Next Article in Journal
Impact of pri-let-7a-1 rs10739971 for Gastric Cancer Predisposition in an Amazon Region
Previous Article in Journal
Effects of L-arginine on Nitric Oxide Synthesis and Larval Metamorphosis of Mytilus coruscus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

MIF Variant rs755622 Is Associated with Severe Crohn’s Disease and Better Response to Anti-TNF Adalimumab Therapy

1
Faculty of Medicine, University of Maribor, Taborska Ulica 8, 2000 Maribor, Slovenia
2
Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova Ulica 17, 2000 Maribor, Slovenia
3
Department for Science and Research, University Medical Centre Maribor, Ljubljanska Ulica 5, 2000 Maribor, Slovenia
*
Author to whom correspondence should be addressed.
Genes 2023, 14(2), 452; https://doi.org/10.3390/genes14020452
Submission received: 18 January 2023 / Revised: 2 February 2023 / Accepted: 7 February 2023 / Published: 9 February 2023
(This article belongs to the Section Pharmacogenetics)

Abstract

:
Crohn’s disease (CD), rheumatoid arthritis, psoriatic arthritis and other inflammatory diseases comprise a group of chronic diseases with immune-mediated pathogenesis which share common pathological pathways, as well as treatment strategies including anti-TNF biologic therapy. However, the response rate to anti-TNF therapy among those diseases varies, and approximately one third of patients do not respond. Since pharmacogenetic studies for anti-TNF therapy have been more frequent for other related diseases and are rare in CD, the aim of our study was to further explore markers associated with anti-TNF response in other inflammatory diseases in Slovenian CD patients treated with the anti-TNF drug adalimumab (ADA). We enrolled 102 CD patients on ADA, for which the response was defined after 4, 12, 20 and 30 weeks of treatment, using an IBDQ questionnaire and blood CRP value. We genotyped 41 SNPs significantly associated with response to anti-TNF treatment in other diseases. We found novel pharmacogenetic association between SNP rs755622 in the gene MIF (macrophage migration inhibitory factor) and SNP rs3740691 in the gene ARFGAP2 in CD patients treated with ADA. The strongest and most consistent association with treatment response was found for the variant rs2275913 in gene IL17A (p = 9.73 × 10−3).

Graphical Abstract

1. Introduction

Crohn’s disease (CD), rheumatoid arthritis (RA), psoriatic arthritis (PA) and other inflammatory diseases comprise a group of chronic diseases with immune-mediated pathogenesis. Since these are all inflammatory diseases, they share common pathological pathways [1]. Tumor necrosis factor α inhibitors (anti-TNF) have improved the treatment of the majority of autoimmune inflammatory complex diseases, and give substantial improvement in cases where convenient treatment using non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids and antibiotics was not successful. Targeting tumor necrosis factor α in IBD allows intestinal healing by blocking TNFR1-dependant intestinal epithelial cell death [2] and inducing cell death in macrophages by binding to transmembrane TNF or by depriving TNFR2-dependent CD4+ T cell survival via NF-κB activation [3]. However, 30–40% of patients have an inadequate response to anti-TNF drugs. It is known that genetic factors influence the response to anti-TNF treatment [4]. Anti-TNF pharmacogenetic studies, including the different anti-TNF drugs infliximab (IFX), adalimumab (ADA) and etanercept (ETN), have so far been performed in RA, inflammatory bowel disease (IBD), PA and ankylosing spondylitis (AS) patients. Most of them have focused on candidate genes known to play a role in susceptibility to disease, and genes implicated in TNFα signaling pathways. Recently, some genome wide association studies (GWAs) of genetic predictors of anti-TNF treatment efficacy have been also performed in RA [5,6] and pediatric IBD [4]. Pharmacogenomic GWA studies are performed on modest sample sizes compared to genetic studies of disease risk. Furthermore, there is a considerable difference between the type of anti-TNF treatment [7] and use of concomitant medications [8]. Moreover, the advent of biosimilars has lowered the treatment cost and increased the drug supply, thereby bringing anti-TNF treatment to even more patients [9]. That is why independent cohort studies are required to validate findings from other cohorts, diseases and GWAs further. The aim of our study is the exploration of previously published anti-TNF response markers in our well-defined Slovenian CD patient cohort treated with ADA.

2. Materials and Methods

2.1. Literature Search and SNP Selection

An extensive literature search was performed in PubMed using various search terms and combinations of these terms, such as anti-TNF, pharmacogenetic study, genome-wide association study, autoimmune diseases, etc. Pharmacogenetic studies of anti-TNF response in autoimmune inflammatory diseases, including Crohn’s disease (CD), ulcerative colitis (UC), RA, PA, AS, spondyloarthritis (SpA) and multiple sclerosis (MS), published between the years 2001 and 2015, were used to identify loci associated with response to biological therapy using anti-TNF agents, including IFX, ADA or ETN. All SNPs that were associated with response to any anti-TNF drug were included.

