Review Open Access
Copyright ©The Author(s) 2023. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Mar 14, 2023; 29(10): 1539-1550
Published online Mar 14, 2023. doi: 10.3748/wjg.v29.i10.1539
Precision medicine in inflammatory bowel disease: Individualizing the use of biologics and small molecule therapies
Eric Cheah, Department of Gastroenterology and Clinical Nutrition, The Royal Children's Hospital Melbourne, Parkville, VIC 3052, Australia
James Guoxian Huang, Department of Paediatrics, Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore 119228, Singapore
James Guoxian Huang, Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore
ORCID number: Eric Cheah (0000-0002-9444-1119); James Guoxian Huang (0000-0002-5869-4194).
Author contributions: Cheah E and Huang JG were involved in the conception of the study, data collection, drafting of the article, critical revision of the article, and final approval of the published version.
Conflict-of-interest statement: All authors have no conflicts of interest to declare.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: James Guoxian Huang, MBBS, Assistant Professor, Attending Doctor, Staff Physician, Department of Pediatrics, Khoo Teck Puat-National University Children's Medical Institute, National University Health System, NUHS Tower Block Level 12, 1E Kent Ridge Road, Singapore 119228, Singapore. james_huang@nuhs.edu.sg
Received: September 29, 2022
Peer-review started: September 29, 2022
First decision: January 3, 2023
Revised: January 17, 2023
Accepted: February 21, 2023
Article in press: February 21, 2023
Published online: March 14, 2023

Abstract

The advent of biologics and small molecules in inflammatory bowel disease (IBD) has marked a significant turning point in the prognosis of IBD, decreasing the rates of corticosteroid dependence, hospitalizations and improving overall quality of life. The introduction of biosimilars has also increased affordability and enhanced access to these otherwise costly targeted therapies. Biologics do not yet represent a complete panacea: A subset of patients do not respond to first-line anti-tumor necrosis factor (TNF)-alpha agents or may subsequently demonstrate a secondary loss of response. Patients who fail to respond to anti-TNF agents typically have a poorer response rate to second-line biologics. It is uncertain which patient would benefit from a different sequencing of biologics or even a combination of biologic agents. The introduction of newer classes of biologics and small molecules may provide alternative therapeutic targets for patients with refractory disease. This review examines the therapeutic ceiling in current treatment strategies of IBD and the potential paradigm shifts in the future.

Key Words: Precision medicine, Therapeutic ceiling, Inflammatory bowel disease, Biologics, Small molecules

Core Tip: Precision medicine and individualizing patient care has been the holy grail in the management of inflammatory bowel disease (IBD). A one-size-fits-all approach, utilizing the current armamentarium of biologics and small molecules, still yields less than ideal clinical outcomes, with significantly high non-response rates. Multiple challenges remain in breaking this therapeutic ceiling: Achieving an early diagnosis of IBD ideally even in the pre-clinical phase; accurately prognosticating the disease course; and tailoring an appropriately sequenced therapy regime to a patient’s disease severity, pharmacokinetic and pharmacodynamic profile.



INTRODUCTION

The incidence of inflammatory bowel disease (IBD) has seen a rise within Asia Pacific. Overall incidence and prevalence rates in Asia are lower than the West, but are on the rise[1,2]. Treatment of IBD has progressed rapidly over the past several decades. A new era in the treatment of IBD began with the development of the chimeric monoclonal anti-tumor necrosis factor (TNF)-alpha antibody cA2 in the early 1990’s. cA2 was subsequently renamed infliximab, and was first licensed by the United States Food and Drug Administration (FDA) in August 1998 for the treatment of Crohn’s disease (CD)[3]. Since the introduction of infliximab, there has been an advent of newer biologics and small molecule agents, along with paradigm shifts in the treatment goals of IBD. The agents currently FDA approved for use include the anti-integrin (vedolizumab), anti-interleukin (IL)-12/23 p40 (ustekinumab), anti-IL-23p19 (risankizumab), oral Janus kinase (JAK) inhibitors (tofacitinib, upadacitinib), and sphingosine-1-phosphate (S1P) receptor modulator (ozanimod). As of 2019, mesenchymal stem cell therapy (darvadstrocel) received a regenerative medicine advanced therapy designation for complex perianal fistulas in adult patients with CD. New targets and treatments being explored in Phase II/III trials include anti-integrin (etrolizumab, ontamalimab), IL-23p19 inhibitors (mirikizumab, brazikumab, guselkumab), oral JAK inhibitor (filgotinib), and S1P modulator (etrasimod)[4].

Current treatment goals in IBD aim to more than just achieve clinical remission. Deep remission, the combination of clinical remission and mucosal healing, represents an important therapeutic target that is now increasingly attainable with the timely use of biologics[5]. Expert consensus statements in the STRIDE[1]/STRIDE-II guidelines, with evidence from the CALM study, have helped us define treat-to-target strategies in adults and children utilizing clinical indices, biomarkers, and endoscopic parameters[6-8]. Aspirational targets include transmural healing in CD and histologic healing in ulcerative colitis (UC).

There is an increasing need to develop newer biologics and small molecules targeting novel cytokine pathways, as current therapeutic options are far from perfect in achieving the above-mentioned treatment targets. A meta-analysis of real-world deep remission rates with anti-TNF agents demonstrated that deep remission was only achieved in 48.6% of CD patients and 43.6% of UC patients at 1 year[9]. In a review by Papamichael et al[10], the rates of primary non-response and non-remission to anti-TNF agents in IBD were between 10%-40% and 50%-80%, respectively. A further 23%-46% of those initial responders or those who achieve remission have a secondary loss of response over time[11]. The clinical remission rates with second-line biologics are also poorer in patients, who have had a prior loss of response to anti-TNF agents, especially those who had a primary non-response[12-14]. Such data distinctly highlight the therapeutic ceiling in current IBD management: how can we optimize current therapies to go beyond this therapeutic ceiling?

BIOMARKERS IN IBD
Predictive and prognostic biomarkers, pharmacogenomics, and response to therapy

Precision and personalized medicine has long been a discussed topic in the management of IBD. It is an aspirational goal to accurately predict those with a complicated and aggressive clinical course, and to administer timely targeted therapy to the individual’s molecular inflammatory profile[15]. Particularly for CD, the emphasis is for appropriately early management within the window of opportunity, before permanent digestive damage is done[16,17]. However, management decisions are still currently made using a one-size-fits-all approach. The conventional strategy is the step-up approach, which will inevitably undertreat patients who are destined to run a more aggressive disease course. With easier access to biologics and small molecules, a top-down approach may be used, which has been shown to improve clinical outcomes in prognostically severe CD[18,19] but this may otherwise expose patients destined to have mild disease to unnecessary risks and overtreatment. This top-down strategy would also be unaffordable in financially constrained health-care settings particularly in the Asia-Pacific region, and may pose a challenge in health jurisdictions that limit the use of expensive novel therapies.

Furthermore, the heterogeneity and variability of the clinical course of IBD between different individuals would mean a suboptimal approach in a substantial group of patients. There is a general consensus that a tailored approach is required, with a need for accurate biomarkers that enable the right patient to be matched to the right treatment (Table 1).

Table 1 Selected list of biomarkers in inflammatory bowel disease.
Biomarker class
Biomarker
Clinical utility
Prognostic biomarkersAnti-ompC, ASCA, ANCA, anti-CBir1, flagellinPrediction of more severe CD phenotype- particularly stricturing and need for surgery
CD 8+ T cell clonal signaturePrediction of more severe disease course and relapse in CD and UC
Surveillance of disease activityFecal calprotectinPredictor of endoscopic disease activity as well as histologic inflammation, and relapse in asymptomatic patients with IBD
MMP-9Associated with disease activity in UC
IL-22Associated with disease activity in CD
Pharmacogenomics and prediction of safetyTPMTRisk of thiopurine adverse reaction
NUDT15Risk of thiopurine adverse reaction, more common in East Asian/Asian populations
Thiopurine metabolites (6TG, 6MMP)Levels associated with adverse drug reaction: myelosuppression, hepatotoxicity. 6TG range also associated with therapy response
Prediction of response to therapyOncostatin MHigher levels predictor of non-response to anti-TNF
TREM-1Low levels predictor of non-response to anti-TNF
HLA-DQA1*05Expression associated with risk of antibody formation to anti-TNF
IL-22Higher level associated with response to anti-IL23p19 (brazikumab)

Stratification of treatment approaches can help identify those of a more complicated course and hence tailoring treatment accordingly. Among prognostic biomarkers at diagnosis predicting a more complex CD phenotype and worse outcomes including stricturing phenotype and need for surgery, identified microbial predictors include circulating antibodies against bacterial antigens such as anti-outer membrane protein C, anti-Saccharomyces cerevisiae antibody, perinuclear anti-neutrophil cytoplasmic antibodies, and anti-CBir1 flagellin. However, it is unclear if these represent a cause or effect of severe disease[20-24].

