy was started more than 30 days before the biologic induction, the risk of discontinuation of the biologic was even lower [129]. Patients starting IMM at the time of IFX induction also displayed lower incidence rates of Abs formation, contrary to those starting concomitant IMM later [113]. At this point it is also worth mentioning that an IMM may as well be added later in the treatment with anti-TNF in case of LOR, since it plays a role not only in preventing, but also in suppressing or even eliminating antibody formation, as well as in recapturing a response [130,131].
Lastly, the use of a combined therapy raises concerns about safety. There is evidence that combination therapy is not associated with more adverse outcomes, but in some specific patient groups there is, nevertheless, still a need for more caution; this is the case because it has been associated with a higher risk of lymphoma and serious infections [132,133]. More specifically, young men may be at a higher risk of hepatosplenic T-cell lymphoma and the elderly may be at a higher risk of lymphoma and infections [134,135].
In conclusion, the combination of anti-TNF agents with an immunomodulator is one of the most efficacious treatments available. Early initiation of combination therapy is recommended as part of the top-down approach for high-risk patients (with risk factors for worse outcomes and complications, aggressive disease behavior) with moderate-to-severe disease, although the evidence regarding adalimumab is not strong enough yet. Combination therapy has also proved beneficial in anti-TNF experienced patients in the case of immunogenicity-related failure. For low-risk patients or for patients with concerns regarding safety, monotherapy is suggested, preserving the IMM addition when LOR or immunogenicity issues arise. Moreover, therapy personalization with the right benefit–risk assessment and the awareness and participation of the patient in the decision making, may lead to the best approach to optimize IBD therapy.

4. Treat-to-Target Approach in IBD

In IBD, like other chronic diseases, it is of high importance to set clear therapeutic targets, in order proper disease management to be adopted and major IBD complications, hospitalizations, surgeries, and poor quality of life to be avoided. According to STRIDE I, concerning IBD treatment, the alleviation of clinical symptoms or else the so-called clinical response/remission and endoscopic remission as well, were identified as the therapeutic goals, short-term and long-term targets, respectively [136]. As for the resolution of clinical symptoms, it became obvious over the years that this primary target was not enough to ensure neither the resolution of bowel inflammation nor the prevention of permanent intestinal damage leading to surgery [137]. In the aforementioned SONIC trial, 50% of CD patients who were in clinical remission, as expressed by the CDAI (Crohn’s Disease Activity Index) score, had endoscopic and/or biochemical (CRP) active disease [138]. Thus, STRIDE II updated the treat-to-target strategy, incorporating serum and fecal biomarkers as the intermediate targets in between the short- (symptomatic response) and long-term targets (endoscopic healing) [17].

4.1. Biomarkers in IBD

The most commonly used biomarkers in everyday clinical practice for IBD patients are serum C-reactive protein (CRP) and fecal calprotectin (FC). According to a meta-analysis, the pooled sensitivity and specificity of CRP and FC were 49% (95% CI 34–64%) and 92% (95% CI 72–96%), and 73% (95% CI 66–79%) and 82% (95% CI 73–88%), respectively. The FC was more sensitive than the CRP in both disease types and was more sensitive in ulcerative colitis than Crohn’s disease [139]. In another more recent meta-analysis, the FC showed a pooled sensitivity of 85%, specificity of 75%, DOR (diagnostic odds ratios) of 16.3 and AUC (area under the curve) of 0.88, for assessing endoscopically active disease [140]. Regarding the FC cut-offs, the best sensitivity (90.6%) was found at 50 μg/g and the best specificity (78.2%) at >100 μg/g. For CD patients, a FC cut-off of 250 mg/g is highly predictive of mucosal healing, whereas a stricter cut-off of 100 mg/g is suggestive of deep healing (mucosal and transmural) [17]. Likewise, in UC different FC cut-offs reflect different degrees of mucosal healing as well, 250 mg/g, 150 mg/g and 100 mg/g endoscopic improvement, mucosal healing and histologic healing, respectively [17]. It is of note that consecutive FC measurements, rather than a single one, are useful for predicting short-term clinical relapses (>261 μg/g with AUC = 0.901, sensitivity 87.2%, specificity 85.3%, p < 0.001) [141].
As mentioned before, while FC may show better sensitivity, CRP has higher specificity in identifying active intestinal disease. Low CRP values have been associated with a lower risk of clinical relapse, with AUC of 0.72 and an optimal cut-off of 1.0 mg/L, with a positive and negative predictive value of 21% and 94%, respectively [142]. Moreover, a high CRP value at the time of anti-TNF discontinuation is associated with a higher relapse risk [143], whereas normalization of CRP after treatment initiation is predictive of maintained response or remission through to w54 in CD patients under IFX [144]. In the ACCENT trial, a CRP decrease > 60% at week 14 was significantly associated with sustained response to IFX [53]. It seems that a baseline CRP and CRP reduction rate could be useful in order to predict the primary non-response or the secondary loss of response to anti-TNF in patients with CD, and could possibly act as a guide for choosing the right therapeutic strategy [145].
The CALM (efficacy and safety of two treatment algorithms in adults with moderate-to-severe Crohn’s disease) study for CD was the first trial to show that patients in the tight-control group (escalation of anti-TNF treatment based on clinical scores, biomarkers or prednisone use in the previous week) achieved higher rates of mucosal healing [CDEIS (Crohn’s Disease Index of Severity) < 4] at week 48 in comparison to the clinical management group (escalation of anti-TNF treatment based only on clinical scores or prednisone use in the previous week) (46% of 122 patients) over (30% of 122 patients), respectively, with an adjusted risk difference of 16·1% (95% CI 3·9–28·3; p = 0·010)) [146]. A subsequent analysis, which included 122 patients from the CALM study with early moderate-to-severe CD, showed that deep remission (CDEIS < 4, absence of/with no deep ulcers or steroids, for eight or more weeks) was significantly associated (aHR (adjusted hazard ration), 0.19; 95% 0.07–0.31) with a decreased risk of severe disease complications, independently of the tight control or conventional management strategy [147].
As far as UC is concerned, in refractory disease treated with IFX, independent predictors of colectomy-free survival were found to be mucosal healing (OR, 4.02; 95% CI, 1.16–13.97; p = 0.028), baseline CRP of 5 mg/L or less (OR, 2.95; 95% CI, 1.26–6.89; p = 0.012) and baseline albumin of 35 g/L or greater (OR, 3.03; 95% CI, 1.12–8.22; p = 0.029) [148]. It is well-established that in UC, FC has a better sensitivity compared to CRP in predicting the endoscopic activity of the disease [136].
Furthermore, FC has been established as a useful predictor of clinical relapses [149]. There is evidence showing that a combination of biomarkers and other factors, such as IFX trough concentrations and Abs to IFX at week 22, are associated with secondary loss of response to anti-TNF [150]. Similarly, a combined endpoint (CDAI < 150 and CRP ≤ 2.9 mg/L and FC improvement) in CD patients after anti-TNF induction at week 12 was found to predict corticosteroid-free remission at week 52 [151]. These results suggest that possibly a combination of clinical scores, IFX concentrations as well as serum and fecal biomarkers could be used to guide and optimize IBD treatment.
On the other hand, opposed to the value of the tight-control approach, highlighted above, there are some recent conflicting data from the STARDUST (treat-to-target versus the standard of care for Crohn’s disease patients treated with ustekinumab) trial, which suggests that implementing the standard of care strategy in patients with CD treated with ustekinumab is not inferior in terms of the endoscopic response at week 48 versus the treat-to-target one [152].

4.2. Future Perspectives for IBD Treatment Targets

As it is already known, inflammation spreads continuously and in a superficial manner in UC, while on the contrary, in a patchy and more transmural manner in CD. For this reason, histological remission in UC and transmural healing in CD could possibly act, in the future, as the next long-term targets after endoscopic remission and mucosal healing. According to a large meta-analysis, which included 28 studies and 2806 patients (2677 UC; 129 CD), there was evidence to suggest that patients with UC who were in endoscopic remission, faced a higher risk of relapse in case of persistent histologic activity (OR 2.41, 95% CI 1.91–3.04). On the contrary, no association was found between histologic activity and relapse in CD [153]. In the same way, according to another meta-analysis, UC patients with histologic remission faced a 63% lower risk of clinical relapse vs. those with persistent histologic activity (RR, 0.37; 95% CI, 0.24–0.56), whereas the annual risk of clinical relapse in those achieving histologic remission was only 5.0% (95% CI, 3.3–7.7) [154].
When it comes to CD, it was shown that transmural healing (defined as a bowel wall thickness of ≤3 mm at bowel sonography) was associated with a higher rate of steroid-free clinical remission (95.6%), lower rates of hospitalization (8.8%) and surgeries (0%) at 1-year versus mucosal (75%, 28.3% and 10%, respectively) and no healing (41%, 66.6% and 35.5%, respectively) (p < 0.001) in patients treated with anti-TNF for 2 years [155]. In another observational study, which included 214 CD patients that underwent MRE (magnetic resonance enterography) and colonoscopy every 6 months, endoscopic remission (OR 0.331 95% CI, 0.178–0.614, p < 0.001) and MRE remission (OR 0.270 95% CI, 0.130–0.564, p < 0.001) were independently associated with a lower risk of adverse outcomes (surgeries, hospitalization, bowel damage) [156]. What is more, early transmural healing as it was expressed by MRE at week 12 after anti-TNF treatment (defined as a 25% decrease in either the Clermont score (odds ratio (OR) = 7.7 (1.7–34.0), p < 0.001) or the Magnetic Resonance Index of Activity (OR = 4.2 (1.3–13.3), p = 0.015)), was shown to be associated with sustained clinical remission (corticosteroid-free remission at week 52) and prevention of long-term bowel damage in CD (HR 0.21 (0.0–0.9); p = 0.037) [157].
Nevertheless, currently neither the transmural healing in CD nor the histological remission in UC constitutes formal targets in the treatment algorithm of IBD. These targets have not yet been incorporated into the treat-to-target IBD strategy and further studies and RCTs are needed in order for the benefits of such a therapeutic algorithm to be proven.