2.2. Patients

We enrolled patients with CD on ADA as described previously [10]. Exclusion criteria were: other complications of CD (e.g., stenosis, abscesses, previous total colectomy), a history of allergy to murine proteins, a serious infection in the previous 3 months, positive test on tuberculosis or active tuberculosis, malignancy, pregnancy and lactation) [10]. Briefly, 102 Slovenian patients with refractory CD were investigated over a period of 30 weeks. Before the first dose of ADA and 4, 12, 20 and 30 weeks after treatment, the response was determined using an IBDQ questionnaire score (clinical response) and blood CRP value (biological response). Both the clinical and biological responses were determined as the difference in the IBDQ or CRP value before and after treatment. Clinical response was defined as an increase in IBDQ by more than 22 points (ΔIBDQ > 22), or as an IBDQ value higher than 170 points [11], and biological response as a decrease in CRP to normal values (<3 mg/L), or a drop in CRP levels by more than 25% [12,13]. The clinical characteristics (including disease location and behavior according to the Montreal classification) of the cohorts are listed in Table 1.

2.3. Genetic Analysis

DNA samples were available for all patients. The DNA was isolated from peripheral blood lymphocytes using a TRI reagent (Sigma, Darmstadt, Germany) according to the manufacturer’s instructions. Genotypes for forty-one (41) SNPs selected from anti-TNF pharmacogenetic studies performed in PA, RA, IBD, CD, UC, MS, AS and SpA were extracted from our genotype data bank. The genotype data bank was obtained using the iChip platform as described previously [14].

2.4. Gene Ontology Analysis

Functional annotation was performed using publicly available functional and biological databases. Gene ontology analysis was performed using the software package CytoScape 3.8.2. [15] with the integrated application ClueGO v2.5.8. [16]. A ClueGO analysis was performed using the following parameters and selected options:
  • Ontology/Pathways selected:
  • Biological Process (13.05.2021)
  • Cellular Component (13.05.2021)
  • Molecular Function (13.05.2021)
  • Evidence selected: only All_Experimental
Statistical significance was defined as a p value lower than 5 × 10−2 after Bonferroni step-down correction (the default selection in ClueGO v2.5.8).
To enhance biological process discovery with gene ontology analysis, the lists of investigated genes were extended to include their interactors. Genes interacting with at least two investigated genes (i.e., genes associated with response to ADA) were obtained from the BIOGRID database [17,18] using the biogridR package [19] for R 4.1.1 [20].

2.5. Statistical Analysis

We used the two-sided Fisher’s exact test to compare genotype and allele frequencies between response to treatment as a categorical variable (response versus non-response) to ADA treatment. To compare continuous data between different genotypes (dominant and recessive models) and alleles or treatment response, we used an independent samples t-test in cases of normal distribution of data (the Kolmogorov–Smirnov test—p > 0.05), or the Mann–Whitney U-test in cases where the data deviated significantly from a normal distribution (the Kolmogorov–Smirnov test of normality—p < 0.05). Fisher combined p-value analysis was performed in cases where associations had been significant according to different statistics calculated from the same sample (multi-phase analysis). For the statistical analysis we used the IBM SPSS Statistics 22.0 statistical package.

3. Results

3.1. Literature Search

Out of 22 studies we collected 73 SNPs from 47 independent loci associated with response to anti-TNF therapy. In total, 50% of the studies (11 out of 22) were performed in IBD patients (IBD pediatric, IBD adults, CD or UC), and 36.4% of the studies (8 out of 22) were performed in RA patients. In total, 64.4% (47) of SNPs were associated with response to anti-TNF therapy in a group of IBD patients, 31.5% (23) in a group of RA patients and 9.6% (7) in a group of AS patients. Two SNPs were associated with anti-TNF response in a group of patients with SpA, one SNP in patients with PA, and one SNP in patients with MS. Table 2 summarizes the pharmacogenetic signals statistically significantly associated with response to anti-TNF therapy in RA, PA, SpA, IBD (CD and UC), pediatric IBD (PED-IBD), AS and MS patients, in the years between 2001 and 2015. Additional variant information is contained within Supplementary Materials (Table S2).

