Published online Mar 20, 2025. doi: 10.5493/wjem.v15.i1.97404
Revised: October 9, 2024
Accepted: November 14, 2024
Published online: March 20, 2025
Processing time: 209 Days and 23.8 Hours
Inflammatory bowel disease (IBD) represents a significant disease burden marked by chronic inflammation and complications that adversely affect patients’ quality of life. Effective diagnostic strategies involve clinical assessments, endoscopic evaluations, imaging studies, and biomarker testing, where early diagnosis is essential for effective management and prevention of long-term complications, highlighting the need for continual advancements in diagnostic methods. The intricate interplay between genetic factors and the outcomes of biological therapy is of critical importance. Unraveling the genetic determinants that influence responses and failures to biological therapy holds significant promise for optimizing treatment strategies for patients with IBD on biologics. Through an in-depth examination of current literature, this review article synthesizes critical genetic markers associated with therapeutic efficacy and resistance in IBD. Under
Core Tip: Understanding the genetic factors that influence the response and failure of biological therapy in inflammatory bowel disease (IBD) is crucial for optimizing treatment strategies. Identifying specific genetic markers can help predict patient outcomes, tailor personalized therapies, and improve efficacy while minimizing adverse effects. This approach enhances clinical decision-making, leading to better management of IBD and improved patient quality of life. Future research should focus on expanding genetic profiling to refine therapeutic interventions.
- Citation: Peruhova M, Stoyanova D, Miteva DG, Kitanova M, Mirchev MB, Velikova T. Genetic factors that predict response and failure of biologic therapy in inflammatory bowel disease. World J Exp Med 2025; 15(1): 97404
- URL: https://www.wjgnet.com/2220-315x/full/v15/i1/97404.htm
- DOI: https://dx.doi.org/10.5493/wjem.v15.i1.97404
Inflammatory bowel disease (IBD) is a chronic inflammatory condition that comprises two entities: ulcerative colitis (UC) and Crohn’s disease (CD). The inflammation in UC continuously affects the colonic mucosa, with no granulomas detected on biopsy[1]. On the other hand, CD is characterized by transmural inflammation and granulomas that can affect any part of the gastrointestinal tract, most commonly the terminal ileum[2]. IBD is considered one of the most frequently diagnosed gastrointestinal diseases, with its incidence and prevalence constantly rising since the second half of the 20th century. This is true for both Europe and North America, as well as the newly industrialized countries of Asia, Africa, and South America. The highest incidence of 505 UC cases and 322 CD cases per 100000 persons has been reported in Norway and Germany, respectively[2]. IBD undoubtedly impairs quality of life, with fatigue, lack of energy, and sleep disturbances being the most common complaints. This is predominantly encountered in women, in patients suffering from CD, and in materially deprived persons[3]. IBD poses a significant disease burden characterized by chronic inflammation, pain, and complications that can severely impact patients' quality of life. Effective diagnostic strategies for IBD include clinical assessments, endoscopic evaluations, imaging studies, and biomarker testing to identify and differentiate the disease accurately. Early diagnosis is crucial for managing IBD effectively and preventing long-term complications, emphasizing the need for ongoing advancements in diagnostic approaches[3].
The treatment of IBD includes conventional therapy with 5-American Society of Anesthesiologists, corticosteroids and non-targeted immunosuppressants, and biological therapy. The biological rationale for using biologics in IBD is based on the known aspects of the disease pathophysiology. In patients with IBD, dysregulation of the immune response leads to the infiltration and accumulation of immune cells, which stimulate the release of various cytokines, chemokines, and growth factors[2]. This cascade may further impact the inflammation and carcinogenesis processes. Immune cells such as regulatory T cells (Tregs), type 2 macrophages, CD4+ T helper 17 cells, CD8+ T cells, and natural killer cells can play roles in either sustaining inflammation in IBD or contributing to disease progression[2].
Traditional gold standard methods for diagnosing IBD, such as endoscopy and histological examination, provide critical insights into mucosal inflammation and tissue morphology but can be invasive and uncomfortable for patients. By contrast, advanced techniques such as capsule endoscopy and biomarkers (e.g., fecal, serum, genetic) offer non-invasive alternatives that enhance patient comfort and convenience[4]. While capsule endoscopy allows visualization of the entire small intestine, fecal calprotectin (FC) testing enables the quick assessment of inflammation levels. Each approach has its advantages and limitations, and the choice of diagnostic method should be tailored to individual patient needs and clinical scenarios to ensure accurate and effective diagnosis[4,5].
Conventional therapy of IBD can induce a clinical response and maintain remission mainly in mild to moderate forms[4,5]. However, a recent meta-analysis showed a modest effect in terms of both induction and maintenance of remission in moderate to severe IBD[6]. Hence, biological therapy emerged as a new class of drugs with the potential to influence treatment failure with conventional therapy. The initial drugs, infliximab and adalimumab, showed excellent clinical and endoscopic efficacy. Still, the subsequent follow-up of patients revealed up to 30% response failure, 50% loss of response over time, and 10% surgical treatment requirement[7].
The new drugs available on the market also show incomplete responses. Vedolizumab, an anti-integrin antibody, achieved endoscopic improvement and remission in 51% and 29% of patients with UC in week 52, respectively. In CD, the same treatment goals were observed in 76% and 48% of cases, respectively[8]. For ustekinumab, an anti-interleukin (IL) 12/23 antibody, clinical response and remission at 1 year were seen in 76.8% and 50.6% of patients with UC, respectively[9]. For CD, the percentage of patients in clinical remission in week 44 was approximately 50%[10]. The non-selective Janus kinase (JAK) inhibitor, tofacitinib, which has only been approved for the treatment of UC, achieved clinical remission in 40.6% of cases in week 52[11]. In comparison, the clinical and endoscopic remission rates of the selective JAK1 inhibitor upadacitinib were 33% and 15% for UC vs 41% and 24% for CD, respectively[12]. In keeping with those mentioned above, there seems to be wide interindividual variation in the efficacy of biological treatment, which can be genetically determined. A recent systematic review published in 2024 by Plaza et al[13] showed that single nucleotide polymorphisms (SNPs) may be associated with a different treatment response towards anti-tumor necrosis factor (TNF), anti-integrins, and anti-IL-12/23 inhibitors. Therefore, a more individualized approach is needed for every patient with IBD.
In this review article, we identify and analyze genetic factors that predict patient response and failure to biologic therapy in IBD, facilitating personalized treatment strategies and improving clinical outcomes for the patients.
A comprehensive literature search was conducted across multiple databases, including PubMed, MEDLINE, Scopus, Web of Science, and Google Scholar, covering the period to May 2024. The search terms used were combinations of key words and Boolean operators: “Inflammatory Bowel Disease” AND (“Genetic Predictors” OR “Genetic Markers”) AND (“Biologic Therapy” OR “Biologic Therapy Response” OR “Biologic Therapy Failure”) AND (“Pharmacogenomics” OR “Genetic Variability”) AND “IBD Treatment”. Approximately 500 papers were retrieved, and relevant articles were selected based on their relevance to the topic, focusing on studies that explored the genetic factors influencing the efficacy and failure of biological therapies in IBD.
Cytochrome P450 (CYP) enzymes are involved in drug metabolism, and genetic polymorphisms in CYP2C19 significantly influence drug metabolism. Variations can result in different enzyme activity levels, categorizing individuals into poor, intermediate, extensive, and ultra-rapid metabolizers. This impacts drug efficacy and safety. For example, poor metabolizers may have higher drug levels, increasing the effectiveness or risk of toxicity for medications metabolized by CYP2C19[14]. Variations in CYP3A4 also affect the metabolism of many drugs used in IBD, contributing to variability in treatment outcomes[15].
Regarding the impact on drug levels and efficacy, thiopurine S-methyltransferase (TPMT) polymorphisms influence the metabolism of thiopurines. Low TPMT activity leads to higher active metabolite levels, increasing efficacy but also the risk of toxicity. TPMT genotyping helps tailor dosing to improve outcomes and reduce side effects[16]. Similarly, N-acetyltransferase 2 polymorphisms can affect the metabolism of certain IBD drugs, impacting their levels and effectiveness[17].
The relevance of genetic variations in drug targets is described for several genes. Polymorphisms in TNF receptors (TNF receptor superfamily member 1A [TNFRSF1A] and TNFRSF1B) can affect the binding and efficacy of anti-TNF therapies such as infliximab and adalimumab. Specific polymorphisms are associated with better or worse responses to these treatments[18].
Variations in the IL-23 receptor (IL-23R) gene influence responses to biologics targeting the IL-23 pathway, such as ustekinumab. Specific IL-23R genotypes are linked to improved treatment responses[19].
When we discuss the implications for treatment outcomes, we should focus on the human leukocyte antigen (HLA)-DQA1*05 allele associated with developing anti-drug antibodies (ADA) against infliximab and adalimumab, reducing their efficacy. Patients with this allele may need closer monitoring and therapy adjustment[20]. Variants in Fc gamma receptor 3A (FCGR3A) can also affect the response to anti-TNF agents by altering drug binding to immune cells, impa
To date, more than 240 non-overlapping genetic loci have been identified as significant risk factors of IBD[21-23]. Among them statistically significant genes include autophagy-related 16-like 1 (ATG16L1), E-cadherin, HLA, hepatocyte nuclear factor 4 alpha, IL-10, IL-10RA, IL-10RB, IL-23R, leucine-rich repeat kinase 2, nucleotide oligomerization domain 2 (NOD2), protein tyrosine phosphatase non-receptor type 2, TNF superfamily member 15, immunity-related GTPase family M, caspase recruitment domain protein 9, and RING finger protein 186, which are linked to innate and adaptive immunity, autophagy, epithelial barrier, innate mucosal defense, Tregs, oxidative stress, IL-10 and IL-23 signaling, and cell apoptosis, among others[22-24].
First, we start by describing the genetic mutations in drug targets. A critical point in IBD treatment is identifying genetic variants associated with an individual's drug response. Mutations in genes encoding drug targets can significantly impact drug efficacy and contribute to treatment resistance. IBD-causing alleles are rich in non-synonymous mutations in their coding region, modulating the protein structure and function and thus affecting drug binding affinity or downstream signaling pathways. Furthermore, 80%-90% of IDB loci are non-synonymous variants due to mutations in their non-coding regions exerting pathogenic effects by modulating the gene expression[25].