5. Concluding Remarks

IBD management has evolved over the past two decades owing to the emergence of several biologics. Anti-TNFs were the first biologics to be approved for this indication and, notably, remain among the most efficacious agents. What is more, IFX and ADA were ranked highest as first-line treatments for the induction of clinical remission in both CD and UC patients, offering a distinct advantage over other therapies [3,158]. However, this does not always preclude an unfavorable outcome for patients, given the progressive disease character and the high likelihood of primary or secondary loss of response. Hence, the need for treatment optimization, even from the outset, has emerged in an effort to make the best use of them, preserve their effectiveness and maximize their durability.
TDM can facilitate decision making through the determination of the treatment failure mechanism. Especially when performed in a reactive setting, but also proactively in certain scenarios, TDM has proved to be beneficial in terms of recapturing a loss of response and prolonging anti-TNF durability and its advantageous outcomes. Similarly, the value of combination treatment of anti-TNF with an immunomodulator has been sufficiently established for IFX and in high-risk patients, or those with previous experience of another anti-TNF. Lastly, the achievement of a tight-disease control, by applying a treat-to-target strategy helps to accomplish improved and efficient management. Setting and reaching short-term targets, like the normalization of CRP or FC, that satisfactorily reflect the current activity enables a more complication-free disease course.
Despite the advances in all the three fields mentioned, there are still limitations and challenges that have to be overcome. Additionally, all these should be implemented in practice with a critical appraisal and an individualized approach. So far, due to the lack of strong evidence recommendations, there is substantial variability in optimization practices. Further, well-designed prospective studies and RCTs are needed to elucidate the role of TDM, combination treatment and tight control to enhance anti-TNF treatment.

Author Contributions

E.O., K.F. and E.T. performed drafting. I.E.K. review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

E.O. has received support from Vianex; I.K. has received support from Abbvie, Genesis, Ferring, Janssen, MSD, Pfizer, Shire, Vianex and Takeda.