3.2. Pharmacogenetic Analysis

No statistically significant associations were detected between the analyzed SNPs and clinical data. Three loci showed strong association with treatment response to ADA in CD patients. The most consistent association during 30 weeks of treatment with ADA was observed between the SNP rs2275913 in gene IL17A and the response measured by the IBDQ. Patients with a GG genotype of SNP rs2275913 had a better response compared to patients with an AA or AG genotype (Figure 1). The strongest statistically significant association was confirmed after 20 weeks of treatment. The average difference in the IBDQ value for patients with the genotype GG was higher (31.9) compared to patients with the AA or AG genotype (13.8, p = 9.73 × 10−3). After 20 weeks of treatment 75.5% of patients with the genotype GG had a positive response to anti-TNF therapy with ADA compared to 54.5% of patients with response with genotype AA or AG (p = 4.67 × 10−2). The same tendency was observed during all 30 weeks of treatment (Table 3). The combined p-value analysis showed the strongest statistical significance after 20 weeks of treatment (p = 6.43 × 10−4).
Consistent association during all 30 weeks of treatment was also observed for the SNP rs755622 in the gene MIF (Figure 2). Patients with a GG genotype showed better response compared to patients with a CC or CG genotype. After 4 weeks of treatment with ADA, patients with GG had a higher deltaIBQ (60.6) compared to patients with a CC or CG genotype (16.4, p = 4.00 × 10−3). The same tendency was also observed after 12, 20 and 30 weeks of treatment. Association has also been observed for biological response measured with CRP, where patients with the genotype GG had a higher deltaCRP after 12 weeks of treatment (11.3) compared to patients with a CC or CG genotype (6.8, p = 0.026). Interestingly, patients with the GG genotype had a significantly lower IBDQ value (122) before treatment compared to patients with CC or CG genotypes (155, p = 0.039).
After four weeks of treatment, a strong, statistically significant association was confirmed for SNP rs3740691 in the gene ARFGAP2 (Figure 3). In the group of patients with genotype AA or AG there were 59.6% of nonresponders compared to 15.1% of nonresponders in the group of patients with genotype GG (p = 1.24 × 10−5). Furthermore, after four weeks of treatment, the average IBDQ value in patients with genotype AA or AG reached only 158.3 points compared to 183.7 points in patients with the genotype GG (p = 2.74 × 10−4). The difference also remained significant after 12 weeks of treatment. The combined p-value analysis showed the strongest statistical significance for SNP rs3740691 in gene ARFGAP2 after 4 weeks of treatment (p = 2.24 × 10−9).
Significant associations were confirmed for genes involved in the regulation of NF-κB signaling, particularly TLR2 (p = 1.48 × 10−3), TLR4 (p = 1.36 × 10−2) and TLR9 (p = 1.98 × 10−2).
Associations between the response to ADA therapy in Slovenian CD patients were altogether found for 28 out of the analyzed 41 SNPs. Our analysis replicated 17 (36.2%) of 47 SNPs associated with anti-TNF response in IBD. Not all SNPs could be replicated reliably due to the nature of IBD and anti-TNF response as a complex trait. The majority of the confirmed associations were between SNPs already associated with response to any anti-TNF drug in IBD (UC or CD) patients. However, the highest overlap was observed between SNPs associated with the response to anti-TNF therapy in AS patients. All associations are presented in Table 3.

3.3. Gene Ontology Analysis

To analyze whether there are specific processes associated with response to anti-TNF therapy with ADA in CD patients, we first performed gene ontology analysis only for genes associated with response (i.e., the genes listed in Table 3). Secondly, we extended the gene ontology analysis to interactors of genes listed in Table 3 obtained from BIOGRID. Finally, we performed gene ontology analysis for all genes reported to be associated with response to anti-TNF therapy in PA, RA, IBD, CD, UC, SpA, MS and AS from Table 2.
Gene ontology analysis of the genes listed in Table 3 showed few significant results, but their extended list containing BIOGRID interactors revealed several enriched GO terms. The genes and their interacting nodes are visualized in Figure 4. Many highly significant enriched GO terms are related to NF-kappaB signaling and TNFα, the most significant being I-kappaB kinase/NF-kappaB signaling (p = 2.76 × 10−37). Other significant terms include death-inducing signaling complex assembly (p = 4.67 × 10−15) and TRIF-dependent Toll-like receptor signaling pathway (p = 1.28 × 10−25).
The addition of genes from Table 2 to the gene ontology analysis did not alter or expand the GO results significantly. The newly identified leading terms are response to bacterium (p = 3.30 × 10−12) and related hyponyms. The full results of the gene ontology analysis are shown in Table S1.
Moreover, pathways of interest were selected and visualized based on genes of interest and their interactors, as well as statistically significant GO results (Figure 5). Figure 5 displays primarily a part of the TNFR1-related pathways, TRIF-dependent Toll-like receptor signaling for TLR3 and TLR4. Figure 5 is based on images published by Rusu et al. [2] and Aluri et al. [39].