A CD8+ T-cell clonal signature was identified to predict worse outcomes and relapse in IBD patients[25,26]. This genomic biomarker was subsequently validated in independent cohorts of newly diagnosed CD and UC patients in the United Kingdom[27]. There is now a trial in progress in the United Kingdom to assess this whole-blood biomarker to guide treatment for newly diagnosed CD patients[28]. It is currently available in clinical use: PredictSure IBD; PredictImmune, Cambridge, United Kingdom.

Other predictive tools include the use of fecal calprotectin as a predictor of endoscopic disease activity as well as histologic inflammation, and fecal calprotectin values have been shown to be predictive of relapse in asymptomatic patients with IBD[29-33]. In a biomarker discovery trial, the EMBARK study, serum matrix metalloproteinase 9 and serum IL-22 were found to be associated with inflammatory disease activity for patients with UC and CD respectively[34].

Pharmacogenomic testing has also become common place in IBD. A commonly used predictor of risk of adverse drug reactions and pharmacologic response is the utility of thiopurine methyltransferase (TPMT) genotyping and metabolite testing, as well as nudix hydrolase 15 (NUDT15) genotyping. Thiopurine use is associated with adverse effects (AEs) in up to 40% of patients[35]. TPMT genotype testing is cost effective, and heterozygous and homozygous TPMT genotypes correlate with AEs. Dose reduction in the TPMT variants significantly reduce adverse hematologic effects without reducing treatment efficacy[36]. NUDT15 variants, first elucidated in a Korean population, have also been more recently described as associated with thiopurine induced myelosuppression and is more predictive of myelosuppression in East Asians[37-39]. Furthermore, thiopurine metabolite testing can aid in dose optimization and compliance[40] and prevents hepatotoxicity by identifying a subgroup of thiopurine-‘shunters’ who preferentially produce the hepatotoxic metabolite 6-methylmercaptopurine.

Apart from this, several biomarkers as predictors of non-response to anti-TNF include higher oncostatin M expression[41,42] and low expression of triggering receptor expressed on myeloid cells 1[43,44]; antibody formation to anti-TNF is associated with the HLA-DQA1*05 genotype[45]. Higher baseline concentrations of serum cytokine IL-22, whose expression is induced by IL-23, is associated with greater likelihood of response to brazikumab[46].

OPTIMIZING AND MAXIMIZING CURRENT BIOLOGIC AGENTS
Therapeutic drug monitoring and drug dosing strategies

Introduction of therapeutic drug monitoring (TDM) has guided our approach in going beyond the therapeutic ceiling. Multiple studies have demonstrated an association with serum drug concentration of biologics, mainly in anti-TNF agents, and outcomes of patients[47-55]. It has assisted us in guiding dose modification (dose escalation or reduction), and informed us of primary or secondary loss of response, thus avoiding persistence of potentially ineffective therapy[56]. The approach of proactive vs reactive therapeutic drug monitoring remains a hotly debated topic[57].

Dashboard systems are clinical decision support tools utilizing computer software modelling to predict ideal personalized medication dosing[58,59]. This ‘model-based dosing’ has long been used by pharmacists and pharmacologists, for example, to dose antibiotics such as aminoglycosides. For anti-TNF dosing, dashboard driven pharmacokinetic (PK) dose optimization considers individual patient covariates including C-reactive protein, albumin, body-weight, sex and also serum drug levels. The PRECISION trial demonstrated that dashboard driven personalized dosing resulted in a significantly higher proportion of patients maintaining clinical remission after 1 year of treatment compared with patients that continued treatment without proactive adjustments: 88% vs 64%, respectively[60]. Utilizing the same PK dashboard system during Infliximab induction, Dubinsky et al[61] recently showed improved infliximab durability; and at 52 wk, 119/123 patients remained on infliximab in steroid free remission. The OPTIMISE trial is underway to evaluate the safety and efficacy of proactive TDM combined PK dashboard-driven infliximab dosing compared with standard of care dosing in patients with CD[62].

“Supratherapeutic” anti-TNF dosing

Numerous exposure-response relationship studies including post-hoc analyses of randomized controlled trials show a positive correlation between biologic drug concentrations and favorable therapeutic outcomes in IBD and other immune-mediated inflammatory diseases; higher drug concentrations are typically associated with improved therapeutic outcomes[63]. Conversely, lower drug concentrations, with or without anti-drug antibodies, are associated with treatment failure and drug discontinuation[64,65].

Multiple clinical studies have provided various TDM targets, especially with the anti-TNF agents infliximab and adalimumab, at various time points that are associated with outcomes of clinical remission[51,66]. Several observational studies have suggested that higher median infliximab concentrations are associated with superior clinical and biochemical remission rates. Given the wide variation in observed concentrations among responders, one may even wonder if the “therapeutic threshold” is identical for all patients and for the different phases of the treatment (induction vs maintenance and active vs quiescent disease).

Yarur et al[67], found that that levels of infliximab ≥ 10 mcg/mL were best associated with fistula healing, though surprisingly, a small number of patients required levels of ≥ 20 mcg/mL to achieve fistula healing. Feng et al[68] demonstrated that on incremental gains analysis, mucosal healing rates gradually increased as infliximab levels went up and reached a brief plateau (> 85%) when the infliximab trough level was 10 μg/mL. However, there was still a small proportion that seemingly benefited from an anti-TNF levels > 12 mcg/mL to achieve mucosal healing. Ungar et al[69] similarly demonstrated in a retrospective study a significant association between serum levels of anti-TNF agents and level of mucosal healing. They went on to propose that serum levels of 6-10 μg/mL for infliximab and 8-12 μg/mL for adalimumab are required to achieve mucosal healing in 80%-90% of patients with IBD.

Several other studies demonstrated that higher trough levels of infliximab and adalimumab are associated with those achieving mucosal healing in CD[68-79]. In a substudy of the TAILORIX trial, Bossuyt et al[71] found that infliximab trough level of 7.8 μg/mL at the end of induction (week 14) was associated with both radiologic response and remission. Continuously high infliximab exposure (infliximab > 5 μg/mL at all time points) was associated with radiologic response.

Low infliximab trough concentrations and the presence of antibodies to infliximab are associated with worse outcomes. Trough concentrations of > 3 μg/mL during maintenance is associated with sustained clinical outcomes[54,70,80,81]. Vande Casteele et al[82] concluded that an appropriate infliximab therapeutic window is between 3 and 7 μg/mL for IBD responders during maintenance therapy based on previous studies, and prospectively validated it in a randomized controlled trial (TAXIT).

The optimal “therapeutic window” for biologics remains to be elucidated, and the upper limit is unclear. While the abovementioned studies observed an association between certain trough concentration ranges and a corresponding degree of disease remission, the ‘ideal’ trough concentration to induce remission may vary between individuals. This may be due to a variety of factors such as an individual’s disease burden and therefore mucosal TNF burden, early vs advanced disease, and an individual’s unique pharmacodynamic makeup. There may be a subset of patients who might benefit from dose escalation to ‘supratherapeutic’ trough concentrations and reassuringly, there is little evidence to indicate greater toxicity with higher infliximab levels.

Sequencing of biologics

While it remains a conventional strategy to offer an anti-TNF agent as the first-line biologic, it has been well established from network meta-analyses that anti-TNF non-responders do not have an optimal response after switching to second-line biologics[12-14]. It thereby raises the question whether certain individuals would benefit from receiving these traditionally second-line biologics as first-line therapy options, and highlights the importance of optimal biologic sequencing.

There are limited head to head trials between biologic agents: In the VARSITY trial (adalimumab vs vedolizumab) for moderate to severe UC, vedolizumab demonstrated superiority in clinical and endoscopic remission but not corticosteroid free clinical remission[83]. In the SEAVUE study (adalimumab vs ustekinumab) for moderate to severe Crohn’s, ustekinumab failed to demonstrate superiority over adalimumab[84].

In the Galaxi-1 study involving participants with moderately to severely active CD, guselkumab was compared to placebo and ustekinumab. It was a phase 2, dose-ranging study[85], not powered to evaluate potential differences in efficacy and safety between guselkumab and ustekinumab.

From the HIBISCUS and GARDENIA trials[86,87] Etrolizumab vs adalimumab or infliximab in moderate to severe UC- failed to demonstrate superiority.

While offering novel alternate pathway biologics as first-line therapy may gain traction in the near future, this approach is often limited by government access in many jurisdictions. Access to newer therapies for adult patients is already limited by licensing authorities, but the pathways remain even more restricted for pediatric patients. Access to biologics in pediatric patients is often on compassionate grounds, due to a significant lag in clinical trials and therefore delaying official approval from medical licensing authorities such as the European Medicines Agency and United States FDA. There is a need for this cohort of patients to have better access to new/emerging therapies through more advanced pharmacogenomic, pharmacokinetic and pharmacodynamic modelling. This is to allow earlier initiation of trials or better ways to “extrapolate” adult data for presentation to licensing authorities to allow for use in children.

Dual biologics and combinations of newer advanced therapies

Combinations of biologic agents and recently combinations of biologics with newer small molecule agents have been attempted to go beyond our current therapeutic ceiling. This concept is not new, however, and was first attempted by Sands et al[88] as a randomized controlled trial comparing the safety and tolerability of patients on infliximab not in remission and adding natalizumab vs a placebo arm. Although the main trial ran for 10 wk and was not powered to assess for differences in efficacy between the groups, there was a higher proportion of patients achieving a clinical response at each time point and this proportion continues to increase over time in the combination biologic group, compared to response rates for the monotherapy arm which remained unchanged.

Since then, there has been much in the literature of dual biologics or with newer small molecule therapies, but as case reports or case series and observational cohort studies (Table 2). The data is largely heterogenous but reassuringly has demonstrated acceptable safety for patients with refractory IBD with no new concerning signals[89,90].

Table 2 Publications of dual biologics.
Ref.
Type
Number of participants/IBD type
Biologic combinations
Therapy duration or follow up (mo)
Outcomes
Buer et al[97], 2018Case series, prospectively followedAdult: 10 (4 CD, 6 UC)Anti-TNF, adding on vedolizumab. Combination was intended as a bridging therapy12-20Clinical: HBI, PMS, 100 % CRem, 50% endoscopic remission. No serious AE (3 minor infections)
Olbjørn et al[98], 2020Case seriesPediatric: 13 (9 CD, 4 UC)Anti-TNF + vedolizumab (8), anti-TNF + ustekinumab (5), (for anti-TNF side effects)N/A3/8 (37.5%) Clinical and biochemical remission
Kwapisz et al[99], 2021Case seriesAdult: 15 (14 CD, 1 UC)8 vedolizumab + anti-TNF, 2 ustekinumab + anti-TNF, 5 vedolizumab + ustekinumab2473% CRes, 44% ERes, 27% SE, 20% surgery
Yang et al[100], 2020Retrospective cohortAdult: 22 (CD)24 combinations: 13 vedolizumab + anti-TNF, 8 vedolizumab + ustekinumab, 3 ustekinumab + anti-TNF1Endoscopic, PRO2 response/remission, CRP, 50% CRes, 36% SF CRem, 43% ERes, 4% SE (1 SLE-1 cancer), 33% surgery
Glassner et al[101], 2020Retrospective cohort5053 combinations: 25 vedolizumab + ustekinumab, multiple other combinations5.5-1350% CRem, 34% ERem, 16% SE, 12% surgery
Privitera et al[102], 2020Case series, indication active IBD and active EIMAdult: 16 (11 CD, 4 UC)Variety of combinations. Most frequent: 3 vedolizumab + adalimumab, 3 vedolizumab + ustekinumab0.5At 6 mo: Response IBD/EIM: 42.8%, 11%; Remission IBD/EIM: 14.2%, 55.5%, AE: 3/16 (18.8%)
Dolinger et al[103], 2021Case seriesPediatric: 16: (CD 7, UC 8, IBD-U 1)Vedolizumab + ustekinumab, vedolizumab + tofacitinib, ustekinumab + tofacitinib6SF remission at 6 mo 12/16 (75%)
Goessens et al[104], 2021Retrospective cohort, heterogenous, active IBD and/or EIMAdult: 98 (CD 58, UC 40)Anti-TNF + vedolizumab, anti-TNF + anti-IL, anti-IL + vedolizumab, tofacitinib + anti-TNF, tofacitinib + vedolizumab, anti-IL + anti-IL, others5-16PGA: Complete or partial improvement was observed in 21/80 (26%) and 35/80 (44%); Mean clinical disease activity for IBD: Significantly higher prior to combination than during combination (2.2 +/- 0.7 vs 1.2 +/- 1.1; P < 0.0001). Simple clinical activity scores (quiescent scores 0, mild scores 1, moderate scores 2 and severe scores 3

In the pipeline, there are several randomized controlled trials evaluating the efficacy of combination biologics. The EXPLORER trial (ClinicalTrials.gov Identifier: NCT02764762) in high-risk CD patients involved triple combination therapy with vedolizumab, adalimumab and methotrexate which has completed but not yet reported.

The phase 2a VEGA study evaluated the safety and efficacy of combination induction therapy with guselkumab plus golimumab (GOL) vs monotherapy with guselkumab or GOL in adults with moderately to severely active UC through to week 12, most recently presented at the European Crohn’s and Colitis Organization 2022 congress[91,92]. A greater proportion of patients who received combination therapy achieved clinical response as judged by Mayo score at week 12 (83.1%) vs guselkumab (74.6%) or GOL (61.1%). Similarly, the proportion of patients who achieved clinical remission in the combination group (36.6%) was greater than that in the monotherapy group (21.1% and 22.2%, respectively). The DUET UC (ClinicalTrials.gov Identifier: NCT05242484) is a phase 2b study of combination therapy with guselkumab and GOL (JNJ-78934804) in participants with moderately to severely active UC that is planned; the DUET CD (ClinicalTrials.gov Identifier: NCT05242471) is a phase 2b study of the same biologic combination in individuals with moderately to severely active CD, and is currently recruiting trial participants.

The other considerations to overcome current therapeutic plateaus with biologic agents include added adjunctive therapies such as vitamin D, curcumin, microbiome alteration via dietary modification, exclusive enteral nutrition and probiotics[93].

CONCLUSION

Ongoing efforts in adding to and optimizing IBD treatments must be commended. However, remission rates are still far from optimal with current treatment approaches. The urgent need to develop new therapeutics also brings us to the challenge that we must meet: Improving the design and delivery of clinical trials, allowing generalizability, be of clinical equipoise and to factor in biomarker discovery. Advances in basic science, translational and clinical aspects of drug development is essential to achieve breakthroughs in IBD therapeutics that meet the needs of patients, physicians and health regulators[94-96]. In conclusion, in addition to the strategies as aforementioned, to go beyond our current therapeutic ceiling requires not only early diagnosis or early stratification but early treatment. This entails incorporating a multiomics approach to better personalize treatment, sequence or combine our therapies, and incorporate the ever-advancing artificial intelligence technology, rather than a one-size-fits all approach[95]. These goals remain attainable and we continue to have a sense of optimism.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author's Membership in Professional Societies: Gastroenterological Society of Singapore.