References

  1. D’Haens, G.R.; van Deventer, S. 25 years of anti-TNF treatment for inflammatory bowel disease: Lessons from the past and a look to the future. Gut 2021, 70, 1396–1405. [Google Scholar] [PubMed]
  2. Al-Bawardy, B.; Shivashankar, R.; Proctor, D.D. Novel and Emerging Therapies for Inflammatory Bowel Disease. Front. Pharmacol. 2021, 12, 651415. [Google Scholar]
  3. Singh, S.; Murad, M.H.; Fumery, M.; Sedano, R.; Jairath, V.; Panaccione, R.; Sandborn, W.J.; Ma, C. Comparative efficacy and safety of biologic therapies for moderate-to-severe Crohn’s disease: A systematic review and network meta-analysis. Lancet Gastroenterol. Hepatol. 2021, 6, 1002–1014. [Google Scholar] [PubMed]
  4. Lasa, J.S.; Olivera, P.A.; Danese, S.; Peyrin-Biroulet, L. Efficacy and safety of biologics and small molecule drugs for patients with moderate-to-severe ulcerative colitis: A systematic review and network meta-analysis. Lancet Gastroenterol. Hepatol. 2022, 7, 161–170. [Google Scholar]
  5. Colombel, J.F.; D’Haens, G.; Lee, W.J.; Petersson, J.; Panaccione, R. Outcomes and Strategies to Support a Treat-to-target Approach in Inflammatory Bowel Disease: A Systematic Review. J. Crohn’s Colitis 2020, 14, 254–266. [Google Scholar]
  6. Gisbert, J.P.; Panés, J. Loss of response and requirement of infliximab dose intensification in Crohn’s disease: A review. Am. J. Gastroenterol. 2009, 104, 760–767. [Google Scholar] [PubMed]
  7. Fine, S.; Papamichael, K.; Cheifetz, A.S. Etiology and Management of Lack or Loss of Response to Anti-Tumor Necrosis Factor Therapy in Patients With Inflammatory Bowel Disease. Gastroenterol. Hepatol. 2019, 15, 656–665. [Google Scholar]
  8. Gisbert, J.P.; Marín, A.C.; McNicholl, A.G.; Chaparro, M. Systematic review with meta-analysis: The efficacy of a second anti-TNF in patients with inflammatory bowel disease whose previous anti-TNF treatment has failed. Aliment. Pharm. Ther. 2015, 41, 613–623. [Google Scholar]
  9. Singh, S.; George, J.; Boland, B.S.; Casteele, N.V.; Sandborn, W.J. Primary Non-Response to Tumor Necrosis Factor Antagonists is Associated with Inferior Response to Second-line Biologics in Patients with Inflammatory Bowel Diseases: A Systematic Review and Meta-analysis. J. Crohn’s Colitis 2018, 12, 635–643. [Google Scholar] [CrossRef]
  10. Steenholdt, C.; Bendtzen, K.; Brynskov, J.; Ainsworth, M.A. Optimizing Treatment with TNF Inhibitors in Inflammatory Bowel Disease by Monitoring Drug Levels and Antidrug Antibodies. Inflamm. Bowel Dis. 2016, 22, 1999–2015. [Google Scholar]
  11. Papamichael, K.; Rakowsky, S.; Rivera, C.; Cheifetz, A.S.; Osterman, M.T. Association between Serum Infliximab trough Concentrations during Maintenance Therapy and Biochemical, Endoscopic, and Histologic Remission in Crohn’s Disease. Inflamm. Bowel Dis. 2018, 24, 2266–2271. [Google Scholar]
  12. Papamichael, K.; Rakowsky, S.; Rivera, C.; Cheifetz, A.S.; Osterman, M.T. Infliximab trough concentrations during maintenance therapy are associated with endoscopic and histologic healing in ulcerative colitis. Aliment. Pharm. Ther. 2018, 47, 478–484. [Google Scholar]
  13. Silva-Ferreira, F.; Afonso, J.; Pinto-Lopes, P.; Magro, F. A Systematic Review on Infliximab and Adalimumab Drug Monitoring: Levels, Clinical Outcomes and Assays. Inflamm. Bowel Dis. 2016, 22, 2289–2301. [Google Scholar] [CrossRef] [PubMed]
  14. Moore, C.; Corbett, G.; Moss, A.C. Systematic Review and Meta-Analysis: Serum Infliximab Levels During Maintenance Therapy and Outcomes in Inflammatory Bowel Disease. J. Crohn’s Colitis 2016, 10, 619–625. [Google Scholar]
  15. Ricciuto, A.; Dhaliwal, J.; Walters, T.D.; Griffiths, A.M.; Church, P.C. Clinical Outcomes With Therapeutic Drug Monitoring in Inflammatory Bowel Disease: A Systematic Review With Meta-Analysis. J. Crohn’s Colitis 2018, 12, 1302–1315. [Google Scholar]
  16. Martins, C.A.; Garcia, K.S.; Queiroz, N.S.F. Multi-utility of therapeutic drug monitoring in inflammatory bowel diseases. Front. Med. 2022, 9, 864888. [Google Scholar]
  17. 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]
  18. 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]
  19. Papamichael, K.; Afif, W.; Drobne, D.; Dubinsky, M.C.; Ferrante, M.; Irving, P.M.; Kamperidis, N.; Kobayashi, T.; Kotze, P.G.; Lambert, J.; et al. Therapeutic drug monitoring of biologics in inflammatory bowel disease: Unmet needs and future perspectives. Lancet Gastroenterol. Hepatol. 2022, 7, 171–185. [Google Scholar]
  20. Lefevre, P.L.C.; Shackelton, L.M.; Casteele, N.V. Factors Influencing Drug Disposition of Monoclonal Antibodies in Inflammatory Bowel Disease: Implications for Personalized Medicine. BioDrugs 2019, 33, 453–468. [Google Scholar]
  21. Nanda, K.S.; Cheifetz, A.S.; Moss, A.C. Impact of antibodies to infliximab on clinical outcomes and serum infliximab levels in patients with inflammatory bowel disease (IBD): A meta-analysis. Am. J. Gastroenterol. 2013, 108, 40–47; quiz 8. [Google Scholar] [CrossRef] [Green Version]
  22. Ungar, B.; Chowers, Y.; Yavzori, M.; Picard, O.; Fudim, E.; Har-Noy, O.; Kopylov, U.; Eliakim, R.; Ben-Horin, S. The temporal evolution of antidrug antibodies in patients with inflammatory bowel disease treated with infliximab. Gut 2014, 63, 1258–1264. [Google Scholar] [CrossRef]
  23. Vande Casteele, N.; Gils, A.; Singh, S.; Ohrmund, L.; Hauenstein, S.; Rutgeerts, P.; Vermeire, S. Antibody response to infliximab and its impact on pharmacokinetics can be transient. Am. J. Gastroenterol. 2013, 108, 962–971. [Google Scholar] [CrossRef] [PubMed]
  24. Lodhia, N.; Rao, S. Updates in therapeutic drug monitoring in inflammatory bowel disease. World J. Gastroenterol. 2022, 28, 2282–2290. [Google Scholar] [CrossRef]
  25. Vande Casteele, N. Assays for measurement of TNF antagonists in practice. Frontline Gastroenterol. 2017, 8, 236–242. [Google Scholar] [CrossRef] [Green Version]
  26. Kharlamova, N.; Hermanrud, C.; Dunn, N.; Ryner, M.; Hambardzumyan, K.; Vivar Pomiano, N.; Marits, P.; Gjertsson, I.; Saevarsdottir, S.; Pullerits, R.; et al. Drug Tolerant Anti-drug Antibody Assay for Infliximab Treatment in Clinical Practice Identifies Positive Cases Earlier. Front. Immunol. 2020, 11, 1365. [Google Scholar] [CrossRef] [PubMed]
  27. Papamichael, K.; Cheifetz, A.S. Therapeutic drug monitoring in inflammatory bowel disease: For every patient and every drug? Curr. Opin. Gastroenterol. 2019, 35, 302–310. [Google Scholar] [CrossRef] [PubMed]
  28. Vermeire, S.; Dreesen, E.; Papamichael, K.; Dubinsky, M.C. How, When, and for Whom Should We Perform Therapeutic Drug Monitoring? Clin. Gastroenterol. Hepatol. 2020, 18, 1291–1299. [Google Scholar] [CrossRef]
  29. Wu, Y.; Wen, A.; Selvanderan, S.P.; Xuan, W.; Andrews, J.M.; Koo, J.H.; Williams, A.-J.; Ng, W.; Connor, S. Management Decisions in Crohn’s Disease Are Changed by Knowledge of Proactive and Reactive Testing of Antitumor Necrosis Factor Drug Levels. Crohn’s Colitis 360 2021, 3, otab042. [Google Scholar] [CrossRef]
  30. Torres, J.; Bonovas, S.; Doherty, G.; Kucharzik, T.; Gisbert, J.P.; Raine, T.; Adamina, M.; Armuzzi, A.; Bachmann, O.; Bager, P.; et al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Medical Treatment. J. Crohn’s Colitis 2019, 14, 4–22. [Google Scholar] [CrossRef] [Green Version]
  31. Lamb, C.A.; Kennedy, N.A.; Raine, T.; Hendy, P.A.; Smith, P.J.; Limdi, J.K.; Hayee, B.; Lomer, M.C.E.; Parkes, G.C.; Selinger, C.; et al. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut 2019, 68 (Suppl 3), s1–s106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Feuerstein, J.D.; Nguyen, G.C.; Kupfer, S.S.; Falck-Ytter, Y.; Singh, S. American Gastroenterological Association Institute Guideline on Therapeutic Drug Monitoring in Inflammatory Bowel Disease. Gastroenterology 2017, 153, 827–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Cheifetz, A.S.; Abreu, M.T.; Afif, W.; Cross, R.K.; Dubinsky, M.C.; Loftus, E.V., Jr.; 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] [PubMed]
  34. Roblin, X.; Boschetti, G.; Duru, G.; Williet, N.; Deltedesco, E.; Phelip, J.M.; Peyrin-Biroulet, L.; Nancey, S.; Flourié, B.; Paul, S. Distinct Thresholds of Infliximab Trough Level Are Associated with Different Therapeutic Outcomes in Patients with Inflammatory Bowel Disease: A Prospective Observational Study. Inflamm. Bowel Dis. 2017, 23, 2048–2053. [Google Scholar] [CrossRef]
  35. Vande Casteele, N.; Khanna, R.; Levesque, B.G.; Stitt, L.; Zou, G.Y.; Singh, S.; Lockton, S.; Hauenstein, S.; Ohrmund, L.; Greenberg, G.R.; et al. The relationship between infliximab concentrations, antibodies to infliximab and disease activity in Crohn’s disease. Gut 2015, 64, 1539–1545. [Google Scholar] [CrossRef]
  36. Ward, M.G.; Warner, B.; Unsworth, N.; Chuah, S.W.; Brownclarke, C.; Shieh, S.; Parkes, M.; Sanderson, J.D.; Arkir, Z.; Reynolds, J.; et al. Infliximab and adalimumab drug levels in Crohn’s disease: Contrasting associations with disease activity and influencing factors. Aliment. Pharm. Ther. 2017, 46, 150–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. 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. Pharm. Ther. 2017, 45, 933–940. [Google Scholar] [CrossRef] [Green Version]
  38. Adedokun, O.J.; Sandborn, W.J.; Feagan, B.G.; Rutgeerts, P.; Xu, Z.; Marano, C.W.; Johanns, J.; Zhou, H.; Davis, H.M.; Cornillie, F.; et al. Association between serum concentration of infliximab and efficacy in adult patients with ulcerative colitis. Gastroenterology 2014, 147, 1296–1307.e5. [Google Scholar] [CrossRef] [Green Version]