Extensive meta-analyses combined with genome-wide association studies (GWAS) have identified specific variants and polymorphisms associated not only with the onset and severity of IBD but also with a role in treatment response in patients undergoing drug therapy[26]. The clinical trials and real-life practice demonstrate the association of some genetic variants with no or limited response (primary non-response or secondary loss of response) to drug treatment[21,26,27] and, in some cases, even worsen it[28].
Genetic variants leading to altered target structure or gene expression are not the only cause of reduced drug effectiveness, as additional causes include drug interactions, development of resistance, and drug quality. For example, changes in physiological conditions, such as the potential of hydrogen and blood flow, can influence drug distribution and metabolism, affecting drug efficacy. The use of multiple drugs can lead to the inhibition or induction of drug-metabolizing enzymes, competition for binding sites, or synergistic or antagonistic effects on drug targets[29].
Some biological drugs can induce immune reactions, leading to the formation of antibodies that neutralize the therapeutic effects of the drug. Immunogenicity with the formation of ADA to biological products is one of the causes of treatment failure in IBDs. The drug concentration, inadequate drug exposure, and high drug clearance can also be responsible for undesirable therapeutic outcomes in patients with IBD. Other factors besides immunogenicity can accelerate the clearance of biologics such as increased body weight, low serum albumin, and even disease status and medications[30]. Biologics have long been used to treat IBD, but guidelines regarding their optimal use are still being researched and developed. Over the past few decades, IBD-related costs have significantly increased due to the frequent administration of TNF-α antagonists and other biological products for treatment[31]. This gives reason to assume that the optimal use of these products is essential to improve the efficacy of therapy and reduce adverse effects.
Various strategies to prevent ADA formation have been investigated. Combining a biological product with an immunomodulator was found to preclude the formation of ADA[32,33]. ADAs decrease by adding or changing immu
The TNF-α antagonists infliximab, adalimumab, golimumab, and certolizumab pegol are used as anti-TNF therapies in the clinical setting of IBD[37,38]. They have different pharmacological profiles and efficacy and can improve remission[39]. However, some patients with IBD either do not respond or have loss of response to treatment over time. Genetic factors are responsible for this inability. Genetic profiling techniques and GWAS have enabled the identification of genetic variants that can influence the treatment response and development of adverse effects[24].
А study revealed genetic associations with primary non-response[40]. The authors found that SNPs in loci DENN domain containing 1B (rs2488397) and aryl hydrocarbon receptor (rs1077773) are most strongly associated with primary non-response. Similarly, they observed genetic associations with time to loss of response. In addition to a number of known IBD susceptibility loci, SNPs in PR domain zinc finger protein 1 (rs62421049), chromosome 21q22.2 (rs2836866), cluster of differentiation 28 (rs3116494), SMAD3 (rs17293632), and interferon (IFN) induced with helicase C domain 1 (rs1990760) were associated[40].
Usually, SNPs with long-term responses are associated with responsiveness to infliximab or adalimumab therapy in patients with IBD. However, some SNPs such as rs396991-GG (FCGR3A), rs6100556-TT (phosphatase and actin regulator 3 [PHACTR3]), rs2241880-AA, rs10210302-CC, and rs2241880-GG (ATG16L1) have shown a reduced clinical response at the end of treatment in pediatric patients with IBD (pIDB)[27,41].
In some cases, the presence of certain variants in the genotype of patients with IDB may worsen the drug treatment. In their research, Zapata-Cobo et al[28] demonstrated that specific SNPs such as rs6908425 (cyclin-dependent kinase 5 regulatory subunit-associated protein 1-like 1), rs2241880 (ATG1L1), rs2188962 (IFN regulatory factor 1 antisense RNA 1), and rs6100556 (PHACTR3) were associated with long-term worse response to anti-TNF drugs in children with IBD.
SNPs with short-term responses do not show a response to drug treatment. For example, rs976881-AA+GA (TNFRSF
The International Organization for the Study of IBD STRIDE-II recommendations from 2021 confirmed that the most critical long-term achievable treatment targets for patients with IBD are clinical remission, endoscopic healing, restoration of quality of life, and absence of disability. With accumulating clinical evidence, serum and fecal biomarkers have been validated as intermediate- or medium-term feasible treatment goals, meaning that at times, treatment could be revisited solely based on these tests to facilitate care in the clinical setting[42].
Based on their low cost and availability, FC and C-reactive protein (CRP) are the two most widely used classical biomarkers in IBD. A meta-analysis that summarized the performance of FC when using all available data, whatever the cutoff values used, showed a pooled sensitivity of 82%, specificity of 72%, and area under the curve of 0.84 for FC in reflecting endoscopic disease activity in CD[43]. The evidence suggests that a reduction in FC and target below a certain threshold have clear prognostic significance, justifying the utilization of this biomarker as a treatment target. FC predicted long-term clinical outcomes when measured 12 weeks after initiating medical treatment[44]. A meta-analysis of six studies showed that patients with elevated FC had a 53% to 83% probability of relapse during the subsequent 2 months to 3 months[45]. Different studies that confirmed FC as a predictor of relapse at the time of anti-TNF discontinuation, predicted subsequent relapse at cutoff values of 50 mg/g to 150 mg/g[46].
Whereas FC has high sensitivity and lower specificity in identifying mucosal inflammation, CRP has the opposite characteristics of higher specificity but low sensitivity[47]. Consequently, high CRP values determined at the time of anti-TNF discontinuation are associated with a higher risk of relapse[48]. CRP normalization at 8 weeks to 14 weeks after treatment predicts remission at 1 year with vedolizumab[49] and anti-TNF success at 2 years[50]. Similarly, CRP > 5 mg/dL in week 22 has been shown to predict secondary loss of response to anti-TNF[51].
Although newer biomarkers are emerging and being tested in practice, none have been successfully validated or proved reliable as a sole predictive tool in personalized medicine in IBD.
Identifying predictive genetic markers is the first step in predicting the response to IBD biological treatment. Although genetic studies are not universally applicable in the clinical field, they can be used to diagnose IBD, predict therapeutic or toxic responses to drugs, and assess the risk, thereby enabling precision medicine for patients. Precisely understanding the underlying immunopathogenic mechanisms of IBD will lead to the development of targeted therapies. Effective and careful consideration of underlying factors, including immunogenicity potential, treatment safety profile, and optimal therapeutic duration in these patients, is needed.
Over the past decade, a range of predictive biomarkers has been identified that promise to provide personalized and effective treatments for patients[52]. There are also some limitations and clinical applications of these biomarkers in monitoring optimized patient outcomes and providing personalized care. Some of the predictive genetic markers are associated with predicting the response to biological treatment in patients with IBD. A favorable clinical response may be associated with polymorphisms in genes such as FCGR3A, TNFRSF1A, IL-6, and IL-1B; conversely, variants of Toll-like receptor 2 (TLR2) and TLR9 show a negative correlation[53].
Some genetic variants in TNFRSF1B and nuclear factor kappa B (NF-kB) genes can affect TNF-α production or the binding of TNF-α blockers to the TNF-α receptor. These, in turn, may influence the primary response to anti-TNF therapy in patients with CD[54] or UC[55].
Polymorphisms in the IL-23R are associated with response in patients with UC, and a polymorphism in the NOD2/CARD15 gene is associated with patients with CD[56]. These data[54] and the genetic variants described above are related to predicting positive, negative, or no response to biological treatment in patients with IBD[55-59].
It is also mandatory to integrate clinical and other biomarkers into clinical practice. Protein markers can provide valuable information for monitoring and therapeutic responses to anti-TNF therapy[60]. Markers such as CRP, human angiopoietin 1 (ANG1), ANG2, carcinoembryonic antigen-related cell adhesion molecule 1, extracellular matrix metalloproteinase inducer, transforming growth factor alpha, matrix metalloproteinases 1-3 (MMP 1-3), MMP-9, IL-6, and some apolipoproteins have been identified as predictive[61]. It should be noted that proteomics has great potential, but various factors influence protein levels and are individual in patients. Therefore, protеin markers alone are not sufficient to be universal markers of therapeutic response in IBD patients.
FC, lactoferrin, and other fecal biomarkers can also be used as potential markers in patients with CD and UC on anti-TNF therapy[62,63]; however, the results of the studies are contradictory. In some cases, high calprotectin levels correlated with better treatment response[64], others inversely[65] or failed to confirm the data[66,67].
The correlation between the gut microbiome and anti-TNF therapy is complex. Still, there is evidence that some microbial markers may be associated with treatment response[68]. Patients with a more diverse gut microbiome respond better to anti-TNF-α therapy, whereas the presence of other species is associated with a negative response[69,70]. In dysbiosis, there is often no or poor response to anti-TNF therapy[71,72]. A recent study by Caenepeel et al[73] investigated different combinations of clinical and microbial data to predict the efficacy of TNF-α treatment. The authors examined certain clinical parameters and microbial dysbiosis, achieving a 73.9% accuracy rate in predicting treatment responses.
Fungi and viruses are also being studied for their correlation with responses to therapy[74-76]. The diversity of these different populations cannot be completely ruled out as misleading in clinical practice, as their amount and types also depend on different factors. Recently, microRNAs (miRNAs) have also been considered potential biomarkers for therapeutic responses in patients with IBD[77].
One study found significant changes in the expression of several miRNAs after anti-TNF treatment in patients with pIDB[78], but another study did not confirm the correlations[79]. Additional studies on miRNAs as possible predictive markers in patients with IBD are needed. Changes in blood or mucosal parameters can also be assessed for anti-TNF therapy's effectiveness. If there is a reduction in TNF-α and IFN-g levels and reduced inflammation at the mucosal level, then anti-TNF therapy is effective[65,80]. Some cytokines have also shown potential as candidate biomarkers in patients with IBD[81].
Many identified biomarkers indicate inflammation and are not specific to IBD alone. Various factors such as age, sex, genetics, biochemical profile, microbial composition, and mucosal conditions influence the therapy response, which may explain why none of these biomarkers have been included in routine clinical practice. For this to happen, future efforts should focus on the robust validation of certain biomarkers in large numbers of patients with IBD.