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Australia

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Al-Mahayri ZN, United Arab Emirates; Zhao G, China S-Editor: Zhang H L-Editor: Filipodia P-Editor: Zhang H

References
1.  Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, Benchimol EI, Panaccione R, Ghosh S, Barkema HW, Kaplan GG. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology. 2012;142:46-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3134]  [Cited by in F6Publishing: 3306]  [Article Influence: 275.5]  [Reference Citation Analysis (1)]
2.  Thia KT, Loftus EV Jr, Sandborn WJ, Yang SK. An update on the epidemiology of inflammatory bowel disease in Asia. Am J Gastroenterol. 2008;103:3167-3182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 371]  [Cited by in F6Publishing: 393]  [Article Influence: 24.6]  [Reference Citation Analysis (1)]
3.  Melsheimer R, Geldhof A, Apaolaza I, Schaible T. Remicade® (infliximab): 20 years of contributions to science and medicine. Biologics. 2019;13:139-178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 56]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
4.  Baumgart DC, Le Berre C. Newer Biologic and Small-Molecule Therapies for Inflammatory Bowel Disease. N Engl J Med. 2021;385:1302-1315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 119]  [Article Influence: 39.7]  [Reference Citation Analysis (0)]
5.  Cholapranee A, Hazlewood GS, Kaplan GG, Peyrin-Biroulet L, Ananthakrishnan AN. Systematic review with meta-analysis: comparative efficacy of biologics for induction and maintenance of mucosal healing in Crohn's disease and ulcerative colitis controlled trials. Aliment Pharmacol Ther. 2017;45:1291-1302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 220]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
6.  Colombel JF, Panaccione R, Bossuyt P, Lukas M, Baert F, Vaňásek T, Danalioglu A, Novacek G, Armuzzi A, Hébuterne X, Travis S, Danese S, Reinisch W, Sandborn WJ, Rutgeerts P, Hommes D, Schreiber S, Neimark E, Huang B, Zhou Q, Mendez P, Petersson J, Wallace K, Robinson AM, Thakkar RB, D'Haens G. Effect of tight control management on Crohn's disease (CALM): a multicentre, randomised, controlled phase 3 trial. Lancet. 2017;390:2779-2789.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 503]  [Cited by in F6Publishing: 560]  [Article Influence: 80.0]  [Reference Citation Analysis (0)]
7.  Peyrin-Biroulet L, Sandborn W, Sands BE, Reinisch W, Bemelman W, Bryant RV, D'Haens G, Dotan I, Dubinsky M, Feagan B, Fiorino G, Gearry R, Krishnareddy S, Lakatos PL, Loftus EV Jr, Marteau P, Munkholm P, Murdoch TB, Ordás I, Panaccione R, Riddell RH, Ruel J, Rubin DT, Samaan M, Siegel CA, Silverberg MS, Stoker J, Schreiber S, Travis S, Van Assche G, Danese S, Panes J, Bouguen G, O'Donnell S, Pariente B, Winer S, Hanauer S, Colombel JF. Selecting Therapeutic Targets in Inflammatory Bowel Disease (STRIDE): Determining Therapeutic Goals for Treat-to-Target. Am J Gastroenterol. 2015;110:1324-1338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1160]  [Cited by in F6Publishing: 1258]  [Article Influence: 139.8]  [Reference Citation Analysis (0)]
8.  Turner D, Ricciuto A, Lewis A, D'Amico F, Dhaliwal J, Griffiths AM, Bettenworth D, Sandborn WJ, Sands BE, Reinisch W, Schölmerich J, Bemelman W, Danese S, Mary JY, Rubin D, Colombel JF, Peyrin-Biroulet L, Dotan I, Abreu MT, Dignass A; International Organization for the Study of IBD. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 473]  [Cited by in F6Publishing: 963]  [Article Influence: 321.0]  [Reference Citation Analysis (0)]
9.  Alipour O, Gualti A, Shao L, Zhang B. Systematic review and meta-analysis: real-world data rates of deep remission with anti-TNFα in inflammatory bowel disease. BMC Gastroenterol. 2021;21:312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
10.  Papamichael K, Gils A, Rutgeerts P, Levesque BG, Vermeire S, Sandborn WJ, Vande Casteele N. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 164]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
11.  Roda G, Jharap B, Neeraj N, Colombel JF. Loss of Response to Anti-TNFs: Definition, Epidemiology, and Management. Clin Transl Gastroenterol. 2016;7:e135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 443]  [Article Influence: 55.4]  [Reference Citation Analysis (0)]
12.  Singh S, George J, Boland BS, Vande Casteele N, Sandborn WJ. 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 Crohns Colitis. 2018;12:635-643.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 138]  [Article Influence: 23.0]  [Reference Citation Analysis (0)]
13.  Singh S, Murad MH, Fumery M, Dulai PS, Sandborn WJ. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 212]  [Article Influence: 53.0]  [Reference Citation Analysis (0)]
14.  Singh S, Murad MH, Fumery M, Sedano R, Jairath V, Panaccione R, Sandborn WJ, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 111]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
15.  Noor NM, Verstockt B, Parkes M, Lee JC. Personalised medicine in Crohn's disease. Lancet Gastroenterol Hepatol. 2020;5:80-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 47]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
16.  Danese S, Fiorino G, Fernandes C, Peyrin-Biroulet L. Catching the therapeutic window of opportunity in early Crohn's disease. Curr Drug Targets. 2014;15:1056-1063.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
17.  Kang B, Choe YH. Early Biologic Treatment in Pediatric Crohn's Disease: Catching the Therapeutic Window of Opportunity in Early Disease by Treat-to-Target. Pediatr Gastroenterol Hepatol Nutr. 2018;21:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 46]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
18.  D'Haens G, Baert F, van Assche G, Caenepeel P, Vergauwe P, Tuynman H, De Vos M, van Deventer S, Stitt L, Donner A, Vermeire S, Van De Mierop FJ, Coche JR, van der Woude J, Ochsenkühn T, van Bodegraven AA, Van Hootegem PP, Lambrecht GL, Mana F, Rutgeerts P, Feagan BG, Hommes D; Belgian Inflammatory Bowel Disease Research Group;  North-Holland Gut Club. Early combined immunosuppression or conventional management in patients with newly diagnosed Crohn's disease: an open randomised trial. Lancet. 2008;371:660-667.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 922]  [Cited by in F6Publishing: 900]  [Article Influence: 56.3]  [Reference Citation Analysis (0)]
19.  Khanna R, Bressler B, Levesque BG, Zou G, Stitt LW, Greenberg GR, Panaccione R, Bitton A, Paré P, Vermeire S, D'Haens G, MacIntosh D, Sandborn WJ, Donner A, Vandervoort MK, Morris JC, Feagan BG; REACT Study Investigators. Early combined immunosuppression for the management of Crohn's disease (REACT): a cluster randomised controlled trial. Lancet. 2015;386:1825-1834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 307]  [Cited by in F6Publishing: 303]  [Article Influence: 33.7]  [Reference Citation Analysis (0)]
20.  Choung RS, Princen F, Stockfisch TP, Torres J, Maue AC, Porter CK, Leon F, De Vroey B, Singh S, Riddle MS, Murray JA, Colombel JF; PREDICTS Study Team. Serologic microbial associated markers can predict Crohn's disease behaviour years before disease diagnosis. Aliment Pharmacol Ther. 2016;43:1300-1310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 90]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
21.  Dubinsky M. What is the role of serological markers in IBD? Dig Dis. 2009;27:259-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
22.  Dubinsky M, Braun J. Diagnostic and Prognostic Microbial Biomarkers in Inflammatory Bowel Diseases. Gastroenterology. 2015;149:1265-1274.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 47]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
23.  Ferrante M, Henckaerts L, Joossens M, Pierik M, Joossens S, Dotan N, Norman GL, Altstock RT, Van Steen K, Rutgeerts P, Van Assche G, Vermeire S. New serological markers in inflammatory bowel disease are associated with complicated disease behaviour. Gut. 2007;56:1394-1403.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 223]  [Cited by in F6Publishing: 222]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
24.  Zhang Z, Li C, Zhao X, Lv C, He Q, Lei S, Guo Y, Zhi F. Anti-Saccharomyces cerevisiae antibodies associate with phenotypes and higher risk for surgery in Crohn's disease: a meta-analysis. Dig Dis Sci. 2012;57:2944-2954.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
25.  Lee JC, Lyons PA, McKinney EF, Sowerby JM, Carr EJ, Bredin F, Rickman HM, Ratlamwala H, Hatton A, Rayner TF, Parkes M, Smith KG. Gene expression profiling of CD8+ T cells predicts prognosis in patients with Crohn disease and ulcerative colitis. J Clin Invest. 2011;121:4170-4179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 208]  [Cited by in F6Publishing: 228]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
26.  Allez M, Auzolle C, Ngollo M, Bottois H, Chardiny V, Corraliza AM, Salas A, Perez K, Stefanescu C, Nancey S, Buisson A, Pariente B, Fumery M, Sokol H, Tréton X, Barnich N, Seksik P, Le Bourhis L; REMIND Study Group. T cell clonal expansions in ileal Crohn's disease are associated with smoking behaviour and postoperative recurrence. Gut. 2019;68:1961-1970.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
27.  Biasci D, Lee JC, Noor NM, Pombal DR, Hou M, Lewis N, Ahmad T, Hart A, Parkes M, McKinney EF, Lyons PA, Smith KGC. A blood-based prognostic biomarker in IBD. Gut. 2019;68:1386-1395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 117]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
28.  Parkes M, Noor NM, Dowling F, Leung H, Bond S, Whitehead L, Upponi S, Kinnon P, Sandham AP, Lyons PA, McKinney EF, Smith KGC, Lee JC. PRedicting Outcomes For Crohn's dIsease using a moLecular biomarkEr (PROFILE): protocol for a multicentre, randomised, biomarker-stratified trial. BMJ Open. 2018;8:e026767.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 41]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
29.  D'Haens G, Ferrante M, Vermeire S, Baert F, Noman M, Moortgat L, Geens P, Iwens D, Aerden I, Van Assche G, Van Olmen G, Rutgeerts P. Fecal calprotectin is a surrogate marker for endoscopic lesions in inflammatory bowel disease. Inflamm Bowel Dis. 2012;18:2218-2224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 553]  [Cited by in F6Publishing: 582]  [Article Influence: 48.5]  [Reference Citation Analysis (0)]
30.  De Vos M, Louis EJ, Jahnsen J, Vandervoort JG, Noman M, Dewit O, Dʼhaens GR, Franchimont D, Baert FJ, Torp RA, Henriksen M, Potvin PM, Van Hootegem PP, Hindryckx PM, Moreels TG, Collard A, Karlsen LN, Kittang E, Lambrecht G, Grimstad T, Koch J, Lygren I, Coche JC, Mana F, Van Gossum A, Belaiche J, Cool MR, Fontaine F, Maisin JM, Muls V, Neuville B, Staessen DA, Van Assche GA, de Lange T, Solberg IC, Vander Cruyssen BJ, Vermeire SA. Consecutive fecal calprotectin measurements to predict relapse in patients with ulcerative colitis receiving infliximab maintenance therapy. Inflamm Bowel Dis. 2013;19:2111-2117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 118]  [Article Influence: 10.7]  [Reference Citation Analysis (0)]
31.  Mao R, Xiao YL, Gao X, Chen BL, He Y, Yang L, Hu PJ, Chen MH. Fecal calprotectin in predicting relapse of inflammatory bowel diseases: a meta-analysis of prospective studies. Inflamm Bowel Dis. 2012;18:1894-1899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 222]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
32.  Schoepfer AM, Beglinger C, Straumann A, Safroneeva E, Romero Y, Armstrong D, Schmidt C, Trummler M, Pittet V, Vavricka SR. Fecal calprotectin more accurately reflects endoscopic activity of ulcerative colitis than the Lichtiger Index, C-reactive protein, platelets, hemoglobin, and blood leukocytes. Inflamm Bowel Dis. 2013;19:332-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 207]  [Article Influence: 18.8]  [Reference Citation Analysis (0)]
33.  Zittan E, Kelly OB, Kirsch R, Milgrom R, Burns J, Nguyen GC, Croitoru K, Van Assche G, Silverberg MS, Steinhart AH. Low Fecal Calprotectin Correlates with Histological Remission and Mucosal Healing in Ulcerative Colitis and Colonic Crohn's Disease. Inflamm Bowel Dis. 2016;22:623-630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 120]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
34.  Faubion WA Jr, Fletcher JG, O'Byrne S, Feagan BG, de Villiers WJ, Salzberg B, Plevy S, Proctor DD, Valentine JF, Higgins PD, Harris JM, Diehl L, Wright L, Tew GW, Luca D, Basu K, Keir ME. EMerging BiomARKers in Inflammatory Bowel Disease (EMBARK) study identifies fecal calprotectin, serum MMP9, and serum IL-22 as a novel combination of biomarkers for Crohn's disease activity: role of cross-sectional imaging. Am J Gastroenterol. 2013;108:1891-1900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 81]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
35.  Ansari A, Arenas M, Greenfield SM, Morris D, Lindsay J, Gilshenan K, Smith M, Lewis C, Marinaki A, Duley J, Sanderson J. Prospective evaluation of the pharmacogenetics of azathioprine in the treatment of inflammatory bowel disease. Aliment Pharmacol Ther. 2008;28:973-983.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 127]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
36.  Coenen MJ, de Jong DJ, van Marrewijk CJ, Derijks LJ, Vermeulen SH, Wong DR, Klungel OH, Verbeek AL, Hooymans PM, Peters WH, te Morsche RH, Newman WG, Scheffer H, Guchelaar HJ, Franke B; TOPIC Recruitment Team. Identification of Patients With Variants in TPMT and Dose Reduction Reduces Hematologic Events During Thiopurine Treatment of Inflammatory Bowel Disease. Gastroenterology. 2015;149:907-17.e7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 132]  [Cited by in F6Publishing: 149]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
37.  Walker GJ, Harrison JW, Heap GA, Voskuil MD, Andersen V, Anderson CA, Ananthakrishnan AN, Barrett JC, Beaugerie L, Bewshea CM, Cole AT, Cummings FR, Daly MJ, Ellul P, Fedorak RN, Festen EAM, Florin TH, Gaya DR, Halfvarson J, Hart AL, Heerasing NM, Hendy P, Irving PM, Jones SE, Koskela J, Lindsay JO, Mansfield JC, McGovern D, Parkes M, Pollok RCG, Ramakrishnan S, Rampton DS, Rivas MA, Russell RK, Schultz M, Sebastian S, Seksik P, Singh A, So K, Sokol H, Subramaniam K, Todd A, Annese V, Weersma RK, Xavier R, Ward R, Weedon MN, Goodhand JR, Kennedy NA, Ahmad T; IBD Pharmacogenetics Study Group. Association of Genetic Variants in NUDT15 With Thiopurine-Induced Myelosuppression in Patients With Inflammatory Bowel Disease. JAMA. 2019;321:773-785.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 107]  [Article Influence: 21.4]  [Reference Citation Analysis (0)]
38.  Yang SK, Hong M, Baek J, Choi H, Zhao W, Jung Y, Haritunians T, Ye BD, Kim KJ, Park SH, Park SK, Yang DH, Dubinsky M, Lee I, McGovern DP, Liu J, Song K. A common missense variant in NUDT15 confers susceptibility to thiopurine-induced leukopenia. Nat Genet. 2014;46:1017-1020.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 412]  [Cited by in F6Publishing: 379]  [Article Influence: 37.9]  [Reference Citation Analysis (0)]
39.  Zhu X, Wang XD, Chao K, Zhi M, Zheng H, Ruan HL, Xin S, Ding N, Hu PJ, Huang M, Gao X. NUDT15 polymorphisms are better than thiopurine S-methyltransferase as predictor of risk for thiopurine-induced leukopenia in Chinese patients with Crohn's disease. Aliment Pharmacol Ther. 2016;44:967-975.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 65]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
40.  Lamb CA, Kennedy NA, Raine T, Hendy PA, Smith PJ, Limdi JK, Hayee B, Lomer MCE, Parkes GC, Selinger C, Barrett KJ, Davies RJ, Bennett C, Gittens S, Dunlop MG, Faiz O, Fraser A, Garrick V, Johnston PD, Parkes M, Sanderson J, Terry H; IBD guidelines eDelphi consensus group, Gaya DR, Iqbal TH, Taylor SA, Smith M, Brookes M, Hansen R, Hawthorne AB. British Society of Gastroenterology consensus guidelines on the management of inflammatory bowel disease in adults. Gut. 2019;68:s1-s106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1230]  [Cited by in F6Publishing: 1184]  [Article Influence: 236.8]  [Reference Citation Analysis (0)]
41.  West NR, Hegazy AN, Owens BMJ, Bullers SJ, Linggi B, Buonocore S, Coccia M, Görtz D, This S, Stockenhuber K, Pott J, Friedrich M, Ryzhakov G, Baribaud F, Brodmerkel C, Cieluch C, Rahman N, Müller-Newen G, Owens RJ, Kühl AA, Maloy KJ, Plevy SE; Oxford IBD Cohort Investigators, Keshav S, Travis SPL, Powrie F. Oncostatin M drives intestinal inflammation and predicts response to tumor necrosis factor-neutralizing therapy in patients with inflammatory bowel disease. Nat Med. 2017;23:579-589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 550]  [Cited by in F6Publishing: 475]  [Article Influence: 67.9]  [Reference Citation Analysis (0)]
42.  Verstockt S, Verstockt B, Machiels K, Vancamelbeke M, Ferrante M, Cleynen I, De Hertogh G, Vermeire S. Oncostatin M Is a Biomarker of Diagnosis, Worse Disease Prognosis, and Therapeutic Nonresponse in Inflammatory Bowel Disease. Inflamm Bowel Dis. 2021;27:1564-1575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 42]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
43.  Verstockt B, Verstockt S, Dehairs J, Ballet V, Blevi H, Wollants WJ, Breynaert C, Van Assche G, Vermeire S, Ferrante M. Low TREM1 expression in whole blood predicts anti-TNF response in inflammatory bowel disease. EBioMedicine. 2019;40:733-742.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 96]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
44.  Verstockt B, Verstockt S, Blevi H, Cleynen I, de Bruyn M, Van Assche G, Vermeire S, Ferrante M. TREM-1, the ideal predictive biomarker for endoscopic healing in anti-TNF-treated Crohn's disease patients? Gut. 2019;68:1531-1533.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 38]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
45.  Sazonovs A, Kennedy NA, Moutsianas L, Heap GA, Rice DL, Reppell M, Bewshea CM, Chanchlani N, Walker GJ, Perry MH, McDonald TJ, Lees CW, Cummings JRF, Parkes M, Mansfield JC, Irving PM, Barrett JC, McGovern D, Goodhand JR, Anderson CA, Ahmad T; PANTS Consortium. HLA-DQA1*05 Carriage Associated With Development of Anti-Drug Antibodies to Infliximab and Adalimumab in Patients With Crohn's Disease. Gastroenterology. 2020;158:189-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 234]  [Cited by in F6Publishing: 229]  [Article Influence: 57.3]  [Reference Citation Analysis (0)]
46.  Sands BE, Chen J, Feagan BG, Penney M, Rees WA, Danese S, Higgins PDR, Newbold P, Faggioni R, Patra K, Li J, Klekotka P, Morehouse C, Pulkstenis E, Drappa J, van der Merwe R, Gasser RA Jr. Efficacy and Safety of MEDI2070, an Antibody Against Interleukin 23, in Patients With Moderate to Severe Crohn's Disease: A Phase 2a Study. Gastroenterology. 2017;153:77-86.e6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 192]  [Article Influence: 27.4]  [Reference Citation Analysis (0)]
47.  Adedokun OJ, Xu Z, Gasink C, Jacobstein D, Szapary P, Johanns J, Gao LL, Davis HM, Hanauer SB, Feagan BG, Ghosh S, Sandborn WJ. Pharmacokinetics and Exposure Response Relationships of Ustekinumab in Patients With Crohn's Disease. Gastroenterology. 2018;154:1660-1671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 166]  [Article Influence: 27.7]  [Reference Citation Analysis (0)]
48.  Baert F, Vande Casteele N, 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 Pharmacol Ther. 2014;40:1324-1332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 58]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
49.  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 Crohns Colitis. 2017;11:549-555.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 37]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
50.  Dreesen E, Verstockt B, Bian S, de Bruyn M, Compernolle G, Tops S, Noman M, Van Assche G, Ferrante M, Gils A, Vermeire S. Evidence to Support Monitoring of Vedolizumab Trough Concentrations in Patients With Inflammatory Bowel Diseases. Clin Gastroenterol Hepatol. 2018;16:1937-1946.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 106]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
51.  Kennedy NA, Heap GA, Green HD, Hamilton B, Bewshea C, Walker GJ, Thomas A, Nice R, Perry MH, Bouri S, Chanchlani N, Heerasing NM, Hendy P, Lin S, Gaya DR, Cummings JRF, Selinger CP, Lees CW, Hart AL, Parkes M, Sebastian S, Mansfield JC, Irving PM, Lindsay J, Russell RK, McDonald TJ, McGovern D, Goodhand JR, Ahmad T; UK Inflammatory Bowel Disease Pharmacogenetics Study Group. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 299]  [Cited by in F6Publishing: 381]  [Article Influence: 76.2]  [Reference Citation Analysis (0)]
52.  Papamichael K, Rakowsky S, Rivera C, Cheifetz AS, Osterman MT. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 41]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
53.  Ungar B, Engel T, Yablecovitch D, Lahat A, Lang A, Avidan B, Har-Noy O, Carter D, Levhar N, Selinger L, Neuman S, Natour OH, Yavzori M, Fudim E, Picard O, Kopylov U, Chowers Y, Naftali T, Broide E, Shachar E, Eliakim R, Ben-Horin S. Prospective Observational Evaluation of Time-Dependency of Adalimumab Immunogenicity and Drug Concentrations: The POETIC Study. Am J Gastroenterol. 2018;113:890-898.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 53]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
54.  Vande Casteele N, Khanna R, Levesque BG, Stitt L, Zou GY, Singh S, Lockton S, Hauenstein S, Ohrmund L, Greenberg GR, Rutgeerts PJ, Gils A, Sandborn WJ, Vermeire S, Feagan BG. The relationship between infliximab concentrations, antibodies to infliximab and disease activity in Crohn's disease. Gut. 2015;64:1539-1545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 204]  [Article Influence: 22.7]  [Reference Citation Analysis (0)]
55.  Williet N, Boschetti G, Fovet M, Di Bernado T, Claudez P, Del Tedesco E, Jarlot C, Rinaldi L, Berger A, Phelip JM, Flourie B, Nancey S, Paul S, Roblin X. Association Between Low Trough Levels of Vedolizumab During Induction Therapy for Inflammatory Bowel Diseases and Need for Additional Doses Within 6 Months. Clin Gastroenterol Hepatol. 2017;15:1750-1757.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 100]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
56.  Feuerstein JD, Nguyen GC, Kupfer SS, Falck-Ytter Y, Singh S; American Gastroenterological Association Institute Clinical Guidelines Committee. American Gastroenterological Association Institute Guideline on Therapeutic Drug Monitoring in Inflammatory Bowel Disease. Gastroenterology. 2017;153:827-834.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 352]  [Cited by in F6Publishing: 389]  [Article Influence: 55.6]  [Reference Citation Analysis (0)]
57.  Papamichael K, Afif W, Drobne D, Dubinsky MC, Ferrante M, Irving PM, Kamperidis N, Kobayashi T, Kotze PG, Lambert J, Noor NM, Roblin X, Roda G, Vande Casteele N, Yarur AJ, Arebi N, Danese S, Paul S, Sandborn WJ, Vermeire S, Cheifetz AS, Peyrin-Biroulet L; International Consortium for Therapeutic Drug Monitoring. Therapeutic drug monitoring of biologics in inflammatory bowel disease: unmet needs and future perspectives. Lancet Gastroenterol Hepatol. 2022;7:171-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 53]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
58.  Mould DR, Dubinsky MC. Dashboard systems: Pharmacokinetic/pharmacodynamic mediated dose optimization for monoclonal antibodies. J Clin Pharmacol. 2015;55 Suppl 3:S51-S59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (1)]
59.  Mould DR, Upton RN, Wojciechowski J, Phan BL, Tse S, Dubinsky MC. Dashboards for Therapeutic Monoclonal Antibodies: Learning and Confirming. AAPS J. 2018;20:76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
60.  Strik AS, Löwenberg M, Mould DR, Berends SE, Ponsioen CI, van den Brande JMH, Jansen JM, Hoekman DR, Brandse JF, Duijvestein M, Gecse KB, de Vries A, Mathôt RA, D'Haens GR. Efficacy of dashboard driven dosing of infliximab in inflammatory bowel disease patients; a randomized controlled trial. Scand J Gastroenterol. 2021;56:145-154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 45]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
61.  Dubinsky MC, Mendiolaza ML, Phan BL, Moran HR, Tse SS, Mould DR. Dashboard-Driven Accelerated Infliximab Induction Dosing Increases Infliximab Durability and Reduces Immunogenicity. Inflamm Bowel Dis. 2022;28:1375-1385.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 31]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
62.  Papamichael K, Jairath V, Zou G, Cohen B, Ritter T, Sands B, Siegel C, Valentine J, Smith M, Vande Casteele N, Dubinsky M, Cheifetz A. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 16]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
63.  Papamichael K, Vogelzang EH, Lambert J, Wolbink G, Cheifetz AS. Therapeutic drug monitoring with biologic agents in immune mediated inflammatory diseases. Expert Rev Clin Immunol. 2019;15:837-848.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 57]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
64.  Sparrow MP, Papamichael K, Ward MG, Riviere P, Laharie D, Paul S, Roblin X. Therapeutic Drug Monitoring of Biologics During Induction to Prevent Primary Non-Response. J Crohns Colitis. 2020;14:542-556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 44]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
65.  Buhl S, Dorn-Rasmussen M, Brynskov J, Ainsworth MA, Bendtzen K, Klausen PH, Bolstad N, Warren DJ, Steenholdt C. Therapeutic thresholds and mechanisms for primary non-response to infliximab in inflammatory bowel disease. Scand J Gastroenterol. 2020;55:884-890.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
66.  Papamichael K, Vande Casteele N, Jeyarajah J, Jairath V, Osterman MT, Cheifetz AS. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 14]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
67.  Yarur AJ, Kanagala V, Stein DJ, Czul F, Quintero MA, Agrawal D, Patel A, Best K, Fox C, Idstein K, Abreu MT. Higher infliximab trough levels are associated with perianal fistula healing in patients with Crohn's disease. Aliment Pharmacol Ther. 2017;45:933-940.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 191]  [Article Influence: 27.3]  [Reference Citation Analysis (1)]
68.  Feng T, Chen B, Ungar B, Qiu Y, Zhang S, He J, Lin S, He Y, Zeng Z, Ben-Horin S, Chen M, Mao R. Association of Infliximab Levels With Mucosal Healing Is Time-Dependent in Crohn's Disease: Higher Drug Exposure Is Required Postinduction Than During Maintenance Treatment. Inflamm Bowel Dis. 2019;25:1813-1821.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
69.  Ungar B, Levy I, Yavne Y, Yavzori M, Picard O, Fudim E, Loebstein R, Chowers Y, Eliakim R, Kopylov U, Ben-Horin S. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 281]  [Article Influence: 35.1]  [Reference Citation Analysis (0)]
70.  Adedokun OJ, Sandborn WJ, Feagan BG, Rutgeerts P, Xu Z, Marano CW, Johanns J, Zhou H, Davis HM, Cornillie F, Reinisch W. Association between serum concentration of infliximab and efficacy in adult patients with ulcerative colitis. Gastroenterology. 2014;147:1296-1307.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 244]  [Cited by in F6Publishing: 249]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
71.  Bossuyt P, Dreesen E, Rimola J, Devuysere S, De Bruecker Y, Vanslembrouck R, Laurent V, Zappa M, Savoye-Collet C, Pariente B, Filippi J, Baert F, D'Haens G, Laharie D, Peyrin-Biroulet L, Vermeire S; TAILORIX study group. Infliximab Exposure Associates With Radiologic Evidence of Healing in Patients With Crohn's Disease. Clin Gastroenterol Hepatol. 2021;19:947-954.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 15]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
72.  Chaparro M, Barreiro-de Acosta M, Echarri A, Almendros R, Barrio J, Llao J, Gomollón F, Vera M, Cabriada JL, Guardiola J, Guerra I, Beltrán B, Roncero O, Busquets D, Taxonera C, Calvet X, Ferreiro-Iglesias R, Ollero Pena V, Bernardo D, Donday MG, Garre A, Godino A, Díaz A, Gisbert JP. Correlation Between Anti-TNF Serum Levels and Endoscopic Inflammation in Inflammatory Bowel Disease Patients. Dig Dis Sci. 2019;64:846-854.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
73.  Clarkston K, Tsai YT, Jackson K, Rosen MJ, Denson LA, Minar P. Development of Infliximab Target Concentrations During Induction in Pediatric Crohn Disease Patients. J Pediatr Gastroenterol Nutr. 2019;69:68-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 45]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
74.  El-Matary W, Walters TD, Huynh HQ, deBruyn J, Mack DR, Jacobson K, Sherlock ME, Church P, Wine E, Carroll MW, Benchimol EI, Lawrence S, Griffiths AM. Higher Postinduction Infliximab Serum Trough Levels Are Associated With Healing of Fistulizing Perianal Crohn's Disease in Children. Inflamm Bowel Dis. 2019;25:150-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 57]  [Article Influence: 11.4]  [Reference Citation Analysis (2)]
75.  Kang B, Choi SY, Choi YO, Lee SY, Baek SY, Sohn I, Choe BH, Lee HJ, Choe YH. Infliximab Trough Levels Are Associated With Mucosal Healing During Maintenance Treatment With Infliximab in Paediatric Crohn's Disease. J Crohns Colitis. 2019;13:189-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
76.  Lyles JL, Mulgund AA, Bauman LE, Su W, Fei L, Chona DL, Sharma P, Etter RK, Hellmann J, Denson LA, Minar P, Dykes DM, Rosen MJ. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 30]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
77.  Roblin X, Boschetti G, Duru G, Williet N, Deltedesco E, Phelip JM, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 17]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
78.  Vande Casteele N, Jeyarajah J, Jairath V, Feagan BG, Sandborn WJ. Infliximab Exposure-Response Relationship and Thresholds Associated With Endoscopic Healing in Patients With Ulcerative Colitis. Clin Gastroenterol Hepatol. 2019;17:1814-1821.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
79.  Zittan E, Steinhart AH, Goldstein P, Milgrom R, Gralnek IM, Silverberg MS. Post-Induction High Adalimumab Drug Levels Predict Biological Remission at Week 24 in Patients With Crohn's Disease. Clin Transl Gastroenterol. 2021;12:e00401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Reference Citation Analysis (0)]
80.  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 Crohns Colitis. 2013;7:736-743.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 193]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
81.  Cornillie F, Hanauer SB, Diamond RH, Wang J, Tang KL, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 279]  [Cited by in F6Publishing: 291]  [Article Influence: 29.1]  [Reference Citation Analysis (0)]
82.  Vande Casteele N, 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;148:1320-9.e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 616]  [Cited by in F6Publishing: 648]  [Article Influence: 72.0]  [Reference Citation Analysis (0)]
83.  Sands BE, Peyrin-Biroulet L, Loftus EV Jr, Danese S, Colombel JF, Törüner M, Jonaitis L, Abhyankar B, Chen J, Rogers R, Lirio RA, Bornstein JD, Schreiber S; VARSITY Study Group. Vedolizumab versus Adalimumab for Moderate-to-Severe Ulcerative Colitis. N Engl J Med. 2019;381:1215-1226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 349]  [Cited by in F6Publishing: 395]  [Article Influence: 79.0]  [Reference Citation Analysis (0)]
84.  Sands BE, Irving PM, Hoops T, Izanec JL, Gao LL, Gasink C, Greenspan A, Allez M, Danese S, Hanauer SB, Jairath V, Kuehbacher T, Lewis JD, Loftus EV Jr, Mihaly E, Panaccione R, Scherl E, Shchukina OB, Sandborn WJ; SEAVUE Study Group. Ustekinumab versus adalimumab for induction and maintenance therapy in biologic-naive patients with moderately to severely active Crohn's disease: a multicentre, randomised, double-blind, parallel-group, phase 3b trial. Lancet. 2022;399:2200-2211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 86]  [Article Influence: 43.0]  [Reference Citation Analysis (0)]
85.  Sandborn WJ, D'Haens GR, Reinisch W, Panés J, Chan D, Gonzalez S, Weisel K, Germinaro M, Frustaci ME, Yang Z, Adedokun OJ, Han C, Panaccione R, Hisamatsu T, Danese S, Rubin DT, Sands BE, Afzali A, Andrews JM, Feagan BG; GALAXI-1 Investigators. Guselkumab for the Treatment of Crohn's Disease: Induction Results From the Phase 2 GALAXI-1 Study. Gastroenterology. 2022;162:1650-1664.e8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 80]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]
86.  Danese S, Colombel JF, Lukas M, Gisbert JP, D'Haens G, Hayee B, Panaccione R, Kim HS, Reinisch W, Tyrrell H, Oh YS, Tole S, Chai A, Chamberlain-James K, Tang MT, Schreiber S; GARDENIA Study Group. Etrolizumab versus infliximab for the treatment of moderately to severely active ulcerative colitis (GARDENIA): a randomised, double-blind, double-dummy, phase 3 study. Lancet Gastroenterol Hepatol. 2022;7:118-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 37]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
87.  Rubin DT, Dotan I, DuVall A, Bouhnik Y, Radford-Smith G, Higgins PDR, Mishkin DS, Arrisi P, Scalori A, Oh YS, Tole S, Chai A, Chamberlain-James K, Lacey S, McBride J, Panés J; HIBISCUS Study Group. Etrolizumab versus adalimumab or placebo as induction therapy for moderately to severely active ulcerative colitis (HIBISCUS): two phase 3 randomised, controlled trials. Lancet Gastroenterol Hepatol. 2022;7:17-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 34]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
88.  Sands BE, Kozarek R, Spainhour J, Barish CF, Becker S, Goldberg L, Katz S, Goldblum R, Harrigan R, Hilton D, Hanauer SB. Safety and tolerability of concurrent natalizumab treatment for patients with Crohn's disease not in remission while receiving infliximab. Inflamm Bowel Dis. 2007;13:2-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 82]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
89.  Ahmed W, Galati J, Kumar A, Christos PJ, Longman R, Lukin DJ, Scherl E, Battat R. Dual Biologic or Small Molecule Therapy for Treatment of Inflammatory Bowel Disease: A Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol. 2022;20:e361-e379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 52]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
90.  Ribaldone DG, Pellicano R, Vernero M, Caviglia GP, Saracco GM, Morino M, Astegiano M. Dual biological therapy with anti-TNF, vedolizumab or ustekinumab in inflammatory bowel disease: a systematic review with pool analysis. Scand J Gastroenterol. 2019;54:407-413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 27]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
91.  Sands BE, Feagan, BG, Sandborn WJ, Shipitofsky N, Marko M, Sheng S, Johanns J, Germinaro M, Vetter M, Panés J. OP36 Efficacy and safety of combination induction therapy with guselkumab and golimumab in participants with moderately-to-severely active Ulcerative Colitis: Results through week 12 of a phase 2a randomized, double-blind, active-controlled, parallel-group, multicenter, proof-of-concept study. J Crohns Colitis. 2022;16: i042-i043.  [PubMed]  [DOI]  [Cited in This Article: ]
92.  Efficacy and Safety of Combination Induction Therapy With Guselkumab and Golimumab in Participants With Moderately to Severely Active Ulcerative Colitis: Results Through Week 12 of a Phase 2a Randomized, Double-Blind, Active-Controlled, Parallel-Group, Multicenter, Proof-of-Concept Study. Gastroenterol Hepatol (N Y). 2022;18:9-10.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Stalgis C, Deepak P, Mehandru S, Colombel JF. Rational Combination Therapy to Overcome the Plateau of Drug Efficacy in Inflammatory Bowel Disease. Gastroenterology. 2021;161:394-399.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
94.  Verstockt B, Parkes M, Lee JC. How Do We Predict a Patient's Disease Course and Whether They Will Respond to Specific Treatments? Gastroenterology. 2022;162:1383-1395.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 27]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
95.  Noor NM, Sousa P, Paul S, Roblin X. Early Diagnosis, Early Stratification, and Early Intervention to Deliver Precision Medicine in IBD. Inflamm Bowel Dis. 2022;28:1254-1264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
96.  Alsoud D, Verstockt B, Fiocchi C, Vermeire S. Breaking the therapeutic ceiling in drug development in ulcerative colitis. Lancet Gastroenterol Hepatol. 2021;6:589-595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 51]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
97.  Buer LCT, Høivik ML, Warren DJ, Medhus AW, Moum BA. Combining Anti-TNF-α and Vedolizumab in the Treatment of Inflammatory Bowel Disease: A Case Series. Inflamm Bowel Dis. 2018;24:997-1004.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
98.  Olbjørn C, Rove JB, Jahnsen J. Combination of Biological Agents in Moderate to Severe Pediatric Inflammatory Bowel Disease: A Case Series and Review of the Literature. Paediatr Drugs. 2020;22:409-416.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 28]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
99.  Kwapisz L, Raffals LE, Bruining DH, Pardi DS, Tremaine WJ, Kane SV, Papadakis KA, Coelho-Prabhu N, Kisiel JB, Heron V, Faubion WA, Loftus EV Jr. Combination Biologic Therapy in Inflammatory Bowel Disease: Experience From a Tertiary Care Center. Clin Gastroenterol Hepatol. 2021;19:616-617.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 49]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
100.  Yang E, Panaccione N, Whitmire N, Dulai PS, Vande Casteele N, Singh S, Boland BS, Collins A, Sandborn WJ, Panaccione R, Battat R. Efficacy and safety of simultaneous treatment with two biologic medications in refractory Crohn's disease. Aliment Pharmacol Ther. 2020;51:1031-1038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 72]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
101.  Glassner K, Oglat A, Duran A, Koduru P, Perry C, Wilhite A, Abraham BP. The use of combination biological or small molecule therapy in inflammatory bowel disease: A retrospective cohort study. J Dig Dis. 2020;21:264-271.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
102.  Privitera G, Onali S, Pugliese D, Renna S, Savarino E, Viola A, Ribaldone DG, Buda A, Bezzio C, Fiorino G, Fantini MC, Scaldaferri F, Guidi L, Danese S, Gasbarrini A, Orlando A, Armuzzi A. Dual Targeted Therapy: a possible option for the management of refractory Inflammatory Bowel Disease. J Crohns Colitis. 2020;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 20]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
103.  Dolinger MT, Spencer EA, Lai J, Dunkin D, Dubinsky MC. Dual Biologic and Small Molecule Therapy for the Treatment of Refractory Pediatric Inflammatory Bowel Disease. Inflamm Bowel Dis. 2021;27:1210-1214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 39]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
104.  Goessens L, Colombel JF, Outtier A, Ferrante M, Sabino J, Judge C, Saeidi R, Rabbitt L, Armuzzi A, Domenech E, Michalopoulos G, Cremer A, García-Alonso FJ, Molnar T, Karmiris K, Gecse K, Van Oostrom J, Löwenberg M, Farkas K, Atreya R, Ribaldone DG, Selinger C, Hoentjen F, Bihin B, Sebastian S; European COMBIO study group, Rahier JF. Safety and efficacy of combining biologics or small molecules for inflammatory bowel disease or immune-mediated inflammatory diseases: A European retrospective observational study. United European Gastroenterol J. 2021;9:1136-1147.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]