  39. Magro, F.; Afonso, J.; Lopes, S.; Coelho, R.; Gonçalves, R.; Caldeira, P.; Lago, P.; de Sousa, H.T.; Ramos, J.; Gonçalves, A.R.; et al. Clinical performance of an infliximab rapid quantification assay. Ther. Adv. Gastroenterol. 2017, 10, 651–660. [Google Scholar] [CrossRef] [Green Version]
  40. Nakase, H.; Motoya, S.; Matsumoto, T.; Watanabe, K.; Hisamatsu, T.; Yoshimura, N.; Ishida, T.; Kato, S.; Nakagawa, T.; Esaki, M.; et al. Significance of measurement of serum trough level and anti-drug antibody of adalimumab as personalised pharmacokinetics in patients with Crohn’s disease: A subanalysis of the DIAMOND trial. Aliment. Pharm. Ther. 2017, 46, 873–882. [Google Scholar] [CrossRef]
  41. Mazor, Y.; Almog, R.; Kopylov, U.; Ben Hur, D.; Blatt, A.; Dahan, A.; Waterman, M.; Ben-Horin, S.; Chowers, Y. Adalimumab drug and antibody levels as predictors of clinical and laboratory response in patients with Crohn’s disease. Aliment. Pharm. Ther. 2014, 40, 620–628. [Google Scholar] [CrossRef]
  42. Morita, Y.; Imaeda, H.; Nishida, A.; Inatomi, O.; Bamba, S.; Sasaki, M.; Tsujikawa, T.; Sugimoto, M.; Andoh, A. Association between serum adalimumab concentrations and endoscopic disease activity in patients with Crohn’s disease. J. Gastroenterol. Hepatol. 2016, 31, 1831–1836. [Google Scholar] [CrossRef] [PubMed]
  43. Juncadella, A.; Papamichael, K.; Vaughn, B.P.; Cheifetz, A.S. Maintenance Adalimumab Concentrations Are Associated with Biochemical, Endoscopic, and Histologic Remission in Inflammatory Bowel Disease. Dig. Dis. Sci. 2018, 63, 3067–3073. [Google Scholar] [CrossRef] [PubMed]
  44. Paul, S.; Moreau, A.C.; Del Tedesco, E.; Rinaudo, M.; Phelip, J.M.; Genin, C.; Peyrin-Biroulet, L.; Roblin, X. Pharmacokinetics of adalimumab in inflammatory bowel diseases: A systematic review and meta-analysis. Inflamm. Bowel Dis. 2014, 20, 1288–1295. [Google Scholar] [CrossRef] [PubMed]
  45. Ungar, B.; Levy, I.; Yavne, Y.; Yavzori, M.; Picard, O.; Fudim, E.; Loebstein, R.; Chowers, Y.; Eliakim, R.; Kopylov, U.; et al. Optimizing Anti-TNF-α Therapy: Serum Levels of Infliximab and Adalimumab Are Associated With Mucosal Healing in Patients With Inflammatory Bowel Diseases. Clin. Gastroenterol. Hepatol. 2016, 14, 550–557.e2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Yarur, A.J.; Jain, A.; Hauenstein, S.I.; Quintero, M.A.; Barkin, J.S.; Deshpande, A.R.; Sussman, D.A.; Singh, S.; Abreu, M.T. Higher Adalimumab Levels Are Associated with Histologic and Endoscopic Remission in Patients with Crohn’s Disease and Ulcerative Colitis. Inflamm. Bowel Dis. 2016, 22, 409–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Morita, Y.; Bamba, S.; Takahashi, K.; Imaeda, H.; Nishida, A.; Inatomi, O.; Sasaki, M.; Tsujikawa, T.; Sugimoto, M.; Andoh, A. Prediction of clinical and endoscopic responses to anti-tumor necrosis factor-α antibodies in ulcerative colitis. Scand. J. Gastroenterol. 2016, 51, 934–941. [Google Scholar] [CrossRef] [PubMed]
  48. Kelly, O.B.; Donnell, S.O.; Stempak, J.M.; Steinhart, A.H.; Silverberg, M.S. Therapeutic Drug Monitoring to Guide Infliximab Dose Adjustment is Associated with Better Endoscopic Outcomes than Clinical Decision Making Alone in Active Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2017, 23, 1202–1209. [Google Scholar] [CrossRef] [Green Version]
  49. Restellini, S.; Chao, C.Y.; Lakatos, P.L.; Aruljothy, A.; Aziz, H.; Kherad, O.; Bitton, A.; Wild, G.; Afif, W.; Bessissow, T. Therapeutic Drug Monitoring Guides the Management of Crohn’s Patients with Secondary Loss of Response to Adalimumab. Inflamm. Bowel Dis. 2018, 24, 1531–1538. [Google Scholar] [CrossRef] [PubMed]
  50. D’Haens, G.; Vermeire, S.; Lambrecht, G.; Baert, F.; Bossuyt, P.; Pariente, B.; Buisson, A.; Bouhnik, Y.; Filippi, J.; Vander Woude, J.; et al. Increasing Infliximab Dose Based on Symptoms, Biomarkers, and Serum Drug Concentrations Does Not Increase Clinical, Endoscopic, and Corticosteroid-Free Remission in Patients with Active Luminal Crohn’s Disease. Gastroenterology 2018, 154, 1343–1351.e1. [Google Scholar] [CrossRef]
  51. Steenholdt, C.; Brynskov, J.; Thomsen, O.; Munck, L.K.; Fallingborg, J.; Christensen, L.A.; Pedersen, G.; Kjeldsen, J.; Jacobsen, B.A.; Oxholm, A.S.; et al. Individualised therapy is more cost-effective than dose intensification in patients with Crohn’s disease who lose response to anti-TNF treatment: A randomised, controlled trial. Gut 2014, 63, 919–927. [Google Scholar] [CrossRef] [PubMed]
  52. Shah, R.; Hoffman, G.R.; El-Dallal, M.; Goldowsky, A.M.; Chen, Y.; Feuerstein, J.D. Is Therapeutic Drug Monitoring for Anti-tumour Necrosis Factor Agents in Adults with Inflammatory Bowel Disease Ready for Standard of Care? A Systematic Review and Meta-analysis. J. Crohn’s Colitis 2020, 14, 1057–1065. [Google Scholar] [CrossRef]
  53. Cornillie, F.; Hanauer, S.B.; Diamond, R.H.; Wang, J.; Tang, K.L.; Xu, Z.; Rutgeerts, P.; Vermeire, S. Postinduction serum infliximab trough level and decrease of C-reactive protein level are associated with durable sustained response to infliximab: A retrospective analysis of the ACCENT I trial. Gut 2014, 63, 1721–1727. [Google Scholar] [CrossRef] [PubMed]
  54. Davidov, Y.; Ungar, B.; Bar-Yoseph, H.; Carter, D.; Haj-Natour, O.; Yavzori, M.; Chowers, Y.; Eliakim, R.; Ben-Horin, S.; Kopylov, U. Association of Induction Infliximab Levels With Clinical Response in Perianal Crohn’s Disease. J. Crohn’s Colitis 2017, 11, 549–555. [Google Scholar] [CrossRef] [PubMed]
  55. Dreesen, E.; Baert, F.; Laharie, D.; Bossuyt, P.; Bouhnik, Y.; Buisson, A.; Lambrecht, G.; Louis, E.; Oldenburg, B.; Pariente, B.; et al. Monitoring a Combination of Calprotectin and Infliximab Identifies Patients With Mucosal Healing of Crohn’s Disease. Clin. Gastroenterol. Hepatol. 2020, 18, 637–646.e11. [Google Scholar] [CrossRef] [PubMed]
  56. Papamichael, K.; Casteele, N.V.; Jeyarajah, J.; Jairath, V.; Osterman, M.T.; Cheifetz, A.S. Higher Postinduction Infliximab Concentrations Are Associated With Improved Clinical Outcomes in Fistulizing Crohn’s Disease: An ACCENT-II Post Hoc Analysis. Am. J. Gastroenterol. 2021, 116, 1007–1014. [Google Scholar] [CrossRef]
  57. 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]
  58. Gonczi, L.; Vegh, Z.; Golovics, P.A.; Rutka, M.; Gecse, K.B.; Bor, R.; Farkas, K.; Szamosi, T.; Bene, L.; Gasztonyi, B.; et al. Prediction of Short- and Medium-term Efficacy of Biosimilar Infliximab Therapy. Do Trough Levels and Antidrug Antibody Levels or Clinical And Biochemical Markers Play the More Important Role? J. Crohn’s Colitis 2016, 11, 697–705. [Google Scholar] [CrossRef] [Green Version]
  59. Kobayashi, T.; Suzuki, Y.; Motoya, S.; Hirai, F.; Ogata, H.; Ito, H.; Sato, N.; Ozaki, K.; Watanabe, M.; Hibi, T. First trough level of infliximab at week 2 predicts future outcomes of induction therapy in ulcerative colitis-results from a multicenter prospective randomized controlled trial and its post hoc analysis. J. Gastroenterol. 2016, 51, 241–251. [Google Scholar] [CrossRef] [Green Version]
  60. Bar-Yoseph, H.; Levhar, N.; Selinger, L.; Manor, U.; Yavzori, M.; Picard, O.; Fudim, E.; Kopylov, U.; Eliakim, R.; Ben-Horin, S.; et al. Early drug and anti-infliximab antibody levels for prediction of primary nonresponse to infliximab therapy. Aliment. Pharm. Ther. 2018, 47, 212–218. [Google Scholar] [CrossRef] [Green Version]
  61. Casteele, N.V.; Jeyarajah, J.; Jairath, V.; Feagan, B.G.; Sandborn, W.J. Infliximab Exposure-Response Relationship and Thresholds Associated with Endoscopic Healing in Patients with Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 2019, 17, 1814–1821.e1. [Google Scholar] [CrossRef] [PubMed]
  62. Papamichael, K.; Van Stappen, T.; Casteele, N.V.; Gils, A.; Billiet, T.; Tops, S.; Claes, K.; Van Assche, G.; Rutgeerts, P.; Vermeire, S.; et al. Infliximab Concentration Thresholds during Induction Therapy Are Associated with Short-term Mucosal Healing in Patients With Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 2016, 14, 543–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Tighe, D.; Smith, S.; O’Connor, A.; Breslin, N.; Ryan, B.; McNamara, D. Positive relationship between infliximab and adalimumab trough levels at completion of induction therapy with clinical response rates, at a tertiary referral center. JGH Open 2017, 1, 4–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Bortlik, M.; Duricova, D.; Malickova, K.; Machkova, N.; Bouzkova, E.; Hrdlicka, L.; Komarek, A.; Lukas, M. Infliximab trough levels may predict sustained response to infliximab in patients with Crohn’s disease. J. Crohn’s Colitis 2013, 7, 736–743. [Google Scholar] [CrossRef] [Green Version]
  65. Ungar, B.; Engel, T.; Yablecovitch, D.; Lahat, A.; Lang, A.; Avidan, B.; Har-Noy, O.; Carter, D.; Levhar, N.; Selinger, L.; et al. Prospective Observational Evaluation of Time-Dependency of Adalimumab Immunogenicity and Drug Concentrations: The POETIC Study. Am. J. Gastroenterol. 2018, 113, 890–898. [Google Scholar] [CrossRef]
  66. Verstockt, B.; Moors, G.; Bian, S.; Van Stappen, T.; Van Assche, G.; Vermeire, S.; Gils, A.; Ferrante, M. Influence of early adalimumab serum levels on immunogenicity and long-term outcome of anti-TNF naive Crohn’s disease patients: The usefulness of rapid testing. Aliment. Pharm. Ther. 2018, 48, 731–739. [Google Scholar] [CrossRef] [PubMed]
  67. Casteele, N.V.; Baert, F.; Bian, S.; Dreesen, E.; Compernolle, G.; Van Assche, G.; Ferrante, M.; Vermeire, S.; Gils, A. Subcutaneous Absorption Contributes to Observed Interindividual Variability in Adalimumab Serum Concentrations in Crohn’s Disease: A Prospective Multicentre Study. J. Crohn’s Colitis 2019, 13, 1248–1256. [Google Scholar] [CrossRef]
  68. Papamichael, K.; Baert, F.; Tops, S.; Assche, G.V.; Rutgeerts, P.; Vermeire, S.; Gils, A.; Ferrante, M. Post-Induction Adalimumab Concentration is Associated with Short-Term Mucosal Healing in Patients with Ulcerative Colitis. J. Crohn’s Colitis 2017, 11, 53–59. [Google Scholar] [CrossRef] [Green Version]
  69. Baert, F.; Casteele, N.V.; Tops, S.; Noman, M.; Van Assche, G.; Rutgeerts, P.; Gils, A.; Vermeire, S.; Ferrante, M. Prior response to infliximab and early serum drug concentrations predict effects of adalimumab in ulcerative colitis. Aliment. Pharm. Ther. 2014, 40, 1324–1332. [Google Scholar] [CrossRef]
  70. Baert, F.; Kondragunta, V.; Lockton, S.; Casteele, N.V.; Hauenstein, S.; Singh, S.; Karmiris, K.; Ferrante, M.; Gils, A.; Vermeire, S. Antibodies to adalimumab are associated with future inflammation in Crohn’s patients receiving maintenance adalimumab therapy: A post hoc analysis of the Karmiris trial. Gut 2016, 65, 1126–1131. [Google Scholar] [CrossRef]
  71. Hindryckx, P.; Novak, G.; Casteele, N.V.; Laukens, D.; Parker, C.; Shackelton, L.M.; Narula, N.; Khanna, R.; Dulai, P.; Levesque, B.G.; et al. Review article: Dose optimisation of infliximab for acute severe ulcerative colitis. Aliment. Pharm. Ther. 2017, 45, 617–630. [Google Scholar] [CrossRef] [Green Version]
  72. Alsoud, D.; Verstockt, B.; Vermeire, S. Letter: Immunogenicity is not the root cause for loss of response to anti-TNF agents in patients with IBD in TDM era. Aliment. Pharm. Ther. 2022, 55, 885–886. [Google Scholar] [CrossRef]
  73. Syversen, S.W.; Goll, G.L.; Jørgensen, K.K.; Sandanger, Ø.; Sexton, J.; Olsen, I.C.; Gehin, J.E.; Warren, D.J.; Brun, M.K.; Klaasen, R.A.; et al. Effect of Therapeutic Drug Monitoring vs Standard Therapy During Infliximab Induction on Disease Remission in Patients With Chronic Immune-Mediated Inflammatory Diseases: A Randomized Clinical Trial. JAMA 2021, 325, 1744–1754. [Google Scholar] [CrossRef] [PubMed]
  74. Casteele, N.V.; Ferrante, M.; Van Assche, G.; Ballet, V.; Compernolle, G.; Van Steen, K.; Simoens, S.; Rutgeerts, P.; Gils, A.; Vermeire, S. Trough concentrations of infliximab guide dosing for patients with inflammatory bowel disease. Gastroenterology 2015, 148, 1320–1329.e3. [Google Scholar] [CrossRef] [PubMed]
  75. Syversen, S.W.; Jahnsen, J.; Haavardsholm, E.A. Therapeutic Drug Monitoring vs Standard Therapy During Maintenance Infliximab Therapy and Control of Immune-Mediated Inflammatory Diseases-Reply. JAMA 2022, 327, 1506–1507. [Google Scholar] [CrossRef]
  76. 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] [PubMed]
  77. Sánchez-Hernández, J.G.; Rebollo, N.; Martin-Suarez, A.; Calvo, M.V.; Muñoz, F. A 3-year prospective study of a multidisciplinary early proactive therapeutic drug monitoring programme of infliximab treatments in inflammatory bowel disease. Br. J. Clin. Pharmacol. 2020, 86, 1165–1175. [Google Scholar] [CrossRef]
  78. Fernandes, S.R.; Bernardo, S.; Simões, C.; Gonçalves, A.R.; Valente, A.; Baldaia, C.; Moura Santos, P.; Correia, L.A.; Tato Marinho, R. Proactive Infliximab Drug Monitoring Is Superior to Conventional Management in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2020, 26, 263–270. [Google Scholar] [CrossRef]
  79. Vaughn, B.P.; Martinez-Vazquez, M.; Patwardhan, V.R.; Moss, A.C.; Sandborn, W.J.; Cheifetz, A.S. Proactive therapeutic concentration monitoring of infliximab may improve outcomes for patients with inflammatory bowel disease: Results from a pilot observational study. Inflamm. Bowel Dis. 2014, 20, 1996–2003. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Syed, N.; Tolaymat, M.; Brown, S.A.; Sivasailam, B.; Cross, R.K. Proactive Drug Monitoring Is Associated With Higher Persistence to Infliximab and Adalimumab Treatment and Lower Healthcare Utilization Compared With Reactive and Clinical Monitoring. Crohns Colitis 360 2020, 2, otaa050. [Google Scholar] [CrossRef]
  81. Lyles, J.L.; Mulgund, A.A.; Bauman, L.E.; Su, W.; Fei, L.; Chona, D.L.; Sharma, P.; Etter, R.K.; Hellmann, J.; Denson, L.A.; et al. Effect of a Practice-wide Anti-TNF Proactive Therapeutic Drug Monitoring Program on Outcomes in Pediatric Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2021, 27, 482–492. [Google Scholar] [CrossRef]
  82. Papamichael, K.; Cheifetz, A.S. Proactive Adalimumab Monitoring in Inflammatory Bowel Disease: Current Data and Future Perspectives. J. Crohn’s Colitis 2020, 14, 878–879. [Google Scholar] [CrossRef]
  83. Papamichael, K.; Juncadella, A.; Wong, D.; Rakowsky, S.; Sattler, L.A.; Campbell, J.P.; Vaughn, B.P.; Cheifetz, A.S. Proactive Therapeutic Drug Monitoring of Adalimumab Is Associated with Better Long-term Outcomes Compared with Standard of Care in Patients with Inflammatory Bowel Disease. J. Crohn’s Colitis 2019, 13, 976–981. [Google Scholar] [CrossRef] [PubMed]
  84. Bernardo, S.; Fernandes, S.; Gonçalves, A.R.; Baldaia, C.; Valente, A.; Moura Santos, P.; Correia, L.; Velosa, J. P453 Efficacy of therapeutic drug monitoring of anti-TNF therapy in the control of patients with inflammatory bowel disease. J. Crohn’s Colitis 2017, 11, S308–S309. [Google Scholar] [CrossRef] [Green Version]
  85. Nguyen, N.H.; Solitano, V.; Vuyyuru, S.K.; MacDonald, J.K.; Syversen, S.W.; Jørgensen, K.K.; Crowley, E.; Ma, C.; Jairath, V.; Singh, S. Proactive Therapeutic Drug Monitoring Versus Conventional Management for Inflammatory Bowel Diseases: A Systematic Review and Meta-Analysis. Gastroenterology 2022, 163, 937–949.e2. [Google Scholar] [CrossRef] [PubMed]
  86. Sethi, S.; Dias, S.; Kumar, A.; Blackwell, J.; Brookes, M.J.; Segal, J.P. Meta-analysis: The efficacy of therapeutic drug monitoring of anti-TNF-therapy in inflammatory bowel disease. Aliment. Pharm. Ther. 2022. [Google Scholar] [CrossRef] [PubMed]
  87. Amiot, A.; Hulin, A.; Belhassan, M.; Andre, C.; Gagniere, C.; Le Baleur, Y.; Farcet, J.P.; Delchier, J.C.; Hüe, S. Therapeutic drug monitoring is predictive of loss of response after de-escalation of infliximab therapy in patients with inflammatory bowel disease in clinical remission. Clin. Res. Hepatol. Gastroenterol. 2016, 40, 90–98. [Google Scholar] [CrossRef]
  88. Lucidarme, C.; Petitcollin, A.; Brochard, C.; Siproudhis, L.; Dewitte, M.; Landemaine, A.; Bellissant, E.; Bouguen, G. Predictors of relapse following infliximab de-escalation in patients with inflammatory bowel disease: The value of a strategy based on therapeutic drug monitoring. Aliment. Pharm. Ther. 2019, 49, 147–154. [Google Scholar] [CrossRef] [PubMed]
  89. Aguas Peris, M.; Bosó, V.; Navarro, B.; Marqués-Miñana, M.R.; Bastida, G.; Beltrán, B.; Iborra, M.; Sáez-González, E.; Monte-Boquet, E.; Poveda-Andrés, J.L.; et al. Serum Adalimumab Levels Predict Successful Remission and Safe Deintensification in Inflammatory Bowel Disease Patients in Clinical Practice. Inflamm. Bowel Diseases. 2017, 23, 1454–1460. [Google Scholar] [CrossRef] [Green Version]
  90. Petitcollin, A.; Brochard, C.; Siproudhis, L.; Tron, C.; Verdier, M.C.; Lemaitre, F.; Lucidarme, C.; Bouguen, G.; Bellissant, É. Pharmacokinetic Parameters of Infliximab Influence the Rate of Relapse after De-Escalation in Adults with Inflammatory Bowel Diseases. Clin. Pharm. Ther. 2019, 106, 605–615. [Google Scholar] [CrossRef]
  91. Drobne, D.; Bossuyt, P.; Breynaert, C.; Cattaert, T.; Casteele, N.V.; Compernolle, G.; Jürgens, M.; Ferrante, M.; Ballet, V.; Wollants, W.J.; et al. Withdrawal of immunomodulators after co-treatment does not reduce trough level of infliximab in patients with Crohn’s disease. Clin. Gastroenterol. Hepatol. 2015, 13, 514–521.e4. [Google Scholar] [CrossRef]
  92. Colombel, J.F.; Adedokun, O.J.; Gasink, C.; Gao, L.L.; Cornillie, F.J.; D’Haens, G.R.; Rutgeerts, P.J.; Reinisch, W.; Sandborn, W.J.; Hanauer, S.B. Combination Therapy With Infliximab and Azathioprine Improves Infliximab Pharmacokinetic Features and Efficacy: A Post Hoc Analysis. Clin. Gastroenterol. Hepatol. 2019, 17, 1525–1532.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Drobne, D.; Kurent, T.; Golob, S.; Švegl, P.; Rajar, P.; Hanžel, J.; Koželj, M.; Novak, G.; Smrekar, N.; Ferkolj, I.; et al. Optimised infliximab monotherapy is as effective as optimised combination therapy, but is associated with higher drug consumption in inflammatory bowel disease. Aliment. Pharm. Ther. 2019, 49, 880–889. [Google Scholar] [CrossRef] [PubMed]
  94. Lega, S.; Phan, B.L.; Rosenthal, C.J.; Gordon, J.; Haddad, N.; Pittman, N.; Benkov, K.J.; Dubinsky, M.C. Proactively Optimized Infliximab Monotherapy Is as Effective as Combination Therapy in IBD. Inflamm. Bowel Dis. 2019, 25, 134–141. [Google Scholar] [CrossRef] [PubMed]
  95. Baert, F.; Drobne, D.; Gils, A.; Casteele, N.V.; Hauenstein, S.; Singh, S.; Lockton, S.; Rutgeerts, P.; Vermeire, S. Early trough levels and antibodies to infliximab predict safety and success of reinitiation of infliximab therapy. Clin. Gastroenterol. Hepatol. 2014, 12, 1474–1481.e2; quiz e91. [Google Scholar] [CrossRef] [Green Version]
  96. Lau, C.; Dubinsky, M.; Melmed, G.; Vasiliauskas, E.; Berel, D.; McGovern, D.; Ippoliti, A.; Shih, D.; Targan, S.; Fleshner, P. The impact of preoperative serum anti-TNFα therapy levels on early postoperative outcomes in inflammatory bowel disease surgery. Ann. Surg. 2015, 261, 487–496. [Google Scholar] [CrossRef] [Green Version]
  97. Cohen, B.L.; Fleshner, P.; Kane, S.V.; Herfarth, H.H.; Palekar, N.; Farraye, F.A.; Leighton, J.A.; Katz, J.A.; Cohen, R.D.; Gerich, M.E.; et al. Prospective Cohort Study to Investigate the Safety of Preoperative Tumor Necrosis Factor Inhibitor Exposure in Patients With Inflammatory Bowel Disease Undergoing Intra-abdominal Surgery. Gastroenterology 2022, 163, 204–221. [Google Scholar] [CrossRef]
  98. Fumery, M.; Seksik, P.; Auzolle, C.; Munoz-Bongrand, N.; Gornet, J.M.; Boschetti, G.; Cotte, E.; Buisson, A.; Dubois, A.; Pariente, B.; et al. Postoperative Complications after Ileocecal Resection in Crohn’s Disease: A Prospective Study from the REMIND Group. Am. J. Gastroenterol. 2017, 112, 337–345. [Google Scholar] [CrossRef]
  99. Waterman, M.; Xu, W.; Dinani, A.; Steinhart, A.H.; Croitoru, K.; Nguyen, G.C.; McLeod, R.S.; Greenberg, G.R.; Cohen, Z.; Silverberg, M.S. Preoperative biological therapy and short-term outcomes of abdominal surgery in patients with inflammatory bowel disease. Gut 2013, 62, 387–394. [Google Scholar] [CrossRef]
  100. Liefferinckx, C.; Bottieau, J.; Toubeau, J.-F.; Thomas, D.; Rahier, J.-F.; Louis, E.; Baert, F.; Dewint, P.; Pouillon, L.; Lambrecht, G.; et al. Collecting New Peak and Intermediate Infliximab Levels to Predict Remission in Inflammatory Bowel Diseases. Inflamm. Bowel Dis. 2021, 28, 208–217. [Google Scholar] [CrossRef]
  101. Yoshihara, T.; Shinzaki, S.; Kawai, S.; Fujii, H.; Iwatani, S.; Yamaguchi, T.; Araki, M.; Hiyama, S.; Inoue, T.; Hayashi, Y.; et al. Tissue Drug Concentrations of Anti-tumor Necrosis Factor Agents Are Associated with the Long-term Outcome of Patients with Crohn’s Disease. Inflamm. Bowel Dis. 2017, 23, 2172–2179. [Google Scholar] [CrossRef]
  102. Afonso, J.; Lopes, S.; Gonçalves, R.; Caldeira, P.; Lago, P.; Tavares de Sousa, H.; Ramos, J.; Gonçalves, A.R.; Ministro, P.; Rosa, I.; et al. Proactive therapeutic drug monitoring of infliximab: A comparative study of a new point-of-care quantitative test with two established ELISA assays. Aliment. Pharmacol. Ther. 2016, 44, 684–692. [Google Scholar] [CrossRef] [PubMed]
  103. Cherry, M.; Dutzer, D.; Nasser, Y.; Berger, A.E.; Roblin, X.; Paul, S. Point-of-Care Assays Could Be Useful for Therapeutic Drug Monitoring of IBD Patients in a Proactive Strategy with Adalimumab. J. Clin. Med. 2020, 9. [Google Scholar] [CrossRef] [PubMed]
  104. Facchin, S.; Buda, A.; Cardin, R.; Agbariah, N.; Zingone, F.; De Bona, M.; Zaetta, D.; Bertani, L.; Ghisa, M.; Barberio, B.; et al. Rapid point-of-care anti-infliximab antibodies detection in clinical practice: Comparison with ELISA and potential for improving therapeutic drug monitoring in IBD patients. Ther. Adv. Gastroenterol. 2021, 14, 1756284821999902. [Google Scholar] [CrossRef]
  105. Dutzer, D.; Nasser, Y.; Berger, A.E.; Roblin, X.; Paul, S. Letter: New thresholds need to be defined when using point of care assays to monitor infliximab trough levels in IBD patients. Aliment. Pharmacol. Ther. 2018, 47, 1571–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Bossuyt, P.; Pouillon, L.; Claeys, S.; D’Haens, S.; Hoefkens, E.; Strubbe, B.; Marichal, D.; Peeters, H. Ultra-proactive Therapeutic Drug Monitoring of Infliximab Based on Point of Care Testing in Inflammatory Bowel Disease: Results of a Pragmatic Trial. J. Crohn’s Colitis 2021, 16, 199–206. [Google Scholar] [CrossRef]
  107. Strik, A.S.; Löwenberg, M.; Mould, D.R.; Berends, S.E.; Ponsioen, C.I.; van den Brande, J.M.H.; Jansen, J.M.; Hoekman, D.R.; Brandse, J.F.; Duijvestein, M.; et al. Efficacy of dashboard driven dosing of infliximab in inflammatory bowel disease patients; a randomized controlled trial. Scand. J. Gastroenterol. 2021, 56, 145–154. [Google Scholar] [CrossRef]
  108. Dubinsky, M.C.; Mendiolaza, M.L.; Phan, B.L.; Moran, H.R.; Tse, S.S.; Mould, D.R. Dashboard-Driven Accelerated Infliximab Induction Dosing Increases Infliximab Durability and Reduces Immunogenicity. Inflamm. Bowel Dis. 2022, 28, 1375–1385. [Google Scholar] [CrossRef]
  109. Papamichael, K.; Jairath, V.; Zou, G.; Cohen, B.; Ritter, T.; Sands, B.; Siegel, C.; Valentine, J.; Smith, M.; Vande Casteele, N.; et al. Proactive infliximab optimisation using a pharmacokinetic dashboard versus standard of care in patients with Crohn’s disease: Study protocol for a randomised, controlled, multicentre, open-label study (the OPTIMIZE trial). BMJ Open 2022, 12, e057656. [Google Scholar] [CrossRef]
  110. Smolen, J.S.; Van Der Heijde, D.M.; Clair, E.W.S.; Emery, P.; Bathon, J.M.; Keystone, E.; Maini, R.N.; Kalden, J.R.; Schiff, M.; Baker, D.; et al. Predictors of joint damage in patients with early rheumatoid arthritis treated with high-dose methotrexate with or without concomitant infliximab: Results from the ASPIRE trial. Arthritis Rheum. 2006, 54, 702–710. [Google Scholar] [CrossRef]
  111. Breedveld, F.C.; Weisman, M.H.; Kavanaugh, A.F.; Cohen, S.B.; Pavelka, K.; Vollenhoven, R.V.; Sharp, J.; Perez, J.L.; Spencer-Green, G.T. The PREMIER study: A multicenter, randomized, double-blind clinical trial of combination therapy with adalimumab plus methotrexate versus methotrexate alone or adalimumab alone in patients with early, aggressive rheumatoid arthritis who had not had previous methotrexate treatment. Arthritis Rheum. 2006, 54, 26–37. [Google Scholar] [PubMed]
  112. Brandse, J.F.; Mathôt, R.A.; van der Kleij, D.; Rispens, T.; Ashruf, Y.; Jansen, J.M.; Rietdijk, S.; Löwenberg, M.; Ponsioen, C.Y.; Singh, S.; et al. Pharmacokinetic Features and Presence of Antidrug Antibodies Associate With Response to Infliximab Induction Therapy in Patients With Moderate to Severe Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 2016, 14, 251–258.e1–2. [Google Scholar] [CrossRef] [PubMed]
  113. van Schaik, T.; Maljaars, J.P.; Roopram, R.K.; Verwey, M.H.; Ipenburg, N.; Hardwick, J.C.; Veenendaal, R.A.; van der Meulen-de Jong, A.E. Influence of combination therapy with immune modulators on anti-TNF trough levels and antibodies in patients with IBD. Inflamm. Bowel Dis. 2014, 20, 2292–2298. [Google Scholar] [CrossRef] [PubMed]
  114. Fraser, A.G.; Orchard, T.R.; Jewell, D.P. The efficacy of azathioprine for the treatment of inflammatory bowel disease: A 30 year review. Gut 2002, 50, 485–489. [Google Scholar] [CrossRef] [PubMed]
  115. Mantzaris, G.J. Thiopurines and Methotrexate Use in IBD Patients in a Biologic Era. Curr. Treat Options Gastroenterol. 2017, 15, 84–104. [Google Scholar] [CrossRef]
  116. Colombel, J.F.; Sandborn, W.J.; Reinisch, W.; Mantzaris, G.J.; Kornbluth, A.; Rachmilewitz, D.; Lichtiger, S.; D’Haens, G.; Diamond, R.H.; Broussard, D.L.; et al. Infliximab, azathioprine, or combination therapy for Crohn’s disease. N. Engl. J. Med. 2010, 362, 1383–1395. [Google Scholar] [CrossRef] [Green Version]
  117. Panaccione, R.; Ghosh, S.; Middleton, S.; Márquez, J.R.; Scott, B.B.; Flint, L.; van Hoogstraten, H.J.; Chen, A.C.; Zheng, H.; Danese, S.; et al. Combination therapy with infliximab and azathioprine is superior to monotherapy with either agent in ulcerative colitis. Gastroenterology 2014, 146, 392–400.e3. [Google Scholar] [CrossRef] [Green Version]
  118. Matsumoto, T.; Motoya, S.; Watanabe, K.; Hisamatsu, T.; Nakase, H.; Yoshimura, N.; Ishida, T.; Kato, S.; Nakagawa, T.; Esaki, M.; et al. Adalimumab Monotherapy and a Combination with Azathioprine for Crohn’s Disease: A Prospective, Randomized Trial. J. Crohn’s Colitis 2016, 10, 1259–1266. [Google Scholar] [CrossRef] [Green Version]
  119. Schröder, O.; Blumenstein, I.; Stein, J. Combining infliximab with methotrexate for the induction and maintenance of remission in refractory Crohn’s disease: A controlled pilot study. Eur. J. Gastroenterol. Hepatol. 2006, 18, 11–16. [Google Scholar] [CrossRef]
  120. Roblin, X.; Williet, N.; Boschetti, G.; Phelip, J.M.; Del Tedesco, E.; Berger, A.E.; Vedrines, P.; Duru, G.; Peyrin-Biroulet, L.; Nancey, S.; et al. Addition of azathioprine to the switch of anti-TNF in patients with IBD in clinical relapse with undetectable anti-TNF trough levels and antidrug antibodies: A prospective randomised trial. Gut 2020, 69, 1206–1212. [Google Scholar] [CrossRef]
  121. Feagan, B.G.; McDonald, J.W.; Panaccione, R.; Enns, R.A.; Bernstein, C.N.; Ponich, T.P.; Bourdages, R.; Macintosh, D.G.; Dallaire, C.; Cohen, A.; et al. Methotrexate in combination with infliximab is no more effective than infliximab alone in patients with Crohn’s disease. Gastroenterology 2014, 146, 681–688.e1. [Google Scholar] [CrossRef] [PubMed]
  122. Targownik, L.E.; Benchimol, E.I.; Bernstein, C.N.; Singh, H.; Tennakoon, A.; Zubieta, A.A.; Coward, S.; Jones, J.; Kaplan, G.G.; Kuenzig, M.E.; et al. Combined Biologic and Immunomodulatory Therapy is Superior to Monotherapy for Decreasing the Risk of Inflammatory Bowel Disease-Related Complications. J. Crohn’s Colitis 2020, 14, 1354–1363. [Google Scholar] [CrossRef] [PubMed]
  123. Mahmoud, R.; Schultheiss, H.P.; Louwers, J.; van der Kaaij, M.; van Hellemondt, B.; Mahmmod, N.; van Boeckel, P.; Jharap, B.; Fidder, H.; Oldenburg, B. Immunomodulator Withdrawal From Anti-TNF Therapy Is Not Associated With Loss of Response in Inflammatory Bowel Disease. Clin. Gastroenterol. Hepatol. 2022, 20, 2577–2587.e6. [Google Scholar] [CrossRef] [PubMed]
  124. Lucas Ramos, J.; Suárez Ferrer, C.; Poza Cordón, J.; Sánchez Azofra, M.; Rueda García, J.L.; Martin Arranz, E.; Yebra Carmona, J.; Andaluz García, I.; Martín Arranz, M.D. Optimization of azathioprine dose in combined treatment with anti-TNF-alpha in inflammatory bowel disease. Gastroenterol. Hepatol. 2021, 44, 337–345. [Google Scholar] [CrossRef]
  125. Nawaz, A.; Glick, L.R.; Chaar, A.; Li, D.K.; Gaidos, J.K.J.; Proctor, D.D.; Al-Bawardy, B. Impact of thiopurine dose in anti-tumor necrosis factor combination therapy on outcomes in inflammatory bowel disease. Ann. Gastroenterol. 2023, 36, 39–44. [Google Scholar] [CrossRef]
  126. Roblin, X.; Boschetti, G.; Williet, N.; Nancey, S.; Marotte, H.; Berger, A.; Phelip, J.M.; Peyrin-Biroulet, L.; Colombel, J.F.; Del Tedesco, E.; et al. Azathioprine dose reduction in inflammatory bowel disease patients on combination therapy: An open-label, prospective and randomised clinical trial. Aliment. Pharm. Ther. 2017, 46, 142–149. [Google Scholar] [CrossRef] [Green Version]
  127. Lambrescak, E.; Vaysse, T.; Allez, M.; Ungar, B.; Gleizes, A.; Hacein-Bey, S.; Chowers, Y.; Roblin, X.; Kopylov, U.; Rachas, A.; et al. Duration of combination therapy and risk of treatment failure in patients with inflammatory bowel disease. Clin. Res. Hepatol. Gastroenterol. 2021, 45, 101503. [Google Scholar] [CrossRef]
  128. Oussalah, A.; Chevaux, J.B.; Fay, R.; Sandborn, W.J.; Bigard, M.A.; Peyrin-Biroulet, L. Predictors of infliximab failure after azathioprine withdrawal in Crohn’s disease treated with combination therapy. Am. J. Gastroenterol. 2010, 105, 1142–1149. [Google Scholar] [CrossRef]
  129. Chen, C.; Hartzema, A.G.; Xiao, H.; Wei, Y.J.; Chaudhry, N.; Ewelukwa, O.; Glover, S.C.; Zimmermann, E.M. Real-world Pattern of Biologic Use in Patients With Inflammatory Bowel Disease: Treatment Persistence, Switching, and Importance of Concurrent Immunosuppressive Therapy. Inflamm. Bowel Dis. 2019, 25, 1417–1427. [Google Scholar] [CrossRef]
  130. Fousekis, F.S.; Papamichael, K.; Kourtis, G.; Albani, E.N.; Orfanidou, A.; Saridi, M.; Katsanos, K.H.; Christodoulou, D.K. The efficacy of immunomodulators in the prevention and suppression of anti-drug antibodies to anti-tumor necrosis factor therapy in inflammatory bowel disease. Ann. Gastroenterol. 2022, 35, 1–7. [Google Scholar] [CrossRef]
  131. Ben-Horin, S.; Waterman, M.; Kopylov, U.; Yavzori, M.; Picard, O.; Fudim, E.; Awadie, H.; Weiss, B.; Chowers, Y. Addition of an immunomodulator to infliximab therapy eliminates antidrug antibodies in serum and restores clinical response of patients with inflammatory bowel disease. Clin. Gastroenterol. Hepatol. 2013, 11, 444–447. [Google Scholar] [CrossRef] [PubMed]
  132. Kirchgesner, J.; Lemaitre, M.; Carrat, F.; Zureik, M.; Carbonnel, F.; Dray-Spira, R. Risk of Serious and Opportunistic Infections Associated With Treatment of Inflammatory Bowel Diseases. Gastroenterology 2018, 155, 337–346.e10. [Google Scholar] [CrossRef] [Green Version]
  133. Lemaitre, M.; Kirchgesner, J.; Rudnichi, A.; Carrat, F.; Zureik, M.; Carbonnel, F.; Dray-Spira, R. Association Between Use of Thiopurines or Tumor Necrosis Factor Antagonists Alone or in Combination and Risk of Lymphoma in Patients with Inflammatory Bowel Disease. JAMA 2017, 318, 1679–1686. [Google Scholar] [CrossRef] [PubMed]
  134. Sturm, A.; Maaser, C.; Mendall, M.; Karagiannis, D.; Karatzas, P.; Ipenburg, N.; Sebastian, S.; Rizzello, F.; Limdi, J.; Katsanos, K.; et al. European Crohn’s and Colitis Organisation Topical Review on IBD in the Elderly. J. Crohn’s Colitis 2017, 11, 263–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Beaugerie, L. Lymphoma: The bête noire of the long-term use of thiopurines in adult and elderly patients with inflammatory bowel disease. Gastroenterology 2013, 145, 927–930. [Google Scholar] [CrossRef] [PubMed]
  136. Peyrin-Biroulet, L.; Sandborn, W.; Sands, B.E.; Reinisch, W.; Bemelman, W.; Bryant, R.V.; D’Haens, G.; Dotan, I.; Dubinsky, M.; Feagan, B.; et al. Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE): Determining Therapeutic Goals for Treat-to-Target. Am. J. Gastroenterol. 2015, 110, 1324–1338. [Google Scholar] [CrossRef]
  137. Bouguen, G.; Peyrin-Biroulet, L. Surgery for adult Crohn’s disease: What is the actual risk? Gut 2011, 60, 1178–1181. [Google Scholar] [CrossRef]
  138. Peyrin-Biroulet, L.; Reinisch, W.; Colombel, J.F.; Mantzaris, G.J.; Kornbluth, A.; Diamond, R.; Rutgeerts, P.; Tang, L.K.; Cornillie, F.J.; Sandborn, W.J. Clinical disease activity, C-reactive protein normalisation and mucosal healing in Crohn’s disease in the SONIC trial. Gut 2014, 63, 88–95; quiz 20. [Google Scholar] [CrossRef]
  139. Mosli, M.H.; Zou, G.; Garg, S.K.; Feagan, S.G.; MacDonald, J.K.; Chande, N.; Sandborn, W.J.; Feagan, B.G. C-Reactive Protein, Fecal Calprotectin, and Stool Lactoferrin for Detection of Endoscopic Activity in Symptomatic Inflammatory Bowel Disease Patients: A Systematic Review and Meta-Analysis. Am. J. Gastroenterol. 2015, 110, 802–819; quiz 20. [Google Scholar] [CrossRef]
  140. Rokkas, T.; Portincasa, P.; Koutroubakis, I.E. Fecal calprotectin in assessing inflammatory bowel disease endoscopic activity: A diagnostic accuracy meta-analysis. J. Gastrointestin. Liver Dis. 2018, 27, 299–306. [Google Scholar] [CrossRef] [Green Version]
  141. Kostas, A.; Siakavellas, S.I.; Kosmidis, C.; Takou, A.; Nikou, J.; Maropoulos, G.; Vlachogiannakos, J.; Papatheodoridis, G.V.; Papaconstantinou, I.; Bamias, G. Fecal calprotectin measurement is a marker of short-term clinical outcome and presence of mucosal healing in patients with inflammatory bowel disease. World J. Gastroenterol. 2017, 23, 7387–7396. [Google Scholar] [CrossRef]
  142. Diederen, K.; Hoekman, D.R.; Leek, A.; Wolters, V.M.; Hummel, T.Z.; de Meij, T.G.; Koot, B.G.; Tabbers, M.M.; Benninga, M.A.; Kindermann, A. Raised faecal calprotectin is associated with subsequent symptomatic relapse, in children and adolescents with inflammatory bowel disease in clinical remission. Aliment. Pharm. Ther. 2017, 45, 951–960. [Google Scholar] [CrossRef] [Green Version]
  143. Gisbert, J.P.; Marín, A.C.; Chaparro, M. Systematic review: Factors associated with relapse of inflammatory bowel disease after discontinuation of anti-TNF therapy. Aliment. Pharm. Ther. 2015, 42, 391–405. [Google Scholar] [CrossRef] [PubMed]
  144. Reinisch, W.; Wang, Y.; Oddens, B.J.; Link, R. C-reactive protein, an indicator for maintained response or remission to infliximab in patients with Crohn’s disease: A post-hoc analysis from ACCENT I. Aliment. Pharm. Ther. 2012, 35, 568–576. [Google Scholar] [CrossRef] [PubMed]
  145. Song, J.H.; Hong, S.N.; Lee, J.E.; Kim, K.; Kim, T.J.; Kim, E.R.; Chang, D.K.; Kim, Y.H. C-Reactive protein reduction rate following initiation of anti-tumor necrosis factor α induction therapy predicts secondary loss of response in patients with Crohn’s disease. Scand. J. Gastroenterol. 2019, 54, 876–885. [Google Scholar] [CrossRef]
  146. Colombel, J.F.; Panaccione, R.; Bossuyt, P.; Lukas, M.; Baert, F.; Vaňásek, T.; Danalioglu, A.; Novacek, G.; Armuzzi, A.; Hébuterne, X.; et al. Effect of tight control management on Crohn’s disease (CALM): A multicentre, randomised, controlled phase 3 trial. Lancet 2017, 390, 2779–2789. [Google Scholar] [CrossRef]
  147. Ungaro, R.C.; Yzet, C.; Bossuyt, P.; Baert, F.J.; Vanasek, T.; D’Haens, G.R.; Joustra, V.W.; Panaccione, R.; Novacek, G.; Reinisch, W.; et al. Deep Remission at 1 Year Prevents Progression of Early Crohn’s Disease. Gastroenterology 2020, 159, 139–147. [Google Scholar] [CrossRef]
  148. Arias, M.T.; Casteele, N.V.; Vermeire, S.; de Buck van Overstraeten, A.; Billiet, T.; Baert, F.; Wolthuis, A.; Van Assche, G.; Noman, M.; Hoffman, I.; et al. A panel to predict long-term outcome of infliximab therapy for patients with ulcerative colitis. Clin. Gastroenterol. Hepatol. 2015, 13, 531–538. [Google Scholar] [CrossRef] [PubMed]
  149. Mao, R.; Xiao, Y.L.; Gao, X.; Chen, B.L.; He, Y.; Yang, L.; Hu, P.J.; Chen, M.H. Fecal calprotectin in predicting relapse of inflammatory bowel diseases: A meta-analysis of prospective studies. Inflamm. Bowel Dis. 2012, 18, 1894–1899. [Google Scholar] [CrossRef]
  150. Roblin, X.; Marotte, H.; Leclerc, M.; Del Tedesco, E.; Phelip, J.M.; Peyrin-Biroulet, L.; Paul, S. Combination of C-reactive protein, infliximab trough levels, and stable but not transient antibodies to infliximab are associated with loss of response to infliximab in inflammatory bowel disease. J. Crohn’s Colitis 2015, 9, 525–531. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Sollelis, E.; Quinard, R.M.; Bouguen, G.; Goutte, M.; Goutorbe, F.; Bouvier, D.; Pereira, B.; Bommelaer, G.; Buisson, A. Combined evaluation of biomarkers as predictor of maintained remission in Crohn’s disease. World J. Gastroenterol. 2019, 25, 2354–2364. [Google Scholar]
  152. Danese, S.; Vermeire, S.; D’Haens, G.; Panés, J.; Dignass, A.; Magro, F.; Nazar, M.; Le Bars, M.; Lahaye, M.; Ni, L.; et al. Treat to target versus standard of care for patients with Crohn’s disease treated with ustekinumab (STARDUST): An open-label, multicentre, randomised phase 3b trial. Lancet Gastroenterol. Hepatol. 2022, 7, 294–306. [Google Scholar]
  153. Gupta, A.; Yu, A.; Peyrin-Biroulet, L.; Ananthakrishnan, A.N. Treat to Target: The Role of Histologic Healing in Inflammatory Bowel Diseases: A Systematic Review and Meta-analysis. Clin. Gastroenterol. Hepatol. 2021, 19, 1800–1813.e4. [Google Scholar]
  154. Yoon, H.; Jangi, S.; Dulai, P.S.; Boland, B.S.; Prokop, L.J.; Jairath, V.; Feagan, B.G.; Sandborn, W.J.; Singh, S. Incremental Benefit of Achieving Endoscopic and Histologic Remission in Patients With Ulcerative Colitis: A Systematic Review and Meta-Analysis. Gastroenterology 2020, 159, 1262–1275.e7. [Google Scholar] [PubMed]
  155. Castiglione, F.; Imperatore, N.; Testa, A.; De Palma, G.D.; Nardone, O.M.; Pellegrini, L.; Caporaso, N.; Rispo, A. One-year clinical outcomes with biologics in Crohn’s disease: Transmural healing compared with mucosal or no healing. Aliment. Pharm. Ther. 2019, 49, 1026–1039. [Google Scholar]
  156. Fernandes, S.R.; Rodrigues, R.V.; Bernardo, S.; Cortez-Pinto, J.; Rosa, I.; da Silva, J.P.; Gonçalves, A.R.; Valente, A.; Baldaia, C.; Santos, P.M.; et al. Transmural Healing Is Associated with Improved Long-term Outcomes of Patients with Crohn’s Disease. Inflamm. Bowel Dis. 2017, 23, 1403–1409. [Google Scholar] [CrossRef]
  157. Messadeg, L.; Hordonneau, C.; Bouguen, G.; Goutorbe, F.; Reimund, J.M.; Goutte, M.; Boucher, A.L.; Scanzi, J.; Reymond, M.; Allimant, C.; et al. Early Transmural Response Assessed Using Magnetic Resonance Imaging Could Predict Sustained Clinical Remission and Prevent Bowel Damage in Patients with Crohn’s Disease Treated with Anti-Tumour Necrosis Factor Therapy. J. Crohn’s Colitis 2020, 14, 1524–1534. [Google Scholar]
  158. Singh, S.; Murad, M.H.; Fumery, M.; Dulai, P.S.; Sandborn, W.J. First- and Second-Line Pharmacotherapies for Patients With Moderate to Severely Active Ulcerative Colitis: An Updated Network Meta-Analysis. Clin. Gastroenterol. Hepatol. 2020, 18, 2179–2191.e6. [Google Scholar]
Table 1. Factors affecting the pharmacokinetics of anti-TNF agents [10,19,20].
Table 1. Factors affecting the pharmacokinetics of anti-TNF agents [10,19,20].
Factors Effect on TCs
Anti-drug antibodies (+)
Combination therapy with immunomodulators
Low albumin levels
High CRP levels
High BMI
Male gender
High serum TNF concentrations
Polymorphisms in the neonatal Fc receptor
Pegylation
↓: decrease; ↑: increase.
Table 3. TDM thresholds during induction treatment and associated short- and long-term therapeutic outcomes for anti-TNF agents.
Table 3. TDM thresholds during induction treatment and associated short- and long-term therapeutic outcomes for anti-TNF agents.
Author,
Year [Ref.]
Study DesignDisease Type Anti-TNF AgentPatients (N)TCs (mg/L)WeekOutcome
Dreesen,
2020 [55]
Post hoc analysis of RCTCDIFX 122>23.1
>10
2
6
Endoscopic remission week 12
Endoscopic remission week 12
Davidov,
2017 [54]
Retrospective CD
fistulizing
IFX36>9.3
>7.3
2
6
Fistula response week 14
Fistula response week 14
Papamichael,
2021 [56]
Post hoc analysis of RCTCD fistulizingIFX 282≥20.2
≥15
≥7.2
2
6
14
Complete remission week 14
Complete remission week 14
Complete remission week 14
(defined as combined complete fistula response and CRP normalization)
Clarkston,
2019 [57]
ProspectiveCD pediatricIFX72≥26.7
≥15.9
2
6
Clinical response week 14
Clinical response week 14
Gonzi,
2017 [58]
ProspectiveCDIFX
(biosimilar)
184>16.9
>20.4
2Clinical response week 14
Clinical remission week 14
Gonzi,
2017 [58]
ProspectiveUCIFX
(biosimilar)
107>11.5
>15.3
2Clinical response week 14
Clinical remission week 14
Gonzi,
2017 [58]
ProspectiveUCIFX
(biosimilar)
107>11.5
>14.5
2Clinical response week 30
Clinical remission week 30
Kobayashi, 2016 [59]Post hoc analysis of RCTUCIFX 82>21.32Clinical response week 14
Bar Yoseph, 2018 [60]Retrospective case controlCDIFX 140<6.82Primary non-response week 14
Vande Castelle, 2019 [61]Post hoc analysis of RCTsUCIFX 484≥18.6
>10.6
≥5.1
≥6.7
2
6
14
14
Mayo endoscopic score ≤1 week 8
Mayo endoscopic score ≤1 week 8
Mayo endoscopic score ≤1 week 30
Mayo endoscopic score 0 week 30
Papamichael, 2016 [62]RetrospectiveUCIFX 101≥28.3
≥15
≥2.1
2
6
14
Short-term mucosal healing weeks 10–12 (Mayo endoscopic score ≤ 1) Short-term mucosal healing weeks 10–12 (Mayo endoscopic score ≤ 1) Short-term mucosal healing weeks 10–12 (Mayo endoscopic score ≤ 1)
Adedokun, 2014 [38]Post hoc analysis of RCTsUCIFX728≥22
>5.1
6
14
Clinical response week 8
Clinical response week 30
Kennedy,
2019 [18]
ProspectiveCDIFX955>714Remission week 14 and 54 (CRP ≤ 3 mg/L and HBI≤ 4, no ongoing steroid therapy, and no exit due to treatment failure)
Cornille,
2014 [53]
Post hoc analysis of RCTCDIFX 291≥ 3.514Sustained clinical response up to week 54
Tighe,
2017 [63]
ProspectiveCD/UCIFX 17>4.814Predicts clinical response week 14
Bortlik,
2013 [64]
RetrospectiveCDIFX84>314Sustained clinical response, decreased risk of treatment failure
Ungar,
2018 [65]
ProspectiveCDADA98>6.72Clinical remission week 14
Verstockt,
2018 [66]
Prospective CDADA116>12
<8.3
4
4
Biological remission at week 12
Positive anti-drug antibodies by week 12
Vande Castelle, 2019 [67] ProspectiveCDADA28>7.34Clinical remission week 12
Papamichael, 2017 [68] Retrospective UCADA43≥7.54Mucosal healing weeks 8–14
Baert,
2014 [69]
RetrospectiveUCADA73≥4.6
≥7
4 Clinical response week 12
Clinical response week 52
Tighe,
2017 [63]
ProspectiveCD/UCADA 18>3.54Predicts clinical response week 4
Baert,
2016 [70]
RetrospectiveCDADA148<54Development of anti-drug antibodies
RCT: randomized clinical trial; CD: Crohn’s disease; UC: ulcerative colitis; IFX: infliximab; ADA: adalimumab; CRP: C-reactive protein; TCs: trough concentrations.
Table 4. Summary of the studies using combination therapy versus monotherapy in patients with IBD.
Table 4. Summary of the studies using combination therapy versus monotherapy in patients with IBD.
Author, Year [Ref.]Study DesignPatients (N)Male (%)Disease Type (UC/CD)Age (Median Years)Disease Duration (Median Years)Follow-up (Months, Median)MonotherapyCombination TherapyConcomitant TherapyPrimary EndpointOutcomes
Colombel, 2010 (SONIC) [116]Randomized, double-blind trial50852CD342.311.5IFX standard dose 5 mg/kgIFX + AZA standard dose 2.5 mg/kgMesalamine, steroidsSteroid-free clinical remission at week 26Combo better outcome vs. monotherapy (p = 0.02)
Panaccione, 2014 (SUCCESS) [117]Randomized, double-blind trial23954UC38NA4IFX standard dose 5 mg/kgIFX + AZA standard dose 2.5 mg/kgSteroids (tapering after the induction)Steroid-free remission at week 16Combo better outcomes vs. monotherapy ( p= 0.017)
Schröder, 2006 [119]Randomized, controlled, open label, clinical trial1942CD 35911.2IFX 5 mg/kgIFX + MTX 20 mg in weeks 0–5 and then 20 mg orally weekly5 ASA steroids (tapering)Clinical remission (CDAI < 150)Combo achieved remission in 91% vs. 50% in monotherapy (p = 0.04), earlier (2 w vs. 18 w)
Matsumoto, 2016 (Diamond trial) [118]Multicentre, randomized, prospective, open label study17672CD31At least 3 months12ADA standard dose (40 mg/2 w)ADA standard dose + AZA 25–100 mg5 ASA, steroidsClinical remission (SCAI < 150) at week 26No difference in clinical remission in the two groups (p = 0.63)
Roblin, 2020 [120]Randomized, open label and prospective trial10049CD/UC39.53.524IFX standard dose (to pts with previous failure to ADA intensified dose)
ADA standard dose (to pts with previous failure to IFX intensified dose)
IFX standard dose + AZA 2.5 mg/Kg
ADA standard dose + AZA 2.5 mg/kg
NAClinical failure and occurrence of undesirable effects at 24 monthsCombo better outcome vs. monotherapy, (p < 0.001)
Feagan, 2014 [121]Double-blind, placebo-controlled trial12656CD39.51011.5IFX (5 mg/kg at weeks 1, 3, 7, 14, 22, 30, 38 and 46)IFX (5 mg/kg at weeks 1, 3, 7, 14, 22, 30, 38 and 46) + MTX (10 mg/w to 20 mg/w at week 3, and to 25 mg/w at week 5 till week 50)Folic acid, antibiotics, steroids taperingTime to clinical failureNo significant difference (p = 0.63)
Targownik, 2020 [122]Single, open label, retrospective clinical trial78,41350.1CD/UCNA (the majority between 25–65)1.2–8.7NAIFX or ADAIFX +MTX/thiopurine,
ADA + MTX/thiopurine
NAThe first occurrence of treatment failure (IBD-related hospitalization, IBD-related surgery, new/recurrent corticosteroid use or anti-TNF switch for 52 weeks)Combo therapy was associated with a significant decrease in treatment ineffectiveness for both CD and UC (aHR 0.77 (95% CI 0.66–0.90) for CD) (0.72 (95% CI 0.62–0.84) for UC)
Mahmoud, 2022 [123]Retrospective cohort study54345CD/UC33.54.120.4IFX/ADA monotherapy after discontinuation of concomitant MTX/thiopurineIFX/ADA + MTX/thiopurineNALOR, detection of anti-drug antibodies to anti-TNF therapyImmunomodulator withdrawal did not increase the risk of LOR (aHR 1.08; 95% CI, 0.72–1.61),but it was associated with an increased risk of anti-drug antibodies in the entire cohort (aHR, 2.14; 95% CI, 1.17–3.94)
MTX: methotrexate; AZA; azathioprine; CD: Crohn’s disease; UC: ulcerative colitis; IFX: infliximab; ADA: adalimumab; NA: not applicable; LOR: loss of response; TCs: trough concentrations.
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

Orfanoudaki, E.; Foteinogiannopoulou, K.; Theodoraki, E.; Koutroubakis, I.E. Recent Advances in the Optimization of Anti-TNF Treatment in Patients with Inflammatory Bowel Disease. J. Clin. Med. 2023, 12, 2452. https://doi.org/10.3390/jcm12072452

AMA Style

Orfanoudaki E, Foteinogiannopoulou K, Theodoraki E, Koutroubakis IE. Recent Advances in the Optimization of Anti-TNF Treatment in Patients with Inflammatory Bowel Disease. Journal of Clinical Medicine. 2023; 12(7):2452. https://doi.org/10.3390/jcm12072452

Chicago/Turabian Style

Orfanoudaki, Eleni, Kalliopi Foteinogiannopoulou, Eirini Theodoraki, and Ioannis E. Koutroubakis. 2023. "Recent Advances in the Optimization of Anti-TNF Treatment in Patients with Inflammatory Bowel Disease" Journal of Clinical Medicine 12, no. 7: 2452. https://doi.org/10.3390/jcm12072452

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