It is a well-established fact that one-third of patients with IBD are primary non-responders to inceptive treatment despite new targeted therapies that have been available recently in clinical practice. Another troubling fact is that half of patients on therapy lose treatment response with time[82]. Presently, there is an insufficient number of biomarkers that can be useful in predicting treatment failure. In clinical practice, validating equitable biomarkers, which can predict treatment response or failure, would greatly help clinicians tailor personalized therapeutic algorithms for managing patients with IBD.
Personalized medicine is the idea that the appropriate medication may be given to the relevant patient at the proper time. This process could only be possible with precise knowledge of the underlying molecular processes causing IBD. Better rates of patient outcomes, reduction of morbidity due to improper treatment, and decline in healthcare costs would all be made possible by such an approach. Creating a personalized medicine approach in IBD is connected with identifying, developing, and validating novel biomarkers to support individualized treatment[83].
A study by Park and Jeen[22] established 240 susceptibility loci for IBD. Understanding the role of these genes in IBD pathogenesis will help to identify novel therapeutic targets. Recently, a plethora of data on the connection between genetic markers and therapeutic response has been published. For example, a study by Jürgens et al[56] demonstrated that therapeutic responses to infliximab were detected in adult patients with CD who were homozygous for the high-risk IL-23R variant compared to low-risk IL-23R variants. Unfortunately, a low percentage of patients have these IL-23R variants; thus, using this marker in clinical practice is unreliable[56].
The CASP9 gene regulates activation of the caspase cascade and the process of cell apoptosis. Thus, polymorphism in CASP9 could affect the process of apoptosis in peripheral blood lymphocytes in patients with IBD. It has been established that polymorphism in the CASP9 gene (rs4645983) is related to short-term non-response to adalimumab[84].
Some data have shown a better response to infliximab in patients with CD with polymorphisms in the CASP9 gene and FAS ligand gene[85]. According to some GWAS, there has been conflicting evidence about the relationship between treatment response and polymorphisms in TNF-encoding genes. Two polymorphisms in the TNF promoter were linked to the responsiveness of patients with IBD to TNF inhibition, according to a 2013 meta-analysis: More often occurring alleles were linked to higher response rates[86].
Better clinical responses have been found to be positively correlated with polymorphisms in the FCGR3A, TLR4, TNFRSF1A, IFN-g, IL-6, and IL-1B genes, whereas variants of TLR2 and TLR9 have shown a negative correlation[87]. In individuals with IBD receiving anti-TNF medication, polymorphisms in TNF, NF-kB, and other cytokine pathways have been connected to better outcomes. For instance, a study by Bank et al[88] demonstrated that in patients with IBD receiving anti-TNF therapy, polymorphisms in TNF, NF-kB, and other cytokine pathways were correlated with a better response to treatment.
A study by Koder et al[84] investigated SNPs in genes that regulate the cell division cycle (cyclin Y; rs12777960 CC), chromatin organization (chromosome 11 open reading frame 30; rs7927894 CC), and synthesis of some proinflammatory cytokines (IL-13; rs1295686 TT). The authors established that these SNPs are related to long-term response to adalimumab[84]. Furthermore, in patients with CD receiving anti-TNF therapy, the HLA-DQA1*05 allele, HLA-DRB1 allele, and polymorphisms at the FCGR3A locus (encoding immunoglobulin G Fc receptor IIIa) have been linked to a higher risk of ADA production[89-91]. Monoclonal antibodies represent large, complex proteins; they can lead to the synthesis of ADA, which are linked to therapy inefficacy. One such chimeric antibody is infliximab[92]. Finding patients at high risk of developing ADA would be very beneficial since concurrent immunosuppression (with thiopurines and methotrexate) lowers the likelihood of developing them[93].
IL-13R alpha 2 is another marker that has been previously discovered by gene array investigations in the mucosal biopsies of patients with IBD[94]. The biomarker, assessed as mRNA expression in the mucosa of patients with IBD before therapy, was recently found to be particularly predictive of the absence of response to anti-TNF in terms of mucosal healing at 6 months. The area under the curve for infliximab and adalimumab was 0.90 and 0.94, respectively, with P < 0.001[95].
The NOD2 gene, which encodes a protein involved in inducing the immune response and connected to the TNF-inflammatory pathway, is linked to both a more aggressive course of the disease and susceptibility to CD[96,97]. According to particular research, NOD2 mutations are linked to a poorer response to anti-TNF therapy[98-100]. In patients with CD receiving TNF antagonist treatment, polymorphisms in the ATG16L1 gene have been linked to improved response rates and prolonged benefits[84]. In actual therapeutic practice, the genetic variants that confer vulnerability to ADA development could be quite helpful in identifying individuals who could benefit from biological therapy. Few studies revealed certain genetic polymorphisms[101-105] and gene variants[106-110] associated with various responses to biological therapy in IBD.
An overview of the currently known genetic factors that influence the response to biological therapy for patients with IBD is presented in Table 1[28,41,53,56,58,84,86-91,98-110].
Genetic markers | Patients | Clinical consequences | Ref. |
Polymorphisms in TLR2, rs11938228, TLR4, TLR9, TNFRSF1A, IFNG, IL-6, and IL-1B (rs4848306, NOD-like receptor thermal protein domain associated protein 3, Janus kinase 2) | IBD | Clinical response in to anti-TNF | Bek et al[53], 2016; Salvador-Martín et al[87], 2019; Steenholdt et al[101], 2012; Medrano et al[102], 2013; Bank et al[88], 2014 |
Polymorphisms in TNF-α promoter (-308 A/G and -857 C/T) | IBD and spondylarthritis | Clinical response to anti-TNF | Tong et al[86], 2013; Song et al[103], 2015 |
Polymorphisms implicated in NF-kB pathway: TLR2, TLR4, TLR9, LY96 (MD-2), CD14, mitogen-activated protein kinase 14 (NIK), TNF-α, TNFRSF1A, TNFAIP3 (A20), IL-1B, IL-1RN, IL-6, IL-17A, IFNG | IBD | Clinical response to anti-TNF (adalimumab) | Bank et al[88], 2014; Song et al[103], 2015; Bek et al[53], 2016 |
Polymorphisms in IL-23R | UC | Early response to infliximab | Jürgens et al[56], 2010 |
HLA-DQA1*05 | CD | Development of ADA against infliximab and adalimumab | Sazonovs et al[89], 2020; Salvador-Martín et al[87], 2023 |
HLA-DRB1 | IBD | Development of ADA against infliximab | Billiet et al[90], 2015 |
Polymorphisms in FcγRIIIa | CD | Development of ADA against infliximab | Louis et al[91], 2004 |
Polymorphisms in NOD2 | CD | Clinical response to anti-TNF | Niess et al[98], 2012 |
Polymorphisms in NOD2 | CD | Loss of response to anti-TNF | Juanola et al[99], 2015 |
Polymorphisms in NOD2 | CD | Lower anti-TNF trough levels | Schäffler et al[100], 2018 |
Polymorphisms in ATG16L1 (C11orf30; rs7927894 CC, CCNY; rs12777960 CC) (rs10210302) | IBD and CD | Clinical response to adalimumab | Koder et al[84], 2015; Zapata-Cobo et al[28], 2023 |
Polymorphisms in FAS, FASL, and CASP9 (apoptotic pharmacogenetic index) | CD | Clinical response to infliximab and adalimumab | Hlavaty et al[104], 2007; Koder et al[84], 2015 |
Multiple polymorphisms (combined clinical-genetic model) | CD and UC | Short-term and long-term to clinical response anti-TNF | Barber et al[58], 2016; Burke et al[105], 2018 |
Polymorphisms in TNFSF4/18, perilipin 2, rs762787, rs9572250, rs144256942, rs523781 | IBD | Clinical response to anti-TNF | Wang et al[106], 2019 |
PHACTR3 (rs6100556) | UC and CD | Response to anti-TNF in children | Zapata-Cobo et al[28], 2023 |
CNTN5 | CD | Thomas et al[107], 2014 | |
FCGR3A | CD and UC | Antibody-dependent immune responses | Curci et al[41], 2021 |
PTGER4 (rs10512734) | CD | Response to adalimumab | Koder et al[84], 2015 |
IL-27 | CD | Response to adalimumab | Koder et al[84], 2015 |
NR12 | CD | Response to adalimumab | Koder et al[84], 2015 |
FASL (rs763110 ) | CD | Clinical response in patients with CD treated with infliximab | Zapata-Cobo et al[28], 2023; Steenholdt et al[101], 2012 |
IRF1-AS1 | UC | Response to anti-TNF in children | Zapata-Cobo et al[28], 2023 |
Polymorphisms in IL-1B | IBD | Response to anti-TNF-α | Bank et al[88], 2014 |
Polymorphisms in IL-18 | IBD | Response to anti-TNF-α | Bek et al[53], 2016 |
TLR2 (rs1816702 CC and rs3804099 TT) | Clinical response to infliximab | Salvador-Martín et al[87], 2019 | |
Polymorphisms in CXCL12 | IBD | Response to anti-TNF-α in children | Zapata-Cobo et al[28], 2023 |
Polymorphisms in IL-10 | IBD | Response to anti-TNF-α in children | Salvador-Martín et al[108], 2020; Salvador-Martín et al[109], 2023 |
Polymorphisms in IL-17 | IBD | Response to anti-TNF-α in children | Salvador-Martín et al[108], 2020; Salvador-Martín et al[109], 2023; Bank et al[88], 2014 |
Polymorphisms in IL-6 | IBD | Response to anti-TNF-α in children | Salvador-Martín et al[108], 2020 |
Gene protein tyrosine phosphatase non-receptor type 2 (rs7234029 AG + GG, CASP9) | IBD | Non-response to anti-TNF and ustekinumab | Hoffmann et al[110], 2021 |
Genomics and precision medicine advancements are revolutionizing the approach to treating IBD. Integrating multi-omics data, including genomics, transcriptomics, proteomics, and metabolomics, allows for a comprehensive understan
Artificial intelligence (AI) is crucial in predicting patient response to biological therapies. Machine learning algorithms can process and analyze large datasets to uncover patterns and predict outcomes based on genetic and clinical information. This predictive capability can significantly enhance clinical decision-making, allowing for selecting the most suitable biological therapy for each patient, thereby improving treatment efficacy and reducing the risk of adverse reactions[111,112].