4. Discussion

In the present study we performed an extensive pharmacogenetic study in CD patients treated with ADA. This is the first replication study of cross-disease anti-TNF signals performed in a well-characterized cohort of refractory CD patients treated specifically with the anti-TNF drug ADA. We identified a novel association between the MIF variant rs755622, severity of CD and response to anti-TNF therapy in CD patients, and also confirmed the IL17A variant rs2275913 as the strongest and most consistent predictor of response to ADA in CD patients during 30 weeks of treatment. For the IL17A variant rs2275913 we found a better response in patients with genotype GG compared to patients with the genotype AA or AG. On average, after 20 weeks of treatment, patients with the GG genotype had a 30-point increase in IBDQ score, a 20-point increase in the IBDQ value, and among patients with the GG genotype, there were 82% of responders compared to 53% of responders with the AA or AG genotype. Our finding is consistent with the observation that IBD patients with an AG or AA genotype of IL17A rs2275913 SNP are associated with nonresponse [26], and is contrary to the finding that female RA patients carrying the GG genotype are characterized by a poor response to anti-TNF treatment [25]. IL-17A, which is produced mainly by Th17 cells, mediates autoimmunity and immune defense against pathogens, and is increased in the intestinal mucosa of patients affected by chronic inflammatory bowel disorders, such as celiac disease, CD, and UC. It was also found that IL-17A is overexpressed in CD strictures compared with non-structured CD areas and gut control [40], and that IL-17A is increased in the inflamed areas of patients with IBD [41], and has a role in epithelial permeability independent of IL-23 [42], further confirming the role of IL17A in the pathogenesis of CD. Indeed, the association with an inflated gut area and gut permeability could open the epithelium to more common host–microbe interactions in the otherwise sterile lamina propria, leading to frequent inflammatory responses and greater odds of chronic and severe disease progression. Meanwhile, MIF is a key cytokine in RA, and changes following anti-TNF therapy were observed in RA almost a decade ago [43]. However, similar changes have not been observed in CD.
For the first time we identified an association between SNP rs755622 in the gene MIF (macrophage migration inhibitory factor) and response to anti-TNF therapy using ADA in patients with CD. We found a better response in patients with the GG genotype for SNP rs755622, in which the difference in the IBDQ score was higher in the 30 week period of treatment compared to patients with a CG or CC genotype. However, patients with the genotype GG were associated with more severe disease prior to the introduction of the anti-TNF therapy. An association between SNP rs755622 and response to anti-TNF therapy has been found in RA patients where minor allele G predicted nonresponse to anti-TNF treatment [24], which is contrary to our results. However, SNP rs755622 was previously associated with more active disease in RA patients [44], where carriers of the minor allele had higher levels of circulating MIF and higher levels of radiological joint damage. Interestingly, AS patients with rs755622 risk allele G also reflect a more active disease [24]. These two observations are consistent with our finding that patients with genotype GG have more severe disease before anti-TNF therapy.
For the first time, we report the association of SNP rs3740691 in gene ARFGAP2 with the response to ADA in CD patients.
Associations between a response to ADA therapy in Slovenian CD patients were altogether found in 28 out of the analyzed 41 SNPs, confirming high overlap with other related diseases. The majority of the confirmed associations were between SNPs already associated with response to any anti-TNF drug in IBD patients. However, the highest overlap was observed between SNPs associated with response to anti-TNF therapy in ankylosing spondylitis (AS) patients. AS is characterized by prominent inflammation of the axial skeleton, although other joints may also be affected. The relationship between IBD and AS has been known for many years. Approximately 5–10% of AS patients have concomitant IBD, either CD or UC, and first-degree relatives of patients with AS are ~3 times more likely to develop CD or UC than unrelated individuals [45,46,47]. Similarly, half of IBD patients develop chronic back pain, and this back pain progresses in about 5 to 10% of IBD patients to become a spondylopathy disorder [48]. Further, current data are consistent with the hypothesis that defective gut mucosal immunity is a major driver of AS, and many genetic associations in AS and IBD overlap [47]. Although genetic studies confirmed the association between AS and CD, the overlap between the pharmacogenetic markers has not been evaluated so far. In our study, a high overlap between markers of response to anti-TNF therapy has been found between AS and CD, further confirming cross-phenotype similarities and associations. From a therapeutic perspective, both infliximab and adalimumab are indicated for use in IBD and AS. Interestingly, another anti-TNF agent, etanercept, has been indicated for AS, but failed to achieve a therapeutic effect in IBD. The current understanding of etanercept’s failure in IBD highlights its ability to bind to soluble, but not transmembrane, TNF, which is believed to be the key source of TNF-related pathogenic effects in IBD [49].
We also wanted to explore the biological pathways and molecular functions involved in the response to anti-TNF treatment in CD, and compare them with other related common autoimmune diseases. Inflammatory response through NF-kappaB signaling has been found to be the most significant biological pathway associated with response to ADA. The results of the GO analysis related to NF-kappaB signaling highlight its importance in anti-TNF response, since NF-κB is the principal mediator of several pro-inflammatory processes in different immune-mediated diseases, including CD. Aberrant changes in genes involved in NF-κB signaling may lead to lower rates of satisfactory anti-TNF response. Additional functional testing is required to explain how variants in genes associated with GO terms related to NF-κB may affect the anti-TNF response.
The GO term result death-inducing signaling complex assembly is likely related to TNFR1 signaling, which mediates both the TNF-induced canonical NF-κB pathway and the TNFR1-dependent death induction. Anti-TNF therapy is also believed to ameliorate aberrant changes in TNFR1-dependent pathways by preventing canonical NF-κB pathway activation and cell death of affected tissue, such as the gut epithelium in IBD and the synovium in RA. In addition, the GO term TRIF-dependent Toll-like receptor signaling pathway may also be related to the described TNFR1-dependent signaling pathway. In IBD, both TLR3 and TRIF can contribute to the TNFR1-dependent pathway, which leads to cell death in the intestinal epithelium [2], but may also activate apoptosis and necroptosis pathways independent of TNFR1.
When comparing all genes associated with response to anti-TNF therapy, the response to bacterium, more specifically, the response to lipopolysaccharide, showed the strongest association, followed again by an inflammatory response through NF-kappaB signaling. So far, few studies have analyzed the biological processes involved in response/nonresponse to anti-TNF therapy. In a GWAS of anti-TNF response in RA patients strong involvement has been confirmed of the biological processes underlying the inflammatory response and cell morphology [6]. In our study, NF-kappaB signaling and bacterium response pathways are recognized as one of the most important processes contributing to a response to ADA.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14020452/s1, Table S1: Full gene ontology results. Table S2: Additional genetic variant information.

Author Contributions

Conceptualization, G.J., M.G. and U.P.; methodology, G.J. and M.G.; validation, G.J. and M.G.; formal analysis, G.J. and U.P.; investigation, G.J. and U.P.; data curation, G.J.; writing—original draft preparation, G.J.; writing—review and editing, M.G. and U.P.; visualization, G.J.; supervision, U.P. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge the financial support from the Slovenian Research Agency, Research Core Funding No. P3-0427 and Research Grant no. J3-9258.

Institutional Review Board Statement

The study was approved by the Slovenian National Committee for Medical Ethics (KME 80/10/07, 21p/12/07). Patients gave informed consent prior to inclusion in the study, and the study was performed in accordance with The Code of Ethics of the World Medical Association (Declaration of Helsinki).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study. Written informed consent was obtained from the participants to publish this paper. The study was in accordance with The Code of Ethics of the World Medical Association (Declaration of Helsinki), and approved by the Republic of Slovenia National Medical Ethics Committee, Ministry of Health, with the reference number KME 80/10/07, 21p/12/07.

Data Availability Statement

The data presented in this study are available on request from the corresponding author. The data are not publicly available due to patient privacy reasons.

Acknowledgments

The authors would like to thank Katja Repnik for technical support.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Blandizzi, C.; Gionchetti, P.; Armuzzi, A.; Caporali, R.; Chimenti, S.; Cimaz, R.; Cimino, L.; Lapadula, G.; Lionetti, P.; Marchesoni, A.; et al. The role of tumour necrosis factor in the pathogenesis of immune-mediated diseases. Int. J. Immunopathol. Pharmacol. 2014, 27, 1–10. [Google Scholar] [CrossRef] [PubMed]
  2. Rusu, I.; Mennillo, E.; Bain, J.L.; Li, Z.; Sun, X.; Ly, K.M.; Rosli, Y.Y.; Naser, M.; Wang, Z.; Advincula, R.; et al. Microbial signals, MyD88, and lymphotoxin drive TNF-independent intestinal epithelial tissue damage. J. Clin. Investig. 2022, 132, e154993. [Google Scholar] [CrossRef] [PubMed]
  3. Billmeier, U.; Dieterich, W.; Neurath, M.F.; Atreya, R. Molecular mechanism of action of anti-tumor necrosis factor antibodies in inflammatory bowel diseases. World J. Gastroenterol. 2016, 22, 9300–9313. [Google Scholar] [CrossRef]
  4. Dubinsky, M.C.; Mei, L.; Friedman, M.; Dhere, T.; Haritunians, T.; Hakonarson, H.; Kim, C.; Glessner, J.; Targan, S.R.; McGovern, D.P.; et al. Genome wide association (GWA) predictors of anti-TNFalpha therapeutic responsiveness in pediatric inflammatory bowel disease. Inflamm. Bowel. Dis. 2010, 16, 1357–1366. [Google Scholar] [CrossRef]
  5. Cui, J.; Stahl, E.A.; Saevarsdottir, S.; Miceli, C.; Diogo, D.; Trynka, G.; Raj, T.; Mirkov, M.U.; Canhao, H.; Ikari, K.; et al. Genome-wide association study and gene expression analysis identifies CD84 as a predictor of response to etanercept therapy in rheumatoid arthritis. PLoS Genet. 2013, 9, e1003394. [Google Scholar] [CrossRef]
  6. Umiċeviċ Mirkov, M.; Cui, J.; Vermeulen, S.H.; Stahl, E.A.; Toonen, E.J.; Makkinje, R.R.; Lee, A.T.; Huizinga, T.W.; Allaart, R.; Barton, A.; et al. Genome-wide association analysis of anti-TNF drug response in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2013, 72, 1375–1381. [Google Scholar] [CrossRef]
  7. Visuri, I.; Eriksson, C.; Olén, O.; Cao, Y.; Mårdberg, E.; Grip, O.; Gustavsson, A.; Hjortswang, H.; Karling, P.; Montgomery, S.; et al. Predictors of drug survival: A cohort study comparing anti-tumour necrosis factor agents using the Swedish inflammatory bowel disease quality register. Aliment. Pharmacol. Ther. 2021, 54, 931–943. [Google Scholar] [CrossRef]
  8. Hong, S.W.; Park, J.; Yoon, H.; Yang, H.R.; Shin, C.M.; Park, Y.S.; Kim, N.; Lee, D.H.; Kim, J.S. Comparison of loss of response between anti-tumor necrosis factor alone and combined use with immunomodulators in patients with inflammatory bowel disease. Korean J. Intern. Med. 2021, 36, S9–S17. [Google Scholar] [CrossRef]
  9. Tursi, A.; Mocci, G.; Allegretta, L.; Aragona, G.; Bianco, M.A.; Colucci, R.; Cuomo, A.; Della Valle, N.; Ferronato, A.; Forti, G.; et al. Comparison of Performances of Adalimumab Biosimilars SB5, APB501, GP2017, and MSB11022 in Treating Patients with Inflammatory Bowel Diseases: A Real-Life, Multicenter, Observational Study. Inflamm. Bowel. Dis. 2022, 28, e145. [Google Scholar] [CrossRef]
  10. Koder, S.; Repnik, K.; Ferkolj, I.; Pernat, C.; Skok, P.; Weersma, R.K.; Potočnik, U. Genetic polymorphism in ATG16L1 gene influences the response to adalimumab in Crohn’s disease patients. Pharmacogenomics 2015, 16, 191–204. [Google Scholar] [CrossRef]
  11. Hlavaty, T.; Persoons, P.; Vermeire, S.; Ferrante, M.; Pierik, M.; Van Assche, G.; Rutgeerts, P. Evaluation of short-term responsiveness and cutoff values of inflammatory bowel disease questionnaire in Crohn’s disease. Inflamm. Bowel. Dis. 2006, 12, 199–204. [Google Scholar] [CrossRef] [PubMed]
  12. Louis, E.; Vermeire, S.; Rutgeerts, P.; De, V.M.; Van Gossum, A.; Pescatore, P.; Fiasse, R.; Pelckmans, P.; Reynaert, H.; D’Haens, G.; et al. A positive response to infliximab in Crohn disease: Association with a higher systemic inflammation before treatment but not with -308 TNF gene polymorphism. Scand. J. Gastroenterol. 2002, 37, 818–824. [Google Scholar] [CrossRef] [PubMed]
  13. Vermeire, S.; Van Gossum, A.; Rutgeerts, P. Laboratory markers in IBD: Useful, magic, or unnecessary toys? Gut 2006, 55, 426–431. [Google Scholar] [CrossRef]
  14. Jostins, L.; Ripke, S.; Weersma, R.K.; Duerr, R.H.; McGovern, D.P.; Hui, K.Y.; Lee, J.C.; Schumm, L.P.; Sharma, Y.; Anderson, C.A.; et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature 2012, 491, 119–124. [Google Scholar] [CrossRef]
  15. Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13, 2498–2504. [Google Scholar] [CrossRef]
  16. Bindea, G.; Mlecnik, B.; Hackl, H.; Charoentong, P.; Tosolini, M.; Kirilovsky, A.; Fridman, W.H.; Pagès, F.; Trajanoski, Z.; Galon, J. ClueGO: A Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 2009, 25, 1091–1093. [Google Scholar] [CrossRef]
  17. Stark, C.; Breitkreutz, B.J.; Reguly, T.; Boucher, L.; Breitkreutz, A.; Tyers, M. BioGRID: A general repository for interaction datasets. Nucleic Acids Res. 2006, 34, D535–D539. [Google Scholar] [CrossRef]
  18. Oughtred, R.; Stark, C.; Breitkreutz, B.J.; Rust, J.; Boucher, L.; Chang, C.; Kolas, N.; O’Donnell, L.; Leung, G.; McAdam, R.; et al. The BioGRID interaction database: 2019 update. Nucleic Acids Res. 2019, 47, D529–D541. [Google Scholar] [CrossRef]
  19. Coutin, N. biogridr: BioGRID R API. 2015. Available online: https://github.com/npjc/biogridr (accessed on 17 October 2022).
  20. Team, R.C. R: A Language and Environment for Statistical Computing. 2020. Available online: https://www.r-project.org/ (accessed on 17 October 2022).
  21. Julià, A.; Rodríguez, J.; Fernández-Sueiro, J.L.; Gratacós, J.; Queiró, R.; Montilla, C.; Torre-Alonso, J.C.; Pérez-Venegas, J.J.; Manrique-Arija, S.; Muñoz-Fernández, S.; et al. PDE3A-SLCO1C1 locus is associated with response to anti-tumor necrosis factor therapy in psoriatic arthritis. Pharmacogenomics 2014, 15, 1763–1769. [Google Scholar] [CrossRef]
  22. Tong, Q.; Zhao, L.; Qian, X.D.; Zhang, L.L.; Xu, X.; Dai, S.M.; Cai, Q.; Zhao, D.B. 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. [Google Scholar] [CrossRef] [Green Version]
  23. Swierkot, J.; Bogunia-Kubik, K.; Nowak, B.; Bialowas, K.; Korman, L.; Gebura, K.; Kolossa, K.; Jeka, S.; Wiland, P. Analysis of associations between polymorphisms within genes coding for tumour necrosis factor (TNF)-alpha and TNF receptors and responsiveness to TNF-alpha blockers in patients with rheumatoid arthritis. Jt. Bone Spine 2015, 82, 94–99. [Google Scholar] [CrossRef] [PubMed]
  24. Schiotis, R.; Sánchez, A.; Escudero, A.; Bartolomé, N.; Szczypiorska, M.; Font, P.; Martínez, A.; Tejedor, D.; Artieda, M.; Mulero, J.; et al. Candidate’s single-nucleotide polymorphism predictors of treatment nonresponse to the first anti-TNF inhibitor in ankylosing spondylitis. Rheumatol. Int. 2014, 34, 793–801. [Google Scholar] [CrossRef] [PubMed]
  25. Bogunia-Kubik, K.; Świerkot, J.; Malak, A.; Wysoczańska, B.; Nowak, B.; Białowąs, K.; Gębura, K.; Korman, L.; Wiland, P. IL-17A, IL-17F and IL-23R Gene Polymorphisms in Polish Patients with Rheumatoid Arthritis. Arch. Immunol. Ther. Exp. 2015, 63, 215–221. [Google Scholar] [CrossRef] [PubMed]
  26. Bank, S.; Andersen, P.S.; Burisch, J.; Pedersen, N.; Roug, S.; Galsgaard, J.; Turino, S.Y.; Brodersen, J.B.; Rashid, S.; Rasmussen, B.K.; et al. Associations between functional polymorphisms in the NFκB signaling pathway and response to anti-TNF treatment in Danish patients with inflammatory bowel disease. Pharmacol. J. 2014, 14, 526–534. [Google Scholar] [CrossRef]
  27. Matsukura, H.; Ikeda, S.; Yoshimura, N.; Takazoe, M.; Muramatsu, M. Genetic polymorphisms of tumour necrosis factor receptor superfamily 1A and 1B affect responses to infliximab in Japanese patients with Crohn’s disease. Aliment. Pharmacol. Ther. 2008, 27, 765–770. [Google Scholar] [CrossRef]
  28. Nishimoto, T.; Seta, N.; Anan, R.; Yamamoto, T.; Kaneko, Y.; Takeuchi, T.; Kuwana, M. A single nucleotide polymorphism of TRAF1 predicts the clinical response to anti-TNF treatment in Japanese patients with rheumatoid arthritis. Clin. Exp. Rheumatol. 2014, 32, 211–217. [Google Scholar]
  29. Sode, J.; Vogel, U.; Bank, S.; Andersen, P.S.; Thomsen, M.K.; Hetland, M.L.; Locht, H.; Heegaard, N.H.; Andersen, V. Anti-TNF treatment response in rheumatoid arthritis patients is associated with genetic variation in the NLRP3-inflammasome. PLoS ONE 2014, 9, e100361. [Google Scholar] [CrossRef]
  30. Duraes, C.; Machado, J.C.; Portela, F.; Rodrigues, S.; Lago, P.; Cravo, M.; Ministro, P.; Marques, M.; Cremers, I.; Freitas, J.; et al. Phenotype-genotype profiles in Crohn’s disease predicted by genetic markers in autophagy-related genes (GOIA study II). Inflamm. Bowel. Dis. 2013, 19, 230–239. [Google Scholar] [CrossRef]
  31. Gregory, A.P.; Dendrou, C.A.; Attfield, K.E.; Haghikia, A.; Xifara, D.K.; Butter, F.; Poschmann, G.; Kaur, G.; Lambert, L.; Leach, O.A.; et al. TNF receptor 1 genetic risk mirrors outcome of anti-TNF therapy in multiple sclerosis. Nature 2012, 488, 508–511. [Google Scholar] [CrossRef]
  32. Plant, D.; Prajapati, R.; Hyrich, K.L.; Morgan, A.W.; Wilson, A.G.; Isaacs, J.D.; Barton, A.; Syndicate, B.i.R.A.G.a.G.S. Replication of association of the PTPRC gene with response to anti-tumor necrosis factor therapy in a large UK cohort. Arthritis Rheum. 2012, 64, 665–670. [Google Scholar] [CrossRef]
  33. Plant, D.; Bowes, J.; Potter, C.; Hyrich, K.L.; Morgan, A.W.; Wilson, A.G.; Isaacs, J.D.; Barton, A.; Consortium, W.T.C.C.; Register, B.S.f.R.B. Genome-wide association study of genetic predictors of anti-tumor necrosis factor treatment efficacy in rheumatoid arthritis identifies associations with polymorphisms at seven loci. Arthritis Rheum. 2011, 63, 645–653. [Google Scholar] [CrossRef] [Green Version]
  34. Hlavaty, T.; Pierik, M.; Henckaerts, L.; Ferrante, M.; Joossens, S.; Van Schuerbeek, N.; Noman, M.; Rutgeerts, P.; Vermeire, S. Polymorphisms in apoptosis genes predict response to infliximab therapy in luminal and fistulizing Crohn’s disease. Aliment. Pharmacol. Ther. 2005, 22, 613–626. [Google Scholar] [CrossRef]
  35. Moroi, R.; Endo, K.; Kinouchi, Y.; Shiga, H.; Kakuta, Y.; Kuroha, M.; Kanazawa, Y.; Shimodaira, Y.; Horiuchi, T.; Takahashi, S.; et al. FCGR3A-158 polymorphism influences the biological response to infliximab in Crohn’s disease through affecting the ADCC activity. Immunogenetics 2013, 65, 265–271. [Google Scholar] [CrossRef]
  36. Jürgens, M.; Laubender, R.P.; Hartl, F.; Weidinger, M.; Seiderer, J.; Wagner, J.; Wetzke, M.; Beigel, F.; Pfennig, S.; Stallhofer, J.; et al. Disease activity, ANCA, and IL23R genotype status determine early response to infliximab in patients with ulcerative colitis. Am. J. Gastroenterol. 2010, 105, 1811–1819. [Google Scholar] [CrossRef]
  37. Lacruz-Guzmán, D.; Torres-Moreno, D.; Pedrero, F.; Romero-Cara, P.; García-Tercero, I.; Trujillo-Santos, J.; Conesa-Zamora, P. Influence of polymorphisms and TNF and IL1β serum concentration on the infliximab response in Crohn’s disease and ulcerative colitis. Eur. J. Clin. Pharmacol. 2013, 69, 431–438. [Google Scholar] [CrossRef]
  38. Taylor, K.D.; Plevy, S.E.; Yang, H.; Landers, C.J.; Barry, M.J.; Rotter, J.I.; Targan, S.R. ANCA pattern and LTA haplotype relationship to clinical responses to anti-TNF antibody treatment in Crohn’s disease. Gastroenterology 2001, 120, 1347–1355. [Google Scholar] [CrossRef]
  39. Aluri, J.; Cooper, M.A.; Schuettpelz, L.G. Toll-Like Receptor Signaling in the Establishment and Function of the Immune System. Cells 2021, 10, 1374. [Google Scholar] [CrossRef]
  40. Biancheri, P.; Pender, S.L.; Ammoscato, F.; Giuffrida, P.; Sampietro, G.; Ardizzone, S.; Ghanbari, A.; Curciarello, R.; Pasini, A.; Monteleone, G.; et al. The role of interleukin 17 in Crohn’s disease-associated intestinal fibrosis. Fibrogenesis Tissue Repair. 2013, 6, 13. [Google Scholar] [CrossRef]
  41. Rovedatti, L.; Kudo, T.; Biancheri, P.; Sarra, M.; Knowles, C.H.; Rampton, D.S.; Corazza, G.R.; Monteleone, G.; Di Sabatino, A.; Macdonald, T.T. Differential regulation of interleukin 17 and interferon gamma production in inflammatory bowel disease. Gut 2009, 58, 1629–1636. [Google Scholar] [CrossRef]
  42. Lee, J.S.; Tato, C.M.; Joyce-Shaikh, B.; Gulen, M.F.; Cayatte, C.; Chen, Y.; Blumenschein, W.M.; Judo, M.; Ayanoglu, G.; McClanahan, T.K.; et al. Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability. Immunity 2015, 43, 727–738. [Google Scholar] [CrossRef]
  43. Wijbrandts, C.A.; van Leuven, S.I.; Boom, H.D.; Gerlag, D.M.; Stroes, E.G.; Kastelein, J.J.; Tak, P.P. Sustained changes in lipid profile and macrophage migration inhibitory factor levels after anti-tumour necrosis factor therapy in rheumatoid arthritis. Ann. Rheum. Dis. 2009, 68, 1316–1321. [Google Scholar] [CrossRef] [PubMed]
  44. Radstake, T.R.; Sweep, F.C.; Welsing, P.; Franke, B.; Vermeulen, S.H.; Geurts-Moespot, A.; Calandra, T.; Donn, R.; van Riel, P.L. Correlation of rheumatoid arthritis severity with the genetic functional variants and circulating levels of macrophage migration inhibitory factor. Arthritis Rheum 2005, 52, 3020–3029. [Google Scholar] [CrossRef] [PubMed]
  45. Mielants, H.; Veys, E.M.; Cuvelier, C.; De Vos, M.; Goemaere, S.; De Clercq, L.; Schatteman, L.; Elewaut, D. The evolution of spondyloarthropathies in relation to gut histology. II. Histological aspects. J. Rheumatol. 1995, 22, 2273–2278. [Google Scholar] [PubMed]
  46. Rudwaleit, M.; Baeten, D. Ankylosing spondylitis and bowel disease. Best Pr. Res. Clin. Rheumatol. 2006, 20, 451–471. [Google Scholar] [CrossRef]
  47. Brown, M.A.; Kenna, T.; Wordsworth, B.P. Genetics of ankylosing spondylitis--insights into pathogenesis. Nat. Rev. Rheumatol. 2016, 12, 81–91. [Google Scholar] [CrossRef]
  48. Ossum, A.M.; Palm, Ø.; Lunder, A.K.; Cvancarova, M.; Banitalebi, H.; Negård, A.; Høie, O.; Henriksen, M.; Moum, B.A.; Høivik, M.L.; et al. Ankylosing Spondylitis and Axial Spondyloarthritis in Patients With Long-term Inflammatory Bowel Disease: Results From 20 Years of Follow-up in the IBSEN Study. J. Crohns Colitis 2018, 12, 96–104. [Google Scholar] [CrossRef]
  49. Sandborn, W.J.; Hanauer, S.B.; Katz, S.; Safdi, M.; Wolf, D.G.; Baerg, R.D.; Tremaine, W.J.; Johnson, T.; Diehl, N.N.; Zinsmeister, A.R. Etanercept for active Crohn’s disease: A randomized, double-blind, placebo-controlled trial. Gastroenterology 2001, 121, 1088–1094. [Google Scholar] [CrossRef]
Figure 1. IBDQ value and difference in the IBDQ value (delta IBDQ) during the 30-week treatment period according to a genotype of SNP rs2275913 in the IL17A gene.
Figure 1. IBDQ value and difference in the IBDQ value (delta IBDQ) during the 30-week treatment period according to a genotype of SNP rs2275913 in the IL17A gene.
Genes 14 00452 g001
Figure 2. IBDQ values and the difference in the IBDQ values (delta IBDQ) during the 30-week treatment period according to the genotype of SNP rs755622 in the MIF gene.
Figure 2. IBDQ values and the difference in the IBDQ values (delta IBDQ) during the 30-week treatment period according to the genotype of SNP rs755622 in the MIF gene.
Genes 14 00452 g002
Figure 3. IBDQ value and the difference in the IBDQ value (delta IBDQ) during the 30-week treatment period according to the genotype of the SNP rs3740691 in the gene ARFGAP2.
Figure 3. IBDQ value and the difference in the IBDQ value (delta IBDQ) during the 30-week treatment period according to the genotype of the SNP rs3740691 in the gene ARFGAP2.
Genes 14 00452 g003
Figure 4. Genes of interest and their interacting genes. The green hexagonal nodes represent genes of interest, also marked with * after the gene name. The pink rