Next Article in Journal
Scheimpflug Corneal Densitometry Patterns at the Graft–Host Interface in DMEK and DSAEK: A 12-Month Longitudinal Comparative Study
Next Article in Special Issue
What Should We Know about Drug Levels and Therapeutic Drug Monitoring during Pregnancy and Breastfeeding in Inflammatory Bowel Disease under Biologic Therapy?
Previous Article in Journal
PSMA PET/CT in Castration-Resistant Prostate Cancer: Myth or Reality?
Previous Article in Special Issue
Higher Adalimumab Trough Levels Are Associated with Histologic Remission and Mucosal Healing in Inflammatory Bowel Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Drug Clearance in Patients with Inflammatory Bowel Disease Treated with Biologics

by
Tina Deyhim
,
Adam S. Cheifetz
and
Konstantinos Papamichael
*
Center for Inflammatory Bowel Diseases, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(22), 7132; https://doi.org/10.3390/jcm12227132
Submission received: 30 October 2023 / Revised: 4 November 2023 / Accepted: 15 November 2023 / Published: 16 November 2023

Abstract

:
Biological therapy is very effective for treating patients with moderate to severe inflammatory bowel disease (IBD). However, up to 40% can have primary non-response, and up to 50% of the patients can experience a loss of response to anti-tumor necrosis factor therapy. These undesirable outcomes can be attributed to either a mechanistic failure or pharmacokinetic (PK) issues characterized by an inadequate drug exposure and a high drug clearance. There are several factors associated with accelerated clearance of biologics including increased body weight, low serum albumin and immunogenicity. Drug clearance has gained a lot of attention recently as cumulative data suggest that there is an association between drug clearance and therapeutic outcomes in patients with IBD. Moreover, clearance is used by model informed precision dosing (MIDP) tools, or PK dashboards, to adjust the dosing for reaching a target drug concentration threshold towards a more personalized application of TDM. However, the role of drug clearance in clinical practice is yet to be determined. This comprehensive review aims to present data regarding the variables affecting the clearance of specific biologics, the association of clearance with therapeutic outcomes and the role of clearance monitoring and MIPD in patients with IBD.

Graphical Abstract

1. Introduction

Biological therapy is very effective for treating patients with moderate to severe inflammatory bowel diseases (IBD) such as Crohn’s disease (CD) and ulcerative colitis (UC). However, up to 40% and 80% of the patients can have primary non-response and primary non-remission to anti-tumor necrosis factor (anti-TNF) therapy, respectively [1]. Moreover, up to half of the patients with IBD may experience secondary loss of response [2]. Therapeutic drug monitoring (TDM) can help to explain these unwanted outcomes attributed either to a mechanistic failure in the presence of adequate drug concentration or pharmacokinetic (PK) issues characterized by an inadequate drug exposure and a high drug clearance [3].
Drug clearance is expressed as the volume of plasma in the vascular compartment that is cleared of drug per unit of time, and it is estimated using Bayesian modeling [3]. Mechanisms underlying the clearance of monoclonal antibodies include intracellular catabolism and endocytosis, increase in target load, protein-losing enteropathy and binding to anti-drug antibodies. There are several factors associated with the above mechanisms that can influence the clearance of biologics including body weight, serum albumin and immunogenicity (Figure 1) [4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46]. In particular, increased body weight, decreased albumin and anti-drug antibodies (ADAs) have been associated with higher clearance for almost all the biologics currently used in IBD (Figure 1).
Drug clearance has gained a lot of attention recently as an increased estimated baseline clearance may identify patients at high risk of underexposure to biologics and PK-related treatment failure. Furthermore, a sudden increase in clearance during therapy can precede the decrease in drug concentrations and may predict a flare prior to the development of symptoms or low drug concentrations. Moreover, cumulative data suggest that there is an association between drug clearance and therapeutic outcomes in patients with IBD (Table 1) [17,21,22,24,32,36,40,47,48,49,50,51].
Finally, clearance is used by model informed precision dosing (MIDP) tools, or PK dashboards, to adjust the dosing for reaching a target drug concentration threshold, typically depending on the desired therapeutic set by the treating physician. However, the role of drug clearance in clinical practice is yet to be determined.
The aim of this comprehensive review is to present data regarding the variables affecting the clearance of specific biologics, the association of clearance with therapeutic outcomes and the role of clearance monitoring and MIPD in patients with IBD.

2. Variables Affecting Clearance of Biologics

2.1. Infliximab

Numerous factors have been shown to accelerate infliximab clearance in patients with IBD. The most frequently identified variables from studies were increased body weight, lower albumin and immunogenicity (Figure 1) [4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20]. Other factors that can lead to high clearance include male gender [5,12,18], induction compared to maintenance therapy [14,21,22], prior biologic therapy [13], an increased Crohn’s disease activity index (CDAI) [11], as well as elevated fecal calprotectin [10,11], C-reactive protein (CRP) [14,23,24] and erythrocyte sedimentation rate (ESR) [10]. On the other hand, concomitant immunomodulators (IMMs) (thiopurines/methotrexate) were found to decrease infliximab clearance by around 15% [6,9,17]. This is probably due to the fact that IMM can prevent or suppress immunogenicity as well as decrease the TNF target-antigen burden and consequently the target-mediated elimination of infliximab; although, the mechanisms by which IMM decrease infliximab clearance have not been clearly defined yet [17].
A post hoc analysis of the REACH and ACCENT I randomized controlled trials (RCTs) showed that the clearance was higher in those who developed ADAs or had low baseline albumin [6]. Moreover, concurrent IMM use decreased infliximab clearance by 14% [6]. Data from the Phase I study of maintenance subcutaneous therapy with the infliximab biosimilar CT-P13 in patients with IBD showed that clearance could be increased by 43.2%, 30.1% and 39% by an elevated body weight (from 70 to 120 kg), lower albumin (from 44 to 32 gr/L) and positive ADAs, respectively [13]. The same study showed that patients previously treated with biologics also exhibited a higher drug clearance compared to anti-TNF naïve patients, probably reflecting a more refractory disease and a higher tendency to develop ADAs [13]. A multi-center prospective study including pediatric patients with CD showed that median clearance was higher in ADA-positive compared to ADA-negative patients (0.0111 L/h vs. 0.0094 L/h, p < 0.001) [8]. A post hoc analysis of the TAILORIX RCT showed that higher infliximab clearance in patients with CD was associated with increased fecal calprotectin, decreased albumin, increased CDAI and immunogenicity [11]. In a PK analysis of the ACT1 and ACT2, RCTs increased body weight, and lower albumin predicted a higher infliximab clearance in UC. The same study also showed that the mean clearance was 47.1% higher for patients with positive ADAs [12]. A prospective PK study regarding patients with IBD showed that concomitant azathioprine use led to a 15.1% decrease in infliximab clearance [17].

2.2. Adalimumab

Several factors have been shown to accelerate adalimumab clearance in patients with IBD including body weight, lower albumin and immunogenicity. (Figure 1) [26,27,28,29,30]. Other factors associated with increased clearance include prior biologic exposure [50], every week dosing [27], UC [29], elevated CRP [29,30] and higher fecal calprotectin [29].
A post hoc analysis of the SERENE CD and UC RCTs identified increased body weight, lower baseline albumin and immunogenicity as variables associated with higher drug clearance [29]. Other factors associated with accelerated adalimumab clearance were UC and elevated baseline fecal calprotectin and CRP levels. The PK analysis of the IMAgINE-1 RCT showed that increased body weight, higher baseline CRP and lower baseline albumin levels were associated with a greater clearance of adalimumab in pediatric CD [30]. In a prospective multicenter study, positive ADAs increased the clearance of a typical patient with CD from 0.330 L/d to 0.525 L/d [26]. Similarly, another study showed a four-fold increase in adalimumab clearance in the presence of ADAs in patients with CD [27].

2.3. Certolizumab Pegol

Variables associated with a higher clearance of certolizumab pegol in patients with CD include increased body weight, lower albumin, immunogenicity and elevated CRP (Figure 1) [31,32].
In a certolizumab pegol PK modeling study, body weight (46.8–100.5 kg) increased the median clearance from 82 to 120%; albumin (32–48 g/L) decreased drug clearance from 123 to 85%; CRP (0.5–54.0 mg/L) increased the median clearance from 83 to 113%; and positive ADAs increased the median clearance by 142–174% [31]. A PK analysis on phase 2 and 3 certolizumab pegol clinical trials demonstrated that the predicted baseline certolizumab pegol clearance of ≥0.5 L/d was associated with a higher probability of a sub-therapeutic drug concentration at week 6 [32]. The same study using the PRECiSE 1 and 2 RCTs datasets identified a baseline certolizumab pegol clearance associated with composite remission (CDAI ≤ 150 and fecal calprotectin concentration ≤ 250 μg/g) at both week 6 [odds ratio (OR): 0.92; 95% confidence interval (CI): 0.87–0.96] and week 26 (OR: 0.93; 95%CI 0.88–0.97) [32].

2.4. Golimumab

Limited data show that the factors associated with a higher golimumab clearance in patients with UC include increased body weight, lower albumin, immunogenicity, previous biological therapy and a lack of concomitant methotrexate use (Figure 1) [33,34].
A population PK model developed on pooled data from studies regarding adults (NCT00487539 and NCT00488631) and children with moderate-to-severe UC (NCT 01900574), as well as children with polyarticular juvenile idiopathic arthritis (NCT01230827), showed that golimumab clearance increased with body weight and immunogenicity, while the clearance decreased with higher baseline albumin and concomitant methotrexate. Patients receiving methotrexate had a 17% lower clearance compared to those on golimumab monotherapy, and positive ADAs were associated with a 21% higher drug clearance [34]. In the same line, in a PK study of 56 patients with moderate-to-severe UC, golimumab clearance was 31% higher when ADAs were detected [33].

2.5. Vedolizumab

Several factors have been identified to accelerate vedolizumab clearance in patients with IBD; the most important ones being increased body weight, lower albumin and immunogenicity (Figure 1) [35,36,37,38,39,40]. Other factors that can lead to a high clearance include prior biological therapy [36,38,39], older age [36], and higher endoscopic Mayo score [35], as well as elevated fecal calprotectin [36,39], CRP [39] and ESR [40].
The prospective multicenter LOVE-CD (NCT02646683) study showed that vedolizumab clearance was higher in patients with CD with lower serum albumin concentrations (+26%, from 41 g/L to 28 g/L), presence of ADAs (+89% compared to no ADAs) and previous exposure to other biologic therapies (+25% compared to no biologic naïve patients) [38]. A PK study of the GEMINI 1 RCT demonstrated that the vedolizumab clearance was higher in patients with UC with a history of prior anti-TNF treatment, lower serum albumin and higher fecal calprotectin [36].

2.6. Ustekinumab

Several factors have been identified to accelerate the ustekinumab clearance in patients with IBD including increased body weight, lower albumin and immunogenicity (Figure 1) [41,42,43,44]. Other factors that can lead to a high clearance include prior exposure to biologics [42,43], increasing fat-free mass [43], male gender [42,44], Asian race [42] and higher CRP levels [42].
Data from four phase IIb/III ustekinumab clinical trials (C0743T26, CNTO1275CRD3001, CNTO1275CRD3002, CNTO1275CRD3003) demonstrated that increased body weight, elevated CRP, decreased albumin, TNF antagonist failure status (11% higher in failed patients), immunogenicity (increased by 13% in positive ADA status), sex (17% higher in males) and race (14% higher in Asian compared to non-Asian races) were associated with a higher clearance in CD [42]. In a PK analysis of UNIFI (NCT02407236), clearance increased non-linearly with body weight in patients with UC [44]. Moreover, a higher ustekinumab clearance was associated with lower albumin, male sex and immunogenicity [44].

2.7. Risankizumab

A PK analysis from a phase I study in healthy participants (NCT05305222) and phase II and III studies in CD (NCT02031276, ADVANCE, MOTIVATE and FORTIFY) identified increased body weight, decreased albumin, increased fecal calprotectin, corticosteroid use, increased creatinine clearance and male gender as variables associated with higher risakinumab clearance (Figure 1) [45]. Neutralizing antibodies and ADAs were not identified as significant covariates for risankizumab clearance [45].

2.8. Mirikizumab

A PK analysis of three RCTs (NCT02589665, NCT03518086, NCT03524092), including 1362 patients with UC, identified increased body weight and decreased albumin as factors associated with higher mirikizumab clearance (Figure 1) [46].

3. Association of Clearance with Therapeutic Outcomes

There is cumulating evidence suggesting that a higher clearance is associated with sub-therapeutic drug concentrations and unwanted therapeutic outcomes in patients with IBD, while a lower drug clearance is associated with clinical, biomarker, endoscopic and deep remission (Table 1).
Regarding infliximab, a PK analysis of the ACT 1 and two RCTs found a linear relationship between the baseline infliximab clearance and week 8 Mayo endoscopic scores (MES) (p < 0.001). Based on a receiver operating characteristic (ROC) curve analysis, a threshold of <0.397 L/d was associated with week 8 MES of ≤ 1 with a sensitivity (SN), specificity (SP), positive predictive value and area under the curve (AUC) of 75%, 48%, 68% and 0.64, respectively [48]. A prospective study of 31 children with IBD showed that patients who achieved deep remission at week 24 had a lower infliximab clearance at week 6 (0.202 L/d vs. 0.269 L/d, p = 0.020) and week 12 (0.215 L/d vs. 0.243 L/day, p = 0.022) compared to patients not achieving deep remission [52]. In a retrospective study of patients with acute severe UC, the median baseline calculated clearance was higher in patients requiring a colectomy at 6 months than in patients without a colectomy (0.733 vs. 0.569 L/d; p = 0.005). An infliximab clearance threshold of 0.627 L/d identified patients who required a colectomy with 80% SN and 82.8% SP (AUC: 0.80) [47]. A multivariable analysis identified the baseline infliximab clearance as the only factor associated with colectomy [47]. A retrospective study of 36 patients with corticosteroid-refractory acute UC showed that the infliximab clearance increased over time in those requiring a colectomy [14]. A prospective study by Peticollin et al. aiming to explore the link between PK parameters and the probability of relapse after de-escalation of infliximab therapy in patients with IBD showed that a drug clearance higher than 0.320 L/d at the time of infliximab de-escalation was associated with a shorter time to relapse [17].
There are only limited data regarding the association of adalimumab clearance with clinical outcomes in patients with IBD (Table 1). A recent retrospective cohort study including patients with CD showed that the median clearance was lower in patients achieving endoscopic remission as compared to those with persistent active endoscopic disease (0.247 L/d vs. 0.326 L/d, p < 0.01) [50]. Of note, there was no significant difference in the median adalimumab concentration between patients with endoscopic remission compared to those without (9.3 μg/mL vs. 11.7 μg/mL), implying that drug clearance may be a more superior PK measure than drug concentration to predict outcomes and a better reflection of inflammatory burden than drug concentration. While highly correlated with one another, clearance performed better than drug concentration alone with respect to all investigated outcomes based on the higher AUC in the ROC curve analysis [50].
Regarding the association of vedolizumab clearance with clinical outcomes in patients with IBD, the ERELATE study showed that baseline vedolizumab clearance thresholds of <0.17 L/d and <0.16 L/d were associated with clinical and deep remission at week 52, respectively [51]. The same study showed that clearance in the lower quartiles was associated with higher rates of favorable therapeutic outcomes, including clinical and deep remission assessed either at week 14 or week 52 [51]. In a propensity-score-based case-matching analysis using data from the GEMINI 1, the RCT clinical response and remission rates at week 14 were 26.6% and 5.9%, respectively, in the highest vedolizumab clearance quartile (0.23 to <0.55 L/d) compared to 65.5% and 35.7, respectively, in the lowest vedolizumab clearance concentration quartile (0.03 to <0.14 L/d) at week 6 [36]. In a prospective multicenter study in children with IBD, starting with a vedolizumab baseline clearance of less than 0.161 L/d predicted a fecal calprotectin remission (<250 µg/g) at the end of the induction [40].
Currently, there are no data available regarding the association of ustekinumab, mirikzumab or risankizumab clearance with therapeutic outcomes in patients with IBD.

4. Clearance Monitoring and Model Informed Precision Dosing

The optimal dose of a biologic is not the same for every patient with IBD due to the high interindividual variability in the monoclonal antibodies’ PK, and one size does not fit all [3]. Clinical decisions based only on TDM are rather empirical as they are based on analog flowcharts or decision trees that refer more to a trial-and-error treatment optimization, underestimating the true value of TDM [53]. One of the most important aspects of PK is clearance. Clearance precedes changes in drug concentrations and can be an early predictor of disease relapse or development of immunogenicity. A recent study showed that a combination of infliximab concentration and clearance was a better predictor of CRP-based clinical remission compared to either one alone [54]. Another study showed that clearance may be even better than drug concentrations for predicting favorable therapeutic outcomes in patients with CD treated with adalimumab [50].
Baseline clearance, although imprecise as it is estimated only based on patients’ clinical and demographic data, can be used to identify patients at high risk of underexposure requiring early proactive TDM and an intensified induction regimen [55]. Drug clearance during biologic therapy is more accurately estimated as drug concentrations are taken into account and can be used by an MIPD tool towards a more personalized implementation of TDM, allowing patient-specific dosing forecasts to accurately achieve a predefined drug concentration target [55]. Of note, clearance monitoring and MIPD do not require TDM at a trough but can operate with intermediate drug concentrations. This allows for more flexibility in the sampling and an extended window of opportunity to adjust dosing. This is particularly important for reactive TDM, as patients are symptomatic and cannot wait until the next drug administration for a clinical decision to be made. A recent study showed an excellent correlation of forecasted infliximab trough concentrations from mid-cycle blood samples with measured trough specimens [56].
MIPD typically uses Bayesian modeling to estimate clearance based on population PK modeling and patient data and a software tool to predict the optimal dosing for achieving a target drug concentration. Preliminary data from retrospective and prospective studies both in adult and pediatric patients with IBD treated with infliximab support the concept of MIPD-based proactive TDM for maintaining therapeutic drug concentrations, showing the benefits of reduced immunogenicity, higher response rates, drug durability and fewer complications [52,57,58,59,60,61,62,63]. Most importantly, the PRECISION RCT (NCT02453776) demonstrated that a PK dashboard-based proactive TDM of infliximab was superior to standard dosing for sustaining remission during maintenance therapy [64].
The real-world impact of infliximab precision-guided dosing on management of patients with IBD was demonstrated by a recent study of 275 patients and 37 providers, where in 58% of cases, providers modified the treatment plans based on the results of the MIDP, including dose modifications (41%) and drug discontinuation (8%). Moreover, all providers reported that MIPD was beneficial in guiding treatment decisions and added more value to their practice than routine TDM [49]. A physiologically based pharmacokinetic model was recently used to predict the PK of anti-TNF agents in pregnant women, fetuses and infants to inform dosing decisions for infliximab, adalimumab and golimumab in pregnancy and vaccination regimens for infants [65]. However, wide utilization of MIPD in clinical practice is hindered by its limited availability, high cost, undetermined optimal TDM sampling based also on the assay used, the lack of clearly defined targets for drug concentrations among different IBD phenotypes and the complexity of bedside implementation. Preliminary data suggest that an MIPD tool can be embedded within the electronic health record, guiding clinical decisions in real time for pediatric patients with CD treated with infliximab or adalimumab [10,66].

5. Conclusions

Cumulative data suggest that clearance monitoring of biologics can predict therapeutic outcomes in IBD. Preliminary data also demonstrate the importance of clearance when estimated by MIPD tools for providing dosing recommendations towards treatment optimization. However, more prospective studies are needed to establish the role of MIPD of biologics in IBD and to investigate the efficacy of a novel therapeutic strategy that includes the combination of MIPD-based proactive TDM and pharmacodynamics monitoring. The ongoing RCTs TITRATE (NCT03937609), MODIFI (NCT04982172), REMODEL-CD (NCT05660746) and OPTIMIZE (NCT04835506) will shed more light on the role of MIPD of infliximab in IBD. Future perspectives regarding the use of MIPD include the incorporation of additional factors such as visceral adipose tissue, human leukocyte antigen haplotypes or drug concentration at the site of inflammation that could increase the accuracy of the estimated clearance and the dosing predictions.

Author Contributions

T.D. and K.P.: manuscript drafting, reviewing and editing. A.S.C.: manuscript reviewing 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

Not applicable.

Conflicts of Interest

K.P. received lecture/speaker fees from Physicians Education Resource LLC and Grifols; scientific advisory board fees from ProciseDx Inc and Scipher Medicine Corporation; and serves as a consultant for Prometheus Laboratories Inc. A.S.C. served as a consultant and/or advisory board member for Janssen, Abbvie, Protagonist, Spherix, Artizan, Food is Good, Clario, Pfizer, Fresenius Kabi, Artugen, ProciseDx, Prometheus, Equillium, Samsung, Arena, Bacainn, Bristol Myers Squibb, Takeda; unbranded speaker for BMS and Abbvie. T.D declares no conflicts of interest.

References

  1. Papamichael, K.; Gils, A.; Rutgeerts, P.; Levesque, B.G.; Vermeire, S.; Sandborn, W.J.; Casteele, V. Role for therapeutic drug monitoring during induction therapy with TNF antagonists in IBD: Evolution in the definition and management of primary nonresponse. Inflamm. Bowel Dis. 2015, 21, 182–197. [Google Scholar] [CrossRef]
  2. Miligkos, M.; Papamichael, K.; Vande Casteele, N.; Mantzaris, G.M.; Gils, A.; Levesque, B.G.; Zintzaras, E. Efficacy and safety profile of anti-tumor necrosis factor-α versus anti-integrin agents for the treatment of Crohn’s disease: A network meta-analysis of indirect comparisons. Clin. Ther. 2016, 38, 1342–1358. [Google Scholar] [CrossRef]
  3. Kantasiripitak, W.; Wang, Z.; Spriet, I.; Ferrante, M.; Dreesen, E. Recent advances in clearance monitoring of monoclonal antibodies in patients with inflammatory bowel diseases. Exp. Rev. Clin. Pharmacol. 2021, 14, 1455–1466. [Google Scholar] [CrossRef]
  4. Edlund, H.; Steenholdt, C.; Ainsworth, M.A.; Goebgen, E.; Brynskov, J.; Thomsen, O.; WHuisinga, W.; Kloft, C. Magnitude of increased infliximab clearance imposed by anti-infliximab antibodies in Crohn’s disease is determined by their concentration. AAPS J. 2017, 19, 223–233. [Google Scholar] [CrossRef]
  5. Fasanmade, A.A.; Adedokun, O.J.; Ford, J.; Hernandez, D.; Johanns, J.; Hu, C.; Davis, H.M.; Zhou, H. Population pharmacokinetic analysis of infliximab in patients with ulcerative colitis. Eur. J. Clin. Pharmacol. 2009, 65, 1211–1228. [Google Scholar] [CrossRef]
  6. Fasanmade, A.A.; Adedokun, O.J.; Blank, M.; Zhou, H.; Davis, H.M. Pharmacokinetic Properties of infliximab in children and adults with Crohn’s disease: A retrospective analysis of data from 2 phase III clinical trials. Clin. Ther. 2011, 33, 946–964. [Google Scholar] [CrossRef]
  7. Fasanmade, A.A.; Adedokun, O.J.; Olson, A.; Strauss, R.; Davis, H.M. Serum albumin concentration: A predictive factor of infliximab pharmacokinetics and clinical response in patients with ulcerative colitis. Int. J. Clin. Pharmacol. Ther. 2010, 48, 297–308. [Google Scholar] [CrossRef]
  8. Colman, R.J.; Xiong, Y.; Mizuno, T.; Hyams, J.S.; Noe, J.D.; Boyle, B.; D’Haens, G.R.; van Limbergen, J.; Chun, K.; Yang, J.; et al. Antibodies-to-infliximab accelerate clearance while dose intensification reverses immunogenicity and recaptures clinical response in paediatric Crohn’s disease. Aliment. Pharmacol. Ther. 2022, 55, 593–603. [Google Scholar] [CrossRef]
  9. Grisic, A.M.; Eser, A.; Huisinga, W.; Reinisch, W.; Kloft, C. Quantitative relationship between infliximab exposure and inhibition of C-reactive protein synthesis to support inflammatory bowel disease management. Br. J. Clin. Pharmacol. 2021, 87, 2374–2384. [Google Scholar] [CrossRef]
  10. Xiong, Y.; Mizuno, T.; Colman, R.; Hyams, J.; Noe, J.D.; Boyle, B.; Tsai, Y.T.; Dong, M.; Jackson, K.; Punt, N.; et al. Real-World infliximab pharmacokinetic study informs an electronic health record-embedded dashboard to guide precision dosing in children with Crohn’s disease. Clin. Pharmacol. Ther. 2021, 109, 1639–1647. [Google Scholar] [CrossRef]
  11. Dreesen, E.; Berends, S.; Laharie, D.; D’Haens, G.; Vermeire, S.; Gils, A.; Mathôt, R. Modelling of the relationship between infliximab exposure, faecal calprotectin and endoscopic remission in patients with Crohn’s disease. Br. J. Clin. Pharmacol. 2021, 87, 106–118. [Google Scholar] [CrossRef]
  12. Adedokun, O.J.; Xu, Z.; Padgett, L.; Blank, M.; Johanns, J.; Griffiths, A.; Ford, J.; Zhou, H.; Guzzo, C.; Davis, H.M.; et al. Pharmacokinetics of infliximab in children with moderate-to-severe ulcerative colitis: Results from a randomized, multicenter, open-label, phase 3 study. Inflamm. Bowel Dis. 2013, 19, 2753–2762. [Google Scholar] [CrossRef]
  13. Hanzel, J.; Bukkems, L.H.; Gecse, K.B.; D’Haens, G.R.; Mathôt, R.A. Population pharmacokinetics of subcutaneous infliximab CT-P13 in Crohn’s disease and ulcerative colitis. Aliment. Pharmacol. Ther. 2021, 54, 1309–1319. [Google Scholar] [CrossRef]
  14. Kevans, D.; Murthy, S.; Mould, D.R.; Silverberg, M.S. Accelerated clearance of infliximab is associated with treatment failure in patients with corticosteroid-refractory acute ulcerative colitis. J. Crohn’s Colitis 2018, 12, 662–669. [Google Scholar] [CrossRef]
  15. Matsuoka, K.; Hamada, S.; Shimizu, M.; Nanki, K.; Mizuno, S.; Kiyohara, H.; Arai, M.; Sugimoto, S.; Iwao, Y.; Ogata, H.; et al. Factors contributing to the systemic clearance of infliximab with long-term administration in Japanese patients with Crohn’s disease: Analysis using population pharmacokinetics. Int. J. Clin. Pharmacol. Ther. 2020, 58, 89. [Google Scholar] [CrossRef]
  16. Petitcollin, A.; Leuret, O.; Tron, C.; Lemaitre, F.; Verdier, M.C.; Paintaud, G.; Bouguen, G.; Willot, S.; Bellissant, E.; Ternant, D. Modeling immunization to infliximab in children with Crohn’s disease using population pharmacokinetics: A pilot study. Inflamm. Bowel Dis. 2018, 24, 1745–1754. [Google Scholar] [CrossRef]
  17. Petitcollin, A.; Brochard, C.; Siproudhis, L. Pharmacokinetic parameters of infliximab influence the rate of relapse after de-escalation in adults with inflammatory bowel diseases. Clin. Pharmacol. Ther. 2019, 106, 605–615. [Google Scholar] [CrossRef]
  18. Buurman, D.J.; Maurer, J.M.; Keizer, R.J.; Kosterink, J.G.W.; Dijkstra, G. Population pharmacokinetics of infliximab in patients with inflammatory bowel disease: Potential implications for dosing in clinical practice. Aliment. Pharmacol. Ther. 2015, 42, 529–539. [Google Scholar] [CrossRef]
  19. Brandse, J.F.; Mould, D.; Smeekes, O.; Ashruf, Y.; Kuin, S.; Strik, A.; van den Brink, G.R.; D’Haens, G.R. A real-life population pharmacokinetic study reveals factors associated with clearance and immunogenicity of infliximab in inflammatory bowel disease. Inflamm. Bowel Dis. 2017, 23, 650–660. [Google Scholar] [CrossRef]
  20. Schreiber, S.; Ben-Horin, S.; Leszczyszyn, J.; Dudkowiak, R.; Lahat, A.; Gawdis-Wojnarska, B.; Pukitis, A.; Horynski, M.; Farkas, K.; Kierkus, J.; et al. Randomized controlled trial: Subcutaneous vs intravenous infliximab CT-P13 maintenance in inflammatory bowel disease. Gastroenterology 2021, 160, 2340–2353. [Google Scholar] [CrossRef]
  21. Whaley, K.G.; Xiong, Y.; Karns, R.; Hyams, J.S.; Kugathasan, S.; Boyle, B.M.; Walters, T.D.; Kelsen, J.; LeLeiko, N.; Shapiro, J.; et al. Multicenter cohort study of infliximab pharmacokinetics and therapy response in pediatric acute severe ulcerative colitis. Clin. Gastroenterol. Hepatol. 2023, 21, 1338–1347. [Google Scholar] [CrossRef] [PubMed]
  22. Vermeire, S.; D’Haens, G.; Laharie, D.; Dreesen, E.; Rabizadeh, S.; Jain, A.; Spencer, E.A.; Panetta, J.C.; Dubinsky, M.; Dervieux, T. Infliximab clearance pre-therapy and during induction predicts long term disease control in inflammatory bowel disease. Gastroenterology 2022, 162, S45–S46. [Google Scholar] [CrossRef]
  23. Le Tilly, O.; Bejan-Angoulvant, T.; Paintaud, G.; Ternant, D. Letter to Dreesen et al. on their article “Modelling of the Relationship Between Infliximab Exposure, Faecal Calprotectin, and Endoscopic Remission in Patients with Crohn’s Disease”—A comprehensive review of infliximab population pharmacokinetic modelling publications. Br. J. Clin. Pharmacol. 2021, 87, 1594–1595. [Google Scholar] [CrossRef] [PubMed]
  24. Chung, A.; Carroll, M.; Almeida, P.; Petrova, A.; Isaac, D.; Mould, D.; Wine, E.; Huynh, H. Early infliximab clearance predicts remission in children with Crohn’s disease. Dig. Dis. Sci. 2023, 68, 1995–2005. [Google Scholar] [CrossRef]
  25. Bauman, L.E.; Xiong, Y.; Mizuno, T.; Minar, P.; Fukuda, T.; Dong, M.; Rosen, M.J.; Vinks, A.A. Improved population pharmacokinetic model for predicting optimized infliximab exposure in pediatric inflammatory bowel disease. Inflamm. Bowel Dis. 2020, 26, 429–439. [Google Scholar] [CrossRef] [PubMed]
  26. Vande Casteele, N.; 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]
  27. Berends, S.E.; Strik, A.S.; Van Selm, J.C.; Löwenberg, M.; Ponsioen, C.Y.; D’Haens, G.R.; Mathôt, R.A. Explaining interpatient variability in adalimumab pharmacokinetics in patients with Crohn’s disease. Ther. Drug Monit. 2018, 40, 202–211. [Google Scholar] [CrossRef]
  28. Ternant, D.; Karmiris, K.; Vermeire, S.; Desvignes, C.; Azzopardi, N.; Bejan-Angoulvant, T.; Van Assche, G.; Paintaud, G. Pharmacokinetics of adalimumab in Crohn’s disease. Eur. J. Clin. Pharmacol. 2015, 71, 1155–1157. [Google Scholar] [CrossRef]
  29. Ponce-Bobadilla, A.V.; Stodtmann, S.; Chen, M.J.; Winzenborg, I.; Mensing, S.; Blaes, J.; Haslberger, T.; Laplanche, L.; Dreher, I.; Mostafa, N.M. Assessing the impact of immunogenicity and improving prediction of trough concentrations: Population pharmacokinetic modeling of adalimumab in patients with Crohn’s disease and ulcerative colitis. Clin. Pharmacokinet. 2023, 62, 623–634. [Google Scholar] [CrossRef]
  30. Sharma, S.; Eckert, D.; Hyams, J.S.; Mensing, S.; Thakkar, R.B.; Robinson, A.M.; Rosh, J.R.; Ruemmele, F.M.; Awni, W.M. Pharmacokinetics and exposure–efficacy relationship of adalimumab in pediatric patients with moderate to severe Crohn’s disease: Results from a randomized, multicenter, phase-3 study. Inflamm. Bowel Dis. 2015, 21, 783–792. [Google Scholar] [CrossRef]
  31. Vande Casteele, N.; Mould, D.R.; Coarse, J.; Hasan, I.; Gils, A.; Feagan, B.; Sandborn, W.J. Accounting for pharmacokinetic variability of certolizumab pegol in patients with Crohn’s disease. Clin. Pharmacokinet. 2017, 56, 1513–1523. [Google Scholar] [CrossRef] [PubMed]
  32. Lefevre, P.L.C.; Dulai, P.S.; Wang, Z.; Guizzetti, L.; Feagan, B.G.; Pop, A.; Yassine, M.; Shackelton, L.M.; Jairath, V.; Sandborn, W.J.; et al. A clinical prediction model to determine probability of response to certolizumab pegol for Crohn’s disease. BioDrugs. 2022, 36, 85–93. [Google Scholar] [CrossRef]
  33. Dreesen, E.; Kantasiripitak, W.; Detrez, I.; Stefanović, S.; Vermeire, S.; Ferrante, M.; Bouillon, T.; Drobne, D.; Gils, A. A Population pharmacokinetic and exposure–response model of golimumab for targeting endoscopic remission in patients with ulcerative colitis. Inflamm. Bowel Dis. 2020, 26, 570–580. [Google Scholar] [CrossRef]
  34. Xu, Y.; Adedokun, O.J.; Chan, D.; Hu, C.; Xu, Z.; Strauss, R.S.; Hyams, J.S.; Turner, D.; Zhou, H. Population pharmacokinetics and exposure-response modeling analyses of golimumab in children with moderately to severely active ulcerative colitis. J. Clin. Pharmacol. 2019, 59, 590–604. [Google Scholar] [CrossRef]
  35. Rosario, M.; Dirks, N.L.; Gastonguay, M.R.; Fasanmade, A.A.; Wyant, T.; Parikh, A.; Sandborn, W.J.; Feagan, B.G.; Reinisch, W.; Fox, I. Population pharmacokinetics-pharmacodynamics of vedolizumab in patients with ulcerative colitis and Crohn’s disease. Aliment. Pharmacol. Ther. 2015, 42, 188–202. [Google Scholar] [CrossRef] [PubMed]
  36. Osterman, M.T.; Rosario, M.; Lasch, K.; Barocas, M.; Wilbur, J.D.; Dirks, N.L.; Gastonguay, M.R. Vedolizumab exposure levels and clinical outcomes in ulcerative colitis: Determining the potential for dose optimisation. Aliment. Pharmacol. Ther. 2019, 49, 408–418. [Google Scholar] [CrossRef] [PubMed]
  37. Okamoto, H.; Dirks, N.L.; Rosario, M.; Hori, T.; Hibi, T. Population pharmacokinetics of vedolizumab in Asian and non-Asian patients with ulcerative colitis and Crohn’s disease. Intest. Res. 2021, 19, 95–105. [Google Scholar] [CrossRef]
  38. Hanzel, J.; Dreesen, E.; Vermeire, S.; Löwenberg, M.; Hoentjen, F.; Bossuyt, P.; Clasquin, E.; Baert, F.J.; D’Haens, G.R.; Mathôt, R. Pharmacokinetic-pharmacodynamic model of vedolizumab for targeting endoscopic remission in patients with Crohn disease: Post-hoc analysis of the LOVE-CD study. Inflamm. Bowel Dis. 2022, 28, 689–699. [Google Scholar] [CrossRef]
  39. Colman, R.J.; Mizuno, T.; Fukushima, K.; Haslam, D.B.; Hyams, J.S.; Boyle, B.; Noe, J.D.; D’Haens, G.R.; Van Limbergen, J.; Chun, K.; et al. Real world population pharmacokinetic study in children and young adults with inflammatory bowel disease discovers novel blood and stool microbial predictors of vedolizumab clearance. Aliment. Pharmacol. Ther. 2023, 57, 524–539. [Google Scholar] [CrossRef]
  40. Colman, R.; Mizuno, T.; Hyams, J.; Noe, J.; Boyle, B.; Denson, L.; Vinks, A.; Minar, P. Real-world vedolizumab pharmacokinetic study in children identifies two novel biomarkers of drug clearance. Gastroenterology 2022, 162, S100. [Google Scholar] [CrossRef]
  41. Wang, Z.; Verstockt, B.; Sabino, J.; Vermeire, S.; Ferrante, M.; Declerck, P.; Dreesen, E. Population pharmacokinetic-pharmacodynamic model-based exploration of alternative ustekinumab dosage regimens for patients with Crohn’s disease. Br. J. Clin. Pharmacol. 2022, 88, 323–335. [Google Scholar] [CrossRef] [PubMed]
  42. Adedokun, O.J.; Xu, Z.; Gasink, C.; Kowalski, K.; Sandborn, W.J.; Feagan, B. Population pharmacokinetics and exposure–response analyses of ustekinumab in patients with moderately to severely active Crohn’s disease. Clin. Ther. 2022, 44, 1336–1355. [Google Scholar] [CrossRef]
  43. Aguiar Zdovc, J.; Hanžel, J.; Kurent, T.; Sever, N.; Koželj, M.; Smrekar, N.; Novak, G.; Štabuc, B.; Dreesen, E.; Thomas, D.; et al. Ustekinumab dosing individualization in crohn’s disease guided by a population pharmacokinetic–pharmacodynamic model. Pharmaceutics 2021, 13, 1587. [Google Scholar] [CrossRef] [PubMed]
  44. Xu, Y.; Hu, C.; Chen, Y.; Miao, X.; Adedokun, O.J.; Xu, Z.; Sharma, A.; Zhou, H. Population pharmacokinetics and exposure-response modeling analyses of ustekinumab in adults with moderately to severely active ulcerative colitis. J. Clin. Pharmacol. 2020, 60, 889–902. [Google Scholar] [CrossRef] [PubMed]
  45. Suleiman, A.A.; Goebel, A.; Bhatnagar, S.; D’Cunha, R.; Liu, W.; Pang, Y. Population pharmacokinetic and exposure–response analyses for efficacy and safety of risankizumab in patients with active Crohn’s disease. Clin. Pharmacol. Ther. 2023, 113, 839–850. [Google Scholar] [CrossRef]
  46. Chua, L.; Friedrich, S.; Zhang, X.C. Mirikizumab pharmacokinetics in patients with moderately to severely active ulcerative colitis: Results from phase III LUCENT Studies. Clin. Pharmacokinet. 2023, 62, 1479–1491. [Google Scholar] [CrossRef]
  47. Battat, R.; Hemperly, A.; Truong, S.; Whitmire, N.; Boland, B.S.; Dulai, P.S.; Holmer, A.K.; Nguyen, N.H.; Singh, S.; Vande Casteele, N.; et al. Baseline clearance of infliximab is associated with requirement for colectomy in patients with acute severe ulcerative colitis. Clin. Gastroenterol. Hepatol. 2021, 19, 511–518. [Google Scholar] [CrossRef]
  48. Vande Casteele, N.; 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. [Google Scholar] [CrossRef]
  49. Abraham, B.P.; Ziring, D.A.; Dervieux, T.; Han, P.A.; Shim, A.; Battat, R. Real-world impact of infliximab precision-guided dosing on management of patients with IBD. Am. J. Manag. Care 2023, 29, S227–S235. [Google Scholar] [CrossRef]
  50. Wright, E.K.; Chaparro, M.; Gionchetti, P.; Hamilton, A.L.; Schulberg, J.; Gisbert, J.P.; Valerii, M.C.; Rizzello, F.; De Cruz, P.; Panetta, J.C.; et al. Adalimumab clearance, rather than trough level, may have greatest relevance to crohn’s disease therapeutic outcomes assessed clinically and endoscopically. J. Crohn’s Colitis, 2023; jjad140, Online ahead of print. [Google Scholar] [CrossRef]
  51. Vande Casteele, N.; Sandborn, W.J.; Feagan, B.G. Real-world multicentre observational study including population pharmacokinetic modelling to evaluate the exposure–response relationship of vedolizumab in inflammatory bowel disease: ERELATE Study. Aliment. Pharmacol. Ther. 2022, 56, 463–476. [Google Scholar] [CrossRef]
  52. Kantasiripitak, W.; Wicha, S.G.; Thomas, D.; Hoffman, I.; Ferrante, M.; Vermeire, S.; Karen van Hoeve, K.; Dreesen, E. A model-based tool for guiding infliximab induction dosing to maximise long-term deep remission in children with inflammatory bowel diseases. J. Crohn’s Colitis 2023, jjad009. [Google Scholar] [CrossRef]
  53. Wang, Z.; Dreesen, E. Therapeutic drug monitoring of anti-tumor necrosis factor agents: Lessons learned and remaining issues. Curr. Opin. Pharmacol. 2020, 55, 53–59. [Google Scholar] [CrossRef] [PubMed]
  54. Dubinsky, M.C.; Rabizadeh, S.; Panetta, J.C.; Spencer, E.A.; Everts-van der Wind, A.; Dervieux, T. The combination of predictive factors of pharmacokinetic origin associates with enhanced disease control during treatment of pediatric Crohn’s disease with infliximab. Pharmaceutics 2023, 15, 2408. [Google Scholar] [CrossRef] [PubMed]
  55. Kantasiripitak, W.; Outtier, A.; Wicha, S.G.; Kensert, K.; Wang, Z.; Sabino, J.; Vermeire, S.; Thomas, D.; Ferrante, M.; Dreesen, E. Multi-model averaging improves the performance of model-guided infliximab dosing in patients with inflammatory bowel diseases. CPT Pharmacomet. Syst. Pharmacol. 2022, 11, 1045–1059. [Google Scholar] [CrossRef]
  56. Primas, C.; Reinisch, W.; Panetta, J.C.; Eser, A.; Mould, D.R.; Dervieux, T. Model informed precision dosing tool forecasts trough infliximab and associates with disease status and tumor necrosis factor-alpha levels of inflammatory bowel diseases. J. Clin. Med. 2022, 11, 3316. [Google Scholar] [CrossRef]
  57. Dubinsky, M.C.; Phan, B.L.; Singh, N.; Rabizadeh, S.; Mould, D.R. Pharmacokinetic dashboard-recommended dosing is different than standard of care dosing in infliximab-treated pediatric IBD patients. AAPS J. 2017, 19, 215–222. [Google Scholar] [CrossRef] [PubMed]
  58. Eser, A.; Primas, C.; Reinisch, S.; Vogelsang, H.; Novacek, G.; Mould, D.R.; Reinisch, W. Prediction of individual serum infliximab concentrations in inflammatory bowel disease by a Bayesian dashboard system. J. Clin. Pharmacol. 2018, 58, 790–802. [Google Scholar] [CrossRef]
  59. 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]
  60. Santacana Juncosa, E.; Rodríguez-Alonso, L.; Padullés Zamora, A.; Guardiola, J.; Rodríguez-Moranta, F.; Nilsson, K.S.; Minguet, J.B.; Rego, F.M.; Codina, H.C.; Zamora, N.P. Bayes-based dosing of infliximab in inflammatory bowel diseases: Short-term efficacy. Br. J. Clin. Pharmacol. 2021, 87, 494–505. [Google Scholar] [CrossRef]
  61. Papamichael, K.; Cheifetz, A.S. Optimizing therapeutic drug monitoring in inflammatory bowel disease: A focus on therapeutic monoclonal antibodies. Expert Opin. Drug Metab. Toxicol. 2021, 17, 1423–1431. [Google Scholar] [CrossRef]
  62. Serrano-Díaz, L.; Iniesta-Navalón, C.; Gómez-Espín, R.; Nicolás-de Prado, I.; Bernal-Morell, E.; Rentero-Redondo, L. Impact of proactive therapeutic drug monitoring of infliximab during the induction phase in IBD patients. A Bayesian Approach. Rev. Esp. Enferm. Dig. 2023, 115, 435–443. [Google Scholar] [CrossRef] [PubMed]
  63. Díaz, L.S.; Navalón, C.I.; Espín, R.G.; Nicolás-de Prado, I.; Bernal-Morell, E.; Rentero-Redondo, L. Impact of proactive of infliximab monitoring using the Bayesian approach in the maintenance phase in patients with inflammatory bowel disease. Gastroenterol. Hepatol. 2023, 46, 504–511. [Google Scholar] [CrossRef] [PubMed]
  64. 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] [PubMed]
  65. Chen, J.; Lin, R.; Guo, G.; Wu, W.; Ke, M.; Ke, C.; Huang, P.; Lin, C. Physiologically-based pharmacokinetic modeling of anti-tumor necrosis factor agents for inflammatory bowel disease patients to predict the withdrawal time in pregnancy and vaccine time in infants. Clin. Pharmacol. Ther. 2023; Online ahead of print. [Google Scholar] [CrossRef]
  66. Colman, R.J.; Samuels, A.; Mizuno, T.; Punt, N.; Vinks, A.A.; Minar, P. Model-informed precision dosing for biologics is now available at the bedside for patients with inflammatory bowel disease. Inflamm. Bowel Dis. 2023, 29, 1342–1346. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Variables associated with higher clearance of biologics in patients with inflammatory bowel disease. CD: Crohn’s disease; IBD: inflammatory bowel disease; UC: ulcerative colitis; ADA; anti-drug antibodies; CRP: C-reactive protein, AZA: azathioprine, MTX: methotrexate, FC: fecal calprotectin, CDAI: Crohn’s disease activity index, PMS: partial Mayo score, WBC: white blood count, ESR: erythrocyte sedimentation rate, EOW: every other week, PCDAI: pediatric CDAI, ALP: alkaline phosphatase; nCD64: neutrophil CD64; ↑ higher; ↓ lower.
Figure 1. Variables associated with higher clearance of biologics in patients with inflammatory bowel disease. CD: Crohn’s disease; IBD: inflammatory bowel disease; UC: ulcerative colitis; ADA; anti-drug antibodies; CRP: C-reactive protein, AZA: azathioprine, MTX: methotrexate, FC: fecal calprotectin, CDAI: Crohn’s disease activity index, PMS: partial Mayo score, WBC: white blood count, ESR: erythrocyte sedimentation rate, EOW: every other week, PCDAI: pediatric CDAI, ALP: alkaline phosphatase; nCD64: neutrophil CD64; ↑ higher; ↓ lower.
Jcm 12 07132 g001
Table 1. Estimated clearance of biologics associated with therapeutic outcomes in patients with inflammatory bowel disease.
Table 1. Estimated clearance of biologics associated with therapeutic outcomes in patients with inflammatory bowel disease.
Author (Year)Study DesignDrugPopulation (N)Estimated
Clearance
Time Point
Estimated Clearance Threshold, L/DayTherapeutic
Outcome
(Time Point)
Battat (2021) [47]RetrospectiveIFXASUC (N = 39)Baseline≥0.627Colectomy
(6 months)
Vande Casteele
(2019) [48]
ACT 1 and 2 RCTsIFXUC (N = 484)Baseline<0.397MES ≤ 1
(week 8)
Vande Casteele
(2019) [48]
ACT 1 and 2 RCTsIFXUC (N = 484)Baseline<0.364MES ≤ 1
(week 30)
Peticollin (2019) [17]ProspectiveIFXIBD (N = 91)At time of
de-escalation
>0.320Relapse following treatment de-escalation
Whaley (2023) [21]ProspectiveIFXASUC * (N = 38)Day 3 after drug initiation>0.480Colectomy
Chung (2023) [24]RetrospectiveIFXCD (N = 85)Baseline<0.230Remission
(5 months)
Chung (2023) [24]RetrospectiveIFXCD (N = 85)Baseline<0.238Remission
(10 months)
Chung (2023) [24]RetrospectiveIFXCD (N = 85)Baseline<0.243Remission
(16 months)
Chung (2023) [24]RetrospectiveIFXCD (N = 85)End of induction<0.230Remission
(5 months)
Chung (2023) [24]RetrospectiveIFXCD (N = 85)End of induction<0.213Remission
(10 months)
Chung (2023) [24]RetrospectiveIFXCD (N = 85)End of induction<0.252Remission
(16 months)
Vermeire (2022) [22]ProspectiveIFXIBD (N = 276)Baseline, weeks 2, 6, 14<0.250CRP-based clinical remission
Abraham (2023) [49]ProspectiveIFXIBD (N = 275)Maintenance>0.294Active disease,
drug discontinuation
Wright (2023) [50]RetrospectiveADMCD (N = 237)Maintenance #<0.350SES-CD < 3
Wright (2023) [50]RetrospectiveADMCD (N = 237)Maintenance #<0.280FC < 100 ug/g
Wright (2023) [50]RetrospectiveADMCD (N = 237)Maintenance #<0.300CRP-based clinical remission
Lefevre (2022) [32]PRECiSE 1 and 2 RCTsCZPCD (N = 964)Baseline>0.500Drug TC < 36 μg/mL (week 6)
Vande Casteele (2022) [51]RetrospectiveVDZIBD (N = 695)Baseline<0.170Clinical remission
(week 52)
Vande Casteele (2022) [51]RetrospectiveVDZIBD (N = 695)Baseline <0.160Deep remission
(week 52)
Osterman
(2019) [36]
GEMINI 1 RCTVDZUC (N = 693)Week 6<0.140Clinical response
(week 14)
Colman (2022) [40]ProspectiveVDZIBD (N = 21)Baseline<0.161FC < 250 mg/g
(week 14)
* Pediatric; # The sampling time for adalimumab pharmacokinetics were matched to the study visit assessment, occurring anytime relative to the last dose. IFX: infliximab; ADM: adalimumab; CZP: certolizumab pegol; VDZ: vedolizumab; ASUC: acute severe ulcerative colitis; IBD: inflammatory bowel disease; MES: Mayo endoscopic score; CD: Crohn’s disease; UC: ulcerative colitis; FC: fecal calprotectin; CRP: C-reactive protein; SES-CD: Simple Endoscopic Score for Crohn’s Disease; RCT: randomized controlled trial; TC: trough concentration.
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

Deyhim, T.; Cheifetz, A.S.; Papamichael, K. Drug Clearance in Patients with Inflammatory Bowel Disease Treated with Biologics. J. Clin. Med. 2023, 12, 7132. https://doi.org/10.3390/jcm12227132

AMA Style

Deyhim T, Cheifetz AS, Papamichael K. Drug Clearance in Patients with Inflammatory Bowel Disease Treated with Biologics. Journal of Clinical Medicine. 2023; 12(22):7132. https://doi.org/10.3390/jcm12227132

Chicago/Turabian Style

Deyhim, Tina, Adam S. Cheifetz, and Konstantinos Papamichael. 2023. "Drug Clearance in Patients with Inflammatory Bowel Disease Treated with Biologics" Journal of Clinical Medicine 12, no. 22: 7132. https://doi.org/10.3390/jcm12227132

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