The potential for novel therapeutic targets based on genetic insights is immense. Emerging therapies, such as those targeting specific genetic pathways implicated in IBD, are showing promise. For example, therapies designed to modulate the immune response or repair intestinal barrier function are developing based on genetic findings. The future landscape of IBD treatment will likely see a shift towards these targeted therapies, which offer the potential for improved patient outcomes and a reduction in the burden of disease. Continued research in this field is essential to fully realizing the benefits of precision medicine in IBD therapy.
In summary, understanding the genetic factors that influence the response and failure of biological therapy in IBD is crucial for advancing treatment approaches. Key genetic factors, such as specific gene polymorphisms, mutations, and epigenetic modifications, play significant roles in determining how patients respond to biological therapies. Identifying these genetic markers enables a more precise prediction of treatment outcomes, paving the way for personalized medicine. Integrating multi-omics data and the application of AI in this field are poised to revolutionize IBD treatment. These advancements will allow for the development of novel therapeutic targets and the optimization of existing treatments, ultimately improving patient outcomes. As we move towards a future where treatment plans are tailored to the individual genetic makeup of patients, the potential for reducing the burden of IBD and enhancing the quality of life for patients is immense. However, the implications for personalized medicine are profound. By leveraging genetic insights, healthcare providers can offer more targeted and effective therapies, minimizing adverse effects and maximizing therapeutic benefits. Continued research and technological advancements will be essential to fully harness the potential of precision medicine in IBD treatment, transforming the clinical management of this complex disease.
1. | Magro F, Gionchetti P, Eliakim R, Ardizzone S, Armuzzi A, Barreiro-de Acosta M, Burisch J, Gecse KB, Hart AL, Hindryckx P, Langner C, Limdi JK, Pellino G, Zagórowicz E, Raine T, Harbord M, Rieder F; European Crohn’s and Colitis Organisation [ECCO]. Third European Evidence-based Consensus on Diagnosis and Management of Ulcerative Colitis. Part 1: Definitions, Diagnosis, Extra-intestinal Manifestations, Pregnancy, Cancer Surveillance, Surgery, and Ileo-anal Pouch Disorders. J Crohns Colitis. 2017;11:649-670. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1024] [Cited by in F6Publishing: 1198] [Article Influence: 171.1] [Reference Citation Analysis (0)] |
2. | Guan Q. A Comprehensive Review and Update on the Pathogenesis of Inflammatory Bowel Disease. J Immunol Res. 2019;2019:7247238. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 227] [Cited by in F6Publishing: 522] [Article Influence: 104.4] [Reference Citation Analysis (0)] |
3. | Rubin GP, Hungin AP, Chinn DJ, Dwarakanath D. Quality of life in patients with established inflammatory bowel disease: a UK general practice survey. Aliment Pharmacol Ther. 2004;19:529-535. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 62] [Cited by in F6Publishing: 47] [Article Influence: 2.4] [Reference Citation Analysis (0)] |
4. | Kühbacher T, Schreiber S, Fölsch UR. Ulcerative colitis: conservative management and long-term effects. Langenbecks Arch Surg. 2004;389:350-353. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Cited by in F6Publishing: 5] [Article Influence: 0.3] [Reference Citation Analysis (0)] |
5. | Sandborn WJ, Feagan BG, Lichtenstein GR. Medical management of mild to moderate Crohn's disease: evidence-based treatment algorithms for induction and maintenance of remission. Aliment Pharmacol Ther. 2007;26:987-1003. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 75] [Cited by in F6Publishing: 67] [Article Influence: 3.9] [Reference Citation Analysis (0)] |
6. | Damião AOMC, de Azevedo MFC, Carlos AS, Wada MY, Silva TVM, Feitosa FC. Conventional therapy for moderate to severe inflammatory bowel disease: A systematic literature review. World J Gastroenterol. 2019;25:1142-1157. [PubMed] [DOI] [Cited in This Article: ] [Cited by in CrossRef: 48] [Cited by in F6Publishing: 54] [Article Influence: 10.8] [Reference Citation Analysis (0)] |
7. | Fanizza J, D'Amico F, Lusetti F, Fasulo E, Allocca M, Furfaro F, Zilli A, Parigi TL, Radice S, Peyrin-Biroulet L, Danese S, Fiorino G. The Role of IL-23 Inhibitors in Crohn's Disease. J Clin Med. 2023;13:224. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
8. | Moens A, Verstockt B, Alsoud D, Sabino J, Ferrante M, Vermeire S. Translating Results from VARSITY to Real World: Adalimumab vs Vedolizumab as First-line Biological in Moderate to Severe IBD. Inflamm Bowel Dis. 2022;28:1135-1142. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Cited by in F6Publishing: 4] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
9. | Taxonera C, Olivares D, López-García ON, Alba C. Meta-analysis: Real-world effectiveness and safety of ustekinumab in patients with ulcerative colitis. Aliment Pharmacol Ther. 2023;57:610-619. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 6] [Reference Citation Analysis (0)] |
10. | Feagan BG, Sandborn WJ, Gasink C, Jacobstein D, Lang Y, Friedman JR, Blank MA, Johanns J, Gao LL, Miao Y, Adedokun OJ, Sands BE, Hanauer SB, Vermeire S, Targan S, Ghosh S, de Villiers WJ, Colombel JF, Tulassay Z, Seidler U, Salzberg BA, Desreumaux P, Lee SD, Loftus EV Jr, Dieleman LA, Katz S, Rutgeerts P; UNITI–IM-UNITI Study Group. Ustekinumab as Induction and Maintenance Therapy for Crohn's Disease. N Engl J Med. 2016;375:1946-1960. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 1053] [Cited by in F6Publishing: 1246] [Article Influence: 155.8] [Reference Citation Analysis (0)] |
11. | Salas A, Hernandez-Rocha C, Duijvestein M, Faubion W, McGovern D, Vermeire S, Vetrano S, Vande Casteele N. JAK-STAT pathway targeting for the treatment of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2020;17:323-337. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 188] [Cited by in F6Publishing: 363] [Article Influence: 90.8] [Reference Citation Analysis (0)] |
12. | Zheng DY, Wang YN, Huang YH, Jiang M, Dai C. Effectiveness and safety of upadacitinib for inflammatory bowel disease: A systematic review and meta-analysis of RCT and real-world observational studies. Int Immunopharmacol. 2024;126:111229. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
13. | Plaza J, Mínguez A, Bastida G, Marqués R, Nos P, Poveda JL, Moret-Tatay I. Genetic Variants Associated with Biological Treatment Response in Inflammatory Bowel Disease: A Systematic Review. Int J Mol Sci. 2024;25:3717. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
14. | Shubbar Q, Alchakee A, Issa KW, Adi AJ, Shorbagi AI, Saber-Ayad M. From genes to drugs: CYP2C19 and pharmacogenetics in clinical practice. Front Pharmacol. 2024;15:1326776. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
15. | Busti AJ. Genetic Polymorphisms of the CYP3A4 Enzyme and Potential Influence on Drug Efficacy and/or Safety. 2015. Available from: https://www.ebmconsult.com/articles/genetic-polymorphisms-cytochrome-p450-cyp3a4-enzyme. [Cited in This Article: ] |
16. | Warner B, Johnston E, Arenas-Hernandez M, Marinaki A, Irving P, Sanderson J. A practical guide to thiopurine prescribing and monitoring in IBD. Frontline Gastroenterol. 2018;9:10-15. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 38] [Cited by in F6Publishing: 52] [Article Influence: 8.7] [Reference Citation Analysis (0)] |
17. | Tomalik-Scharte D, Lazar A, Fuhr U, Kirchheiner J. The clinical role of genetic polymorphisms in drug-metabolizing enzymes. Pharmacogenomics J. 2008;8:4-15. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 98] [Cited by in F6Publishing: 101] [Article Influence: 5.9] [Reference Citation Analysis (0)] |
18. | Kumar M, Murugesan S, Ibrahim N, Elawad M, Al Khodor S. Predictive biomarkers for anti-TNF alpha therapy in IBD patients. J Transl Med. 2024;22:284. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 4] [Reference Citation Analysis (0)] |
19. | Siakavellas SI, Bamias G. Role of the IL-23/IL-17 axis in Crohn's disease. Discov Med. 2012;14:253-262. [PubMed] [Cited in This Article: ] |
20. | Guardiola Capón J, Iborra M, Padró A, de la Peña L, Serra K, Martin-arranz MD, Domènech E, Fernandez A, Mesonero F, Gonzalez-muñoza C, Ferreiro-iglesias R, Navarro P, Martín-cardona A, Sicilia B, Sierra-ausin M, Calvet X, Marquez L, de Francisco R, Cañete F, Gutierrez A, García-lópez S, Rivero M, Hinojosa J, Iglesias-flores E, Nos P, Riestra S, Bosca-watts M, Zabana Y, Castro B, Barreiro M, Garcia-planella E, Ricart E, De Francisco R, Suris G, Ruiz-cerulla A, Rodriguez-alonso L, Orobitg J, Rodríguez-moranta F. OP37 Effect of the HLA-DQA1*05 allele on the efficacy of ustekinumab in patients with Crohn's Disease. Multicenter study based on the ENEIDA registry of GETECCU. J Crohns Colitis. 2024;18:i67-i67. [DOI] [Cited in This Article: ] |
21. | Privitera G, Pugliese D, Rapaccini GL, Gasbarrini A, Armuzzi A, Guidi L. Predictors and Early Markers of Response to Biological Therapies in Inflammatory Bowel Diseases. J Clin Med. 2021;10:853. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 8] [Cited by in F6Publishing: 19] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
22. | Park SC, Jeen YT. Genetic Studies of Inflammatory Bowel Disease-Focusing on Asian Patients. Cells. 2019;8:404. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 28] [Cited by in F6Publishing: 44] [Article Influence: 8.8] [Reference Citation Analysis (0)] |
23. | Antunes JC, Seabra CL, Domingues JM, Teixeira MO, Nunes C, Costa-Lima SA, Homem NC, Reis S, Amorim MTP, Felgueiras HP. Drug Targeting of Inflammatory Bowel Diseases by Biomolecules. Nanomaterials (Basel). 2021;11:2035. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 6] [Cited by in F6Publishing: 12] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
24. | Lauro R, Mannino F, Irrera N, Squadrito F, Altavilla D, Squadrito G, Pallio G, Bitto A. Pharmacogenetics of Biological Agents Used in Inflammatory Bowel Disease: A Systematic Review. Biomedicines. 2021;9:1748. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 16] [Cited by in F6Publishing: 17] [Article Influence: 5.7] [Reference Citation Analysis (0)] |
25. | McGovern DP, Kugathasan S, Cho JH. Genetics of Inflammatory Bowel Diseases. Gastroenterology. 2015;149:1163-1176. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 250] [Cited by in F6Publishing: 271] [Article Influence: 30.1] [Reference Citation Analysis (1)] |
26. | Puca P, Capobianco I, Coppola G, Di Vincenzo F, Trapani V, Petito V, Laterza L, Pugliese D, Lopetuso LR, Scaldaferri F. Cellular and Molecular Determinants of Biologic Drugs Resistance and Therapeutic Failure in Inflammatory Bowel Disease. Int J Mol Sci. 2024;25:2789. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
27. | Ye BD, McGovern DP. Genetic variation in IBD: progress, clues to pathogenesis and possible clinical utility. Expert Rev Clin Immunol. 2016;12:1091-1107. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 67] [Cited by in F6Publishing: 59] [Article Influence: 7.4] [Reference Citation Analysis (0)] |
28. | Zapata-Cobo P, Salvador-Martín S, Velasco M, Palomino LM, Clemente S, Segarra O, Moreno-Álvarez A, Fernández-Lorenzo A, Pérez-Moneo B, Montraveta M, Sánchez C, Tolín M, Loverdos I, Fobelo MJ, Navas-López VM, Magallares L, García-Romero R, Sánchez-Hernández JG, Rodríguez A, Bossacoma F, Balboa MJ, Salcedo E, Sanjurjo-Sáez M, López-Fernández LA. Polymorphisms indicating risk of inflammatory bowel disease or antigenicity to anti-TNF drugs as biomarkers of response in children. Pharmacol Res. 2023;194:106859. [PubMed] [DOI] [Cited in This Article: ] [Reference Citation Analysis (0)] |
29. | Bechtold B, Clarke J. Multi-factorial pharmacokinetic interactions: unraveling complexities in precision drug therapy. Expert Opin Drug Metab Toxicol. 2021;17:397-412. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 4] [Cited by in F6Publishing: 7] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
30. | Deyhim T, Cheifetz AS, Papamichael K. Drug Clearance in Patients with Inflammatory Bowel Disease Treated with Biologics. J Clin Med. 2023;12:7132. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Cited by in F6Publishing: 2] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
31. | Bots SJA, Hoekman DR, Benninga MA, Ponsioen CY, D'Haens GR, Löwenberg M. Patterns of anti-TNF use and associated treatment outcomes in inflammatory bowel disease patients: results from an analysis of Dutch health insurance claims data. Neth J Med. 2017;75:432-442. [PubMed] [Cited in This Article: ] |
32. | Colombel JF, Sandborn WJ, Reinisch W, Mantzaris GJ, Kornbluth A, Rachmilewitz D, Lichtiger S, D'Haens G, Diamond RH, Broussard DL, Tang KL, van der Woude CJ, Rutgeerts P; SONIC Study Group. Infliximab, azathioprine, or combination therapy for Crohn's disease. N Engl J Med. 2010;362:1383-1395. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2221] [Cited by in F6Publishing: 2273] [Article Influence: 162.4] [Reference Citation Analysis (0)] |
33. | Panaccione R, Ghosh S, Middleton S, Márquez JR, Scott BB, Flint L, van Hoogstraten HJ, Chen AC, Zheng H, Danese S, Rutgeerts P. Combination therapy with infliximab and azathioprine is superior to monotherapy with either agent in ulcerative colitis. Gastroenterology. 2014;146:392-400. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 614] [Cited by in F6Publishing: 648] [Article Influence: 64.8] [Reference Citation Analysis (0)] |
34. | Ben-Horin S, Waterman M, Kopylov U, Yavzori M, Picard O, Fudim E, Awadie H, Weiss B, Chowers Y. Addition of an immunomodulator to infliximab therapy eliminates antidrug antibodies in serum and restores clinical response of patients with inflammatory bowel disease. Clin Gastroenterol Hepatol. 2013;11:444-447. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 243] [Cited by in F6Publishing: 240] [Article Influence: 21.8] [Reference Citation Analysis (0)] |
35. | Strik AS, van den Brink GR, Ponsioen C, Mathot R, Löwenberg M, D'Haens GR. Suppression of anti-drug antibodies to infliximab or adalimumab with the addition of an immunomodulator in patients with inflammatory bowel disease. Aliment Pharmacol Ther. 2017;45:1128-1134. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 88] [Cited by in F6Publishing: 99] [Article Influence: 14.1] [Reference Citation Analysis (0)] |
36. | Adedokun OJ, Gunn GR, Leu JH, Gargano C, Xu Z, Sandborn WJ, Rutgeerts P, Shankar G. Immunogenicity of Golimumab and its Clinical Relevance in Patients With Ulcerative Colitis. Inflamm Bowel Dis. 2019;25:1532-1540. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 9] [Cited by in F6Publishing: 9] [Article Influence: 1.8] [Reference Citation Analysis (0)] |
37. | Masuda H, Iwai S, Tanaka T, Hayakawa S. Expression of IL-8, TNF-alpha and IFN-gamma m-RNA in ulcerative colitis, particularly in patients with inactive phase. J Clin Lab Immunol. 1995;46:111-123. [PubMed] [Cited in This Article: ] |
38. | Sedger LM, McDermott MF. TNF and TNF-receptors: From mediators of cell death and inflammation to therapeutic giants - past, present and future. Cytokine Growth Factor Rev. 2014;25:453-472. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 502] [Cited by in F6Publishing: 549] [Article Influence: 54.9] [Reference Citation Analysis (0)] |
39. | Gareb B, Otten AT, Frijlink HW, Dijkstra G, Kosterink JGW. Review: Local Tumor Necrosis Factor-α Inhibition in Inflammatory Bowel Disease. Pharmaceutics. 2020;12:539. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 56] [Cited by in F6Publishing: 46] [Article Influence: 11.5] [Reference Citation Analysis (0)] |
40. | Yoon SM, Haritunians T, Chhina S, Liu Z, Yang S, Landers C, Li D, Ye BD, Shih D, Vasiliauskas EA, Ippoliti A, Rabizadeh S, Targan SR, Melmed GY, McGovern DPB. Colonic Phenotypes Are Associated with Poorer Response to Anti-TNF Therapies in Patients with IBD. Inflamm Bowel Dis. 2017;23:1382-1393. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 19] [Cited by in F6Publishing: 15] [Article Influence: 2.1] [Reference Citation Analysis (0)] |
41. | Curci D, Lucafò M, Cifù A, Fabris M, Bramuzzo M, Martelossi S, Franca R, Decorti G, Stocco G. Pharmacogenetic variants of infliximab response in young patients with inflammatory bowel disease. Clin Transl Sci. 2021;14:2184-2192. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Cited by in F6Publishing: 3] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
42. | 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: 1256] [Article Influence: 418.7] [Reference Citation Analysis (0)] |
43. | Rokkas T, Portincasa P, Koutroubakis IE. Fecal calprotectin in assessing inflammatory bowel disease endoscopic activity: a diagnostic accuracy meta-analysis. J Gastrointestin Liver Dis. 2018;27:299-306. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 42] [Cited by in F6Publishing: 72] [Article Influence: 14.4] [Reference Citation Analysis (0)] |
44. | Haisma SM, Verkade HJ, Scheenstra R, van der Doef HPJ, Bodewes FAJA, van Rheenen PF. Time-to-reach Target Calprotectin Level in Newly Diagnosed Patients With Inflammatory Bowel Disease. J Pediatr Gastroenterol Nutr. 2019;69:466-473. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 12] [Cited by in F6Publishing: 16] [Article Influence: 3.2] [Reference Citation Analysis (0)] |
45. | Heida A, Park KT, van Rheenen PF. Clinical Utility of Fecal Calprotectin Monitoring in Asymptomatic Patients with Inflammatory Bowel Disease: A Systematic Review and Practical Guide. Inflamm Bowel Dis. 2017;23:894-902. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 87] [Cited by in F6Publishing: 96] [Article Influence: 13.7] [Reference Citation Analysis (0)] |
46. | Kennedy NA, Warner B, Johnston EL, Flanders L, Hendy P, Ding NS, Harris R, Fadra AS, Basquill C, Lamb CA, Cameron FL, Murray CD, Parkes M, Gooding I, Ahmad T, Gaya DR, Mann S, Lindsay JO, Gordon J, Satsangi J, Hart A, McCartney S, Irving P; UK Anti-TNF withdrawal study group, Lees CW. Relapse after withdrawal from anti-TNF therapy for inflammatory bowel disease: an observational study, plus systematic review and meta-analysis. Aliment Pharmacol Ther. 2016;43:910-923. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 68] [Cited by in F6Publishing: 85] [Article Influence: 10.6] [Reference Citation Analysis (0)] |
47. | Nakarai A, Kato J, Hiraoka S, Inokuchi T, Takei D, Morito Y, Akita M, Takahashi S, Hori K, Harada K, Okada H, Yamamoto K. Slight increases in the disease activity index and platelet count imply the presence of active intestinal lesions in C-reactive protein-negative Crohn's disease patients. Intern Med. 2014;53:1905-1911. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 10] [Cited by in F6Publishing: 15] [Article Influence: 1.5] [Reference Citation Analysis (0)] |
48. | Gisbert JP, Marín AC, Chaparro M. Systematic review: factors associated with relapse of inflammatory bowel disease after discontinuation of anti-TNF therapy. Aliment Pharmacol Ther. 2015;42:391-405. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 74] [Cited by in F6Publishing: 88] [Article Influence: 9.8] [Reference Citation Analysis (0)] |
49. | Stallmach A, Langbein C, Atreya R, Bruns T, Dignass A, Ende K, Hampe J, Hartmann F, Neurath MF, Maul J, Preiss JC, Schmelz R, Siegmund B, Schulze H, Teich N, von Arnim U, Baumgart DC, Schmidt C. Vedolizumab provides clinical benefit over 1 year in patients with active inflammatory bowel disease - a prospective multicenter observational study. Aliment Pharmacol Ther. 2016;44:1199-1212. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 116] [Cited by in F6Publishing: 127] [Article Influence: 15.9] [Reference Citation Analysis (0)] |
50. | Echarri A, Ollero V, Barreiro-de Acosta M, Fernández-Villaverde A, Hernández V, Lorenzo A, Pereira S, Carpio D, Castro J; EIGA Group. Clinical, biological, and endoscopic responses to adalimumab in antitumor necrosis factor-naive Crohn's disease: predictors of efficacy in clinical practice. Eur J Gastroenterol Hepatol. 2015;27:430-435. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 16] [Cited by in F6Publishing: 14] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
51. | Roblin X, Marotte H, Leclerc M, Del Tedesco E, Phelip JM, Peyrin-Biroulet L, Paul S. Combination of C-reactive protein, infliximab trough levels, and stable but not transient antibodies to infliximab are associated with loss of response to infliximab in inflammatory bowel disease. J Crohns Colitis. 2015;9:525-531. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 73] [Cited by in F6Publishing: 82] [Article Influence: 9.1] [Reference Citation Analysis (0)] |
52. | Neurath MF. Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat Immunol. 2019;20:970-979. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 248] [Cited by in F6Publishing: 404] [Article Influence: 80.8] [Reference Citation Analysis (0)] |
53. | Bek S, Nielsen JV, Bojesen AB, Franke A, Bank S, Vogel U, Andersen V. Systematic review: genetic biomarkers associated with anti-TNF treatment response in inflammatory bowel diseases. Aliment Pharmacol Ther. 2016;44:554-567. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 71] [Cited by in F6Publishing: 76] [Article Influence: 9.5] [Reference Citation Analysis (0)] |
54. | Netz U, Carter JV, Eichenberger MR, Dryden GW, Pan J, Rai SN, Galandiuk S. Genetic polymorphisms predict response to anti-tumor necrosis factor treatment in Crohn's disease. World J Gastroenterol. 2017;23:4958-4967. [PubMed] [DOI] [Cited in This Article: ] [Cited by in CrossRef: 26] [Cited by in F6Publishing: 25] [Article Influence: 3.6] [Reference Citation Analysis (0)] |
55. | Bank S, Andersen PS, Burisch J, Pedersen N, Roug S, Galsgaard J, Turino SY, Brodersen JB, Rashid S, Rasmussen BK, Avlund S, Olesen TB, Hoffmann HJ, Nexø BA, Sode J, Vogel U, Andersen V. Genetically determined high activity of IL-12 and IL-18 in ulcerative colitis and TLR5 in Crohns disease were associated with non-response to anti-TNF therapy. Pharmacogenomics J. 2018;18:87-97. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 31] [Cited by in F6Publishing: 37] [Article Influence: 5.3] [Reference Citation Analysis (0)] |
56. | Jürgens M, Laubender RP, Hartl F, Weidinger M, Seiderer J, Wagner J, Wetzke M, Beigel F, Pfennig S, Stallhofer J, Schnitzler F, Tillack C, Lohse P, Göke B, Glas J, Ochsenkühn T, Brand S. Disease activity, ANCA, and IL23R genotype status determine early response to infliximab in patients with ulcerative colitis. Am J Gastroenterol. 2010;105:1811-1819. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 116] [Cited by in F6Publishing: 127] [Article Influence: 9.1] [Reference Citation Analysis (0)] |
57. | Louis EJ, Watier HE, Schreiber S, Hampe J, Taillard F, Olson A, Thorne N, Zhang H, Colombel JF. Polymorphism in IgG Fc receptor gene FCGR3A and response to infliximab in Crohn's disease: a subanalysis of the ACCENT I study. Pharmacogenet Genomics. 2006;16:911-914. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 62] [Cited by in F6Publishing: 65] [Article Influence: 3.8] [Reference Citation Analysis (0)] |
58. | Barber GE, Yajnik V, Khalili H, Giallourakis C, Garber J, Xavier R, Ananthakrishnan AN. Genetic Markers Predict Primary Non-Response and Durable Response To Anti-TNF Biologic Therapies in Crohn's Disease. Am J Gastroenterol. 2016;111:1816-1822. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 64] [Cited by in F6Publishing: 73] [Article Influence: 9.1] [Reference Citation Analysis (0)] |
59. | Planell N, Masamunt MC, Leal RF, Rodríguez L, Esteller M, Lozano JJ, Ramírez A, Ayrizono MLS, Coy CSR, Alfaro I, Ordás I, Visvanathan S, Ricart E, Guardiola J, Panés J, Salas A. Usefulness of Transcriptional Blood Biomarkers as a Non-invasive Surrogate Marker of Mucosal Healing and Endoscopic Response in Ulcerative Colitis. J Crohns Colitis. 2017;11:1335-1346. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 35] [Cited by in F6Publishing: 45] [Article Influence: 6.4] [Reference Citation Analysis (0)] |
60. | Kalla R, Adams AT, Bergemalm D, Vatn S, Kennedy NA, Ricanek P, Lindstrom J, Ocklind A, Hjelm F, Ventham NT, Ho GT, Petren C, Repsilber D, Söderholm J, Pierik M, D'Amato M, Gomollón F, Olbjorn C, Jahnsen J, Vatn MH, Halfvarson J, Satsangi J. Serum proteomic profiling at diagnosis predicts clinical course, and need for intensification of treatment in inflammatory bowel disease. J Crohns Colitis. 2021;15:699-708. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 31] [Cited by in F6Publishing: 39] [Article Influence: 13.0] [Reference Citation Analysis (0)] |
61. | Gisbert JP, Chaparro M. Clinical Usefulness of Proteomics in Inflammatory Bowel Disease: A Comprehensive Review. J Crohns Colitis. 2019;13:374-384. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 25] [Cited by in F6Publishing: 31] [Article Influence: 6.2] [Reference Citation Analysis (0)] |
62. | Bohra A, Mohamed G, Vasudevan A, Lewis D, Van Langenberg DR, Segal JP. The Utility of Faecal Calprotectin, Lactoferrin and Other Faecal Biomarkers in Discriminating Endoscopic Activity in Crohn's Disease: A Systematic Review and Meta-Analysis. Biomedicines. 2023;11:1408. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 11] [Reference Citation Analysis (0)] |
63. | Ueno N, Sugiyama Y, Kobayashi Y, Murakami Y, Iwama T, Sasaki T, Kunogi T, Takahashi K, Tanaka K, Ando K, Kashima S, Inaba Y, Moriichi K, Tanabe H, Taruishi M, Saitoh Y, Okumura T, Fujiya M. Fecal calprotectin is a useful biomarker for predicting the clinical outcome of granulocyte and monocyte adsorptive apheresis in ulcerative colitis patients: a prospective observation study. BMC Gastroenterol. 2021;21:316. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 7] [Cited by in F6Publishing: 4] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
64. | Benítez JM, García-Sánchez V. Faecal calprotectin: Management in inflammatory bowel disease. World J Gastrointest Pathophysiol. 2015;6:203-209. [PubMed] [DOI] [Cited in This Article: ] [Cited by in CrossRef: 17] [Cited by in F6Publishing: 14] [Article Influence: 1.6] [Reference Citation Analysis (1)] |
65. | Cui G, Fan Q, Li Z, Goll R, Florholmen J. Evaluation of anti-TNF therapeutic response in patients with inflammatory bowel disease: Current and novel biomarkers. EBioMedicine. 2021;66:103329. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 10] [Cited by in F6Publishing: 44] [Article Influence: 14.7] [Reference Citation Analysis (0)] |
66. | Hassan EA, Ramadan HK, Ismael AA, Mohamed KF, El-Attar MM, Alhelali I. Noninvasive biomarkers as surrogate predictors of clinical and endoscopic remission after infliximab induction in patients with refractory ulcerative colitis. Saudi J Gastroenterol. 2017;23:238-245. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 6] [Reference Citation Analysis (0)] |
67. | Angelison L, Almer S, Eriksson A, Karling P, Fagerberg U, Halfvarson J, Thörn M, Björk J, Hindorf U, Löfberg R, Bajor A, Hjortswang H, Hammarlund P, Grip O, Torp J, Marsal J, Hertervig E; Swedish Organization for the Study of Inflammatory Bowel diseases (SOIBD). Long-term outcome of infliximab treatment in chronic active ulcerative colitis: a Swedish multicentre study of 250 patients. Aliment Pharmacol Ther. 2017;45:519-532. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 22] [Cited by in F6Publishing: 28] [Article Influence: 4.0] [Reference Citation Analysis (0)] |
68. | Ding NS, McDonald JAK, Perdones-Montero A, Rees DN, Adegbola SO, Misra R, Hendy P, Penez L, Marchesi JR, Holmes E, Sarafian MH, Hart AL. Metabonomics and the Gut Microbiome Associated With Primary Response to Anti-TNF Therapy in Crohn's Disease. J Crohns Colitis. 2020;14:1090-1102. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 36] [Cited by in F6Publishing: 64] [Article Influence: 16.0] [Reference Citation Analysis (0)] |
69. | Singh N, Rabizadeh S, Jossen J, Pittman N, Check M, Hashemi G, Phan BL, Hyams JS, Dubinsky MC. Multi-Center Experience of Vedolizumab Effectiveness in Pediatric Inflammatory Bowel Disease. Inflamm Bowel Dis. 2016;22:2121-2126. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 75] [Cited by in F6Publishing: 61] [Article Influence: 7.6] [Reference Citation Analysis (0)] |
70. | Lee JWJ, Plichta D, Hogstrom L, Borren NZ, Lau H, Gregory SM, Tan W, Khalili H, Clish C, Vlamakis H, Xavier RJ, Ananthakrishnan AN. Multi-omics reveal microbial determinants impacting responses to biologic therapies in inflammatory bowel disease. Cell Host Microbe. 2021;29:1294-1304. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 20] [Cited by in F6Publishing: 101] [Article Influence: 33.7] [Reference Citation Analysis (0)] |
71. | Jones-Hall YL, Nakatsu CH. The Intersection of TNF, IBD and the Microbiome. Gut Microbes. 2016;7:58-62. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 40] [Cited by in F6Publishing: 42] [Article Influence: 5.3] [Reference Citation Analysis (0)] |
72. | Peters CP, Eshuis EJ, Toxopeüs FM, Hellemons ME, Jansen JM, D'Haens GR, Fockens P, Stokkers PC, Tuynman HA, van Bodegraven AA, Ponsioen CY; North Holland GUT club. Adalimumab for Crohn's disease: long-term sustained benefit in a population-based cohort of 438 patients. J Crohns Colitis. 2014;8:866-875. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 57] [Cited by in F6Publishing: 63] [Article Influence: 6.3] [Reference Citation Analysis (0)] |
73. | Caenepeel C, Falony G, Machiels K, Verstockt B, Goncalves PJ, Ferrante M, Sabino J, Raes J, Vieira-Silva S, Vermeire S. Dysbiosis and Associated Stool Features Improve Prediction of Response to Biological Therapy in Inflammatory Bowel Disease. Gastroenterology. 2024;166:483-495. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3] [Reference Citation Analysis (0)] |
74. | Sokol H, Leducq V, Aschard H, Pham HP, Jegou S, Landman C, Cohen D, Liguori G, Bourrier A, Nion-Larmurier I, Cosnes J, Seksik P, Langella P, Skurnik D, Richard ML, Beaugerie L. Fungal microbiota dysbiosis in IBD. Gut. 2017;66:1039-1048. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 658] [Cited by in F6Publishing: 820] [Article Influence: 117.1] [Reference Citation Analysis (0)] |
75. | Kowalska-Duplaga K, Krawczyk A, Sroka-Oleksiak A, Salamon D, Wędrychowicz A, Fyderek K, Gosiewski T. Dependence of Colonization of the Large Intestine by Candida on the Treatment of Crohn's Disease. Pol J Microbiol. 2019;68:121-126. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 11] [Cited by in F6Publishing: 15] [Article Influence: 3.0] [Reference Citation Analysis (0)] |
76. | Ungaro F, Massimino L, D'Alessio S, Danese S. The gut virome in inflammatory bowel disease pathogenesis: From metagenomics to novel therapeutic approaches. United European Gastroenterol J. 2019;7:999-1007. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 22] [Cited by in F6Publishing: 39] [Article Influence: 7.8] [Reference Citation Analysis (0)] |
77. | James JP, Riis LB, Malham M, Høgdall E, Langholz E, Nielsen BS. MicroRNA Biomarkers in IBD-Differential Diagnosis and Prediction of Colitis-Associated Cancer. Int J Mol Sci. 2020;21:7893. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 30] [Cited by in F6Publishing: 65] [Article Influence: 16.3] [Reference Citation Analysis (0)] |
78. | Batra SK, Heier CR, Diaz-Calderon L, Tully CB, Fiorillo AA, van den Anker J, Conklin LS. Serum miRNAs Are Pharmacodynamic Biomarkers Associated With Therapeutic Response in Pediatric Inflammatory Bowel Disease. Inflamm Bowel Dis. 2020;26:1597-1606. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 18] [Cited by in F6Publishing: 34] [Article Influence: 8.5] [Reference Citation Analysis (0)] |
79. | Papaconstantinou I, Kapizioni C, Legaki E, Xourgia E, Karamanolis G, Gklavas A, Gazouli M. Association of miR-146 rs2910164, miR-196a rs11614913, miR-221 rs113054794 and miR-224 rs188519172 polymorphisms with anti-TNF treatment response in a Greek population with Crohn's disease. World J Gastrointest Pharmacol Ther. 2017;8:193-200. [PubMed] [DOI] [Cited in This Article: ] [Cited by in CrossRef: 9] [Cited by in F6Publishing: 12] [Article Influence: 1.7] [Reference Citation Analysis (0)] |
80. | Olsen T, Goll R, Cui G, Christiansen I, Florholmen J. TNF-alpha gene expression in colorectal mucosa as a predictor of remission after induction therapy with infliximab in ulcerative colitis. Cytokine. 2009;46:222-227. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 60] [Cited by in F6Publishing: 63] [Article Influence: 4.2] [Reference Citation Analysis (0)] |
81. | Dahlén R, Magnusson MK, Bajor A, Lasson A, Ung KA, Strid H, Öhman L. Global mucosal and serum cytokine profile in patients with ulcerative colitis undergoing anti-TNF therapy. Scand J Gastroenterol. 2015;50:1118-1126. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 26] [Cited by in F6Publishing: 38] [Article Influence: 4.8] [Reference Citation Analysis (0)] |
82. | 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: 416] [Article Influence: 83.2] [Reference Citation Analysis (0)] |
83. | Chang JT. Pathophysiology of Inflammatory Bowel Diseases. N Engl J Med. 2020;383:2652-2664. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 274] [Cited by in F6Publishing: 626] [Article Influence: 156.5] [Reference Citation Analysis (0)] |
84. | Koder S, Repnik K, Ferkolj I, Pernat C, Skok P, Weersma RK, Potočnik U. Genetic polymorphism in ATG16L1 gene influences the response to adalimumab in Crohn's disease patients. Pharmacogenomics. 2015;16:191-204. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 35] [Cited by in F6Publishing: 37] [Article Influence: 4.1] [Reference Citation Analysis (0)] |
85. | Vermeire S, Louis E, Carbonez A, Van Assche G, Noman M, Belaiche J, De Vos M, Van Gossum A, Pescatore P, Fiasse R, Pelckmans P, Reynaert H, D'Haens G, Rutgeerts P; Belgian Group of Infliximab Expanded Access Program in Crohn's Disease. Demographic and clinical parameters influencing the short-term outcome of anti-tumor necrosis factor (infliximab) treatment in Crohn's disease. Am J Gastroenterol. 2002;97:2357-2363. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 160] [Cited by in F6Publishing: 164] [Article Influence: 7.5] [Reference Citation Analysis (0)] |
86. | Tong Q, Zhao L, Qian XD, Zhang LL, Xu X, Dai SM, Cai Q, Zhao DB. Association of TNF-α polymorphism with prediction of response to TNF blockers in spondyloarthritis and inflammatory bowel disease: a meta-analysis. Pharmacogenomics. 2013;14:1691-1700. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 23] [Cited by in F6Publishing: 24] [Article Influence: 2.2] [Reference Citation Analysis (0)] |
87. | Salvador-Martín S, López-Cauce B, Nuñez O, Laserna-Mendieta EJ, García MI, Lobato E, Abarca-Zabalía J, Sanjurjo-Saez M, Lucendo AJ, Marín-Jiménez I, Menchén LA, López-Fernández LA. Genetic predictors of long-term response and trough levels of infliximab in crohn's disease. Pharmacol Res. 2019;149:104478. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 11] [Cited by in F6Publishing: 19] [Article Influence: 3.8] [Reference Citation Analysis (0)] |
88. | Bank S, Andersen PS, Burisch J, Pedersen N, Roug S, Galsgaard J, Turino SY, Brodersen JB, Rashid S, Rasmussen BK, Avlund S, Olesen TB, Hoffmann HJ, Thomsen MK, Thomsen VØ, Frydenberg M, Nexø BA, Sode J, Vogel U, Andersen V. Associations between functional polymorphisms in the NFκB signaling pathway and response to anti-TNF treatment in Danish patients with inflammatory bowel disease. Pharmacogenomics J. 2014;14:526-534. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 93] [Cited by in F6Publishing: 105] [Article Influence: 10.5] [Reference Citation Analysis (0)] |
89. | 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: 251] [Article Influence: 62.8] [Reference Citation Analysis (0)] |
90. | Billiet T, Vande Casteele N, Van Stappen T, Princen F, Singh S, Gils A, Ferrante M, Van Assche G, Cleynen I, Vermeire S. Immunogenicity to infliximab is associated with HLA-DRB1. Gut. 2015;64:1344-1345. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 47] [Cited by in F6Publishing: 50] [Article Influence: 5.6] [Reference Citation Analysis (0)] |
91. | Louis E, El Ghoul Z, Vermeire S, Dall'Ozzo S, Rutgeerts P, Paintaud G, Belaiche J, De Vos M, Van Gossum A, Colombel JF, Watier H. Association between polymorphism in IgG Fc receptor IIIa coding gene and biological response to infliximab in Crohn's disease. Aliment Pharmacol Ther. 2004;19:511-519. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 173] [Cited by in F6Publishing: 155] [Article Influence: 7.8] [Reference Citation Analysis (0)] |
92. | Vermeire S, Gils A, Accossato P, Lula S, Marren A. Immunogenicity of biologics in inflammatory bowel disease. Therap Adv Gastroenterol. 2018;11:1756283X17750355. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 157] [Cited by in F6Publishing: 162] [Article Influence: 27.0] [Reference Citation Analysis (0)] |
93. | Colombel JF, Adedokun OJ, Gasink C, Gao LL, Cornillie FJ, D'Haens GR, Rutgeerts PJ, Reinisch W, Sandborn WJ, Hanauer SB. Combination Therapy With Infliximab and Azathioprine Improves Infliximab Pharmacokinetic Features and Efficacy: A Post Hoc Analysis. Clin Gastroenterol Hepatol. 2019;17:1525-1532. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 84] [Cited by in F6Publishing: 119] [Article Influence: 23.8] [Reference Citation Analysis (0)] |
94. | Arijs I, Li K, Toedter G, Quintens R, Van Lommel L, Van Steen K, Leemans P, De Hertogh G, Lemaire K, Ferrante M, Schnitzler F, Thorrez L, Ma K, Song XY, Marano C, Van Assche G, Vermeire S, Geboes K, Schuit F, Baribaud F, Rutgeerts P. Mucosal gene signatures to predict response to infliximab in patients with ulcerative colitis. Gut. 2009;58:1612-1619. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 271] [Cited by in F6Publishing: 289] [Article Influence: 19.3] [Reference Citation Analysis (0)] |
95. | Verstockt B, Verstockt S, Creyns B, Tops S, Van Assche G, Gils A, Ceuppens JL, Vermeire S, Ferrante M, Breynaert C. Mucosal IL13RA2 expression predicts nonresponse to anti-TNF therapy in Crohn's disease. Aliment Pharmacol Ther. 2019;49:572-581. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 52] [Cited by in F6Publishing: 44] [Article Influence: 8.8] [Reference Citation Analysis (0)] |
96. | Ogura Y, Bonen DK, Inohara N, Nicolae DL, Chen FF, Ramos R, Britton H, Moran T, Karaliuskas R, Duerr RH, Achkar JP, Brant SR, Bayless TM, Kirschner BS, Hanauer SB, Nuñez G, Cho JH. A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature. 2001;411:603-606. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 3555] [Cited by in F6Publishing: 3431] [Article Influence: 149.2] [Reference Citation Analysis (1)] |
97. | Weersma RK, Stokkers PC, van Bodegraven AA, van Hogezand RA, Verspaget HW, de Jong DJ, van der Woude CJ, Oldenburg B, Linskens RK, Festen EA, van der Steege G, Hommes DW, Crusius JB, Wijmenga C, Nolte IM, Dijkstra G; Dutch Initiative on Crohn and Colitis (ICC). Molecular prediction of disease risk and severity in a large Dutch Crohn's disease cohort. Gut. 2009;58:388-395. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 127] [Cited by in F6Publishing: 142] [Article Influence: 9.5] [Reference Citation Analysis (0)] |
98. | Niess JH, Klaus J, Stephani J, Pflüger C, Degenkolb N, Spaniol U, Mayer B, Lahr G, von Boyen GB. NOD2 polymorphism predicts response to treatment in Crohn's disease--first steps to a personalized therapy. Dig Dis Sci. 2012;57:879-886. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 28] [Cited by in F6Publishing: 24] [Article Influence: 2.0] [Reference Citation Analysis (0)] |
99. | Juanola O, Moratalla A, Gutiérrez A, Sempere L, Zapater P, Giménez P, Almenta I, Peiró G, González-Navajas JM, Such JF, Francés R. Anti-TNF-alpha loss of response is associated with a decreased percentage of FoxP3+ T cells and a variant NOD2 genotype in patients with Crohn's disease. J Gastroenterol. 2015;50:758-768. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 10] [Cited by in F6Publishing: 10] [Article Influence: 1.1] [Reference Citation Analysis (0)] |
100. | Schäffler H, Geiss D, Gittel N, Rohde S, Huth A, Glass Ä, Brandhorst G, Jaster R, Lamprecht G. Mutations in the NOD2 gene are associated with a specific phenotype and lower anti-tumor necrosis factor trough levels in Crohn's disease. J Dig Dis. 2018;19:678-684. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 10] [Cited by in F6Publishing: 15] [Article Influence: 2.5] [Reference Citation Analysis (0)] |
101. | Steenholdt C, Enevold C, Ainsworth MA, Brynskov J, Thomsen OØ, Bendtzen K. Genetic polymorphisms of tumour necrosis factor receptor superfamily 1b and fas ligand are associated with clinical efficacy and/or acute severe infusion reactions to infliximab in Crohn's disease. Aliment Pharmacol Ther. 2012;36:650-659. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 33] [Cited by in F6Publishing: 37] [Article Influence: 3.1] [Reference Citation Analysis (0)] |
102. | Medrano LM, Taxonera C, Márquez A, Barreiro-de Acosta M, Gómez-García M, González-Artacho C, Pérez-Calle JL, Bermejo F, Lopez-Sanromán A, Martín Arranz MD, Gisbert JP, Mendoza JL, Martín J, Urcelay E, Núñez C. Role of TNFRSF1B polymorphisms in the response of Crohn's disease patients to infliximab. Hum Immunol. 2014;75:71-75. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 33] [Cited by in F6Publishing: 45] [Article Influence: 4.5] [Reference Citation Analysis (0)] |
103. | Song GG, Seo YH, Kim JH, Choi SJ, Ji JD, Lee YH. Association between TNF-α (-308 A/G, -238 A/G, -857 C/T) polymorphisms and responsiveness to TNF-α blockers in spondyloarthropathy, psoriasis and Crohn's disease: a meta-analysis. Pharmacogenomics. 2015;16:1427-1437. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 33] [Cited by in F6Publishing: 34] [Article Influence: 3.8] [Reference Citation Analysis (0)] |
104. | Hlavaty T, Ferrante M, Henckaerts L, Pierik M, Rutgeerts P, Vermeire S. Predictive model for the outcome of infliximab therapy in Crohn's disease based on apoptotic pharmacogenetic index and clinical predictors. Inflamm Bowel Dis. 2007;13:372-379. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 56] [Cited by in F6Publishing: 46] [Article Influence: 2.7] [Reference Citation Analysis (0)] |
105. | Burke KE, Khalili H, Garber JJ, Haritunians T, McGovern DPB, Xavier RJ, Ananthakrishnan AN. Genetic Markers Predict Primary Nonresponse and Durable Response to Anti-Tumor Necrosis Factor Therapy in Ulcerative Colitis. Inflamm Bowel Dis. 2018;24:1840-1848. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 28] [Cited by in F6Publishing: 27] [Article Influence: 4.5] [Reference Citation Analysis (0)] |
106. | Wang MH, Friton JJ, Raffals LE, Leighton JA, Pasha SF, Picco MF, Cushing KC, Monroe K, Nix BD, Newberry RD, Faubion WA. Novel Genetic Risk Variants Can Predict Anti-TNF Agent Response in Patients With Inflammatory Bowel Disease. J Crohns Colitis. 2019;13:1036-1043. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 8] [Cited by in F6Publishing: 8] [Article Influence: 1.6] [Reference Citation Analysis (0)] |
107. | Thomas D, Gazouli M, Karantanos T, Rigoglou S, Karamanolis G, Bramis K, Zografos G, Theodoropoulos GE. Association of rs1568885, rs1813443 and rs4411591 polymorphisms with anti-TNF medication response in Greek patients with Crohn's disease. World J Gastroenterol. 2014;20:3609-3614. [PubMed] [DOI] [Cited in This Article: ] [Cited by in CrossRef: 18] [Cited by in F6Publishing: 17] [Article Influence: 1.7] [Reference Citation Analysis (0)] |
108. | Salvador-Martín S, Bossacoma F, Pujol-Muncunill G, Navas-López VM, Gallego-Fernández C, Viada J, Muñoz-Codoceo R, Magallares L, Martínez-Ojinaga E, Moreno-Álvarez A, Solar-Boga A, Segarra O, Clemente S, Rodriguez-Martinez A, Alvarez-Vayo C, Loverdos I, Merino-Bohórquez V, Balboa-Vega MJ, Blanca-García JA, Fobelo MJ, Millán-Jiménez A, García-Romero R, Sanchez C, Tolín M, Caldas RG, Eizaguirre FJ, Sánchez-Hernandez JG, Torres-Peral R, Aznal E, García-González X, Sanjurjo-Sáez M, López-Fernández LA. Genetic Predictors of Long-term Response to Antitumor Necrosis Factor Agents in Pediatric Inflammatory Bowel Disease. J Pediatr Gastroenterol Nutr. 2020;71:508-515. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 5] [Cited by in F6Publishing: 4] [Article Influence: 1.0] [Reference Citation Analysis (0)] |
109. | Salvador-Martín S, Zapata-Cobo P, Velasco M, Palomino LM, Clemente S, Segarra O, Sánchez C, Tolín M, Moreno-Álvarez A, Fernández-Lorenzo A, Pérez-Moneo B, Loverdos I, Navas López VM, Millán A, Magallares L, Torres-Peral R, García-Romero R, Pujol-Muncunill G, Merino-Bohorquez V, Rodríguez A, Salcedo E, López-Cauce B, Marín-Jiménez I, Menchén L, Laserna-Mendieta E, Lucendo AJ, Sanjurjo-Sáez M, López-Fernández LA. Association between HLA DNA Variants and Long-Term Response to Anti-TNF Drugs in a Spanish Pediatric Inflammatory Bowel Disease Cohort. Int J Mol Sci. 2023;24:1797. [PubMed] [DOI] [Cited in This Article: ] [Cited by in F6Publishing: 10] [Reference Citation Analysis (0)] |
110. | Hoffmann P, Lamerz D, Hill P, Kirchner M, Gauss A. Gene Polymorphisms of NOD2, IL23R, PTPN2 and ATG16L1 in Patients with Crohn's Disease: On the Way to Personalized Medicine? Genes (Basel). 2021;12:866. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 2] [Cited by in F6Publishing: 18] [Article Influence: 6.0] [Reference Citation Analysis (0)] |
111. | Chen G, Shen J. Artificial Intelligence Enhances Studies on Inflammatory Bowel Disease. Front Bioeng Biotechnol. 2021;9:635764. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 4] [Cited by in F6Publishing: 4] [Article Influence: 1.3] [Reference Citation Analysis (0)] |
112. | Russo V, Lallo E, Munnia A, Spedicato M, Messerini L, D'Aurizio R, Ceroni EG, Brunelli G, Galvano A, Russo A, Landini I, Nobili S, Ceppi M, Bruzzone M, Cianchi F, Staderini F, Roselli M, Riondino S, Ferroni P, Guadagni F, Mini E, Peluso M. Artificial Intelligence Predictive Models of Response to Cytotoxic Chemotherapy Alone or Combined to Targeted Therapy for Metastatic Colorectal Cancer Patients: A Systematic Review and Meta-Analysis. Cancers (Basel). 2022;14:4012. [PubMed] [DOI] [Cited in This Article: ] [Cited by in Crossref: 5] [Cited by in F6Publishing: 6] [Article Influence: 3.0] [Reference Citation Analysis (0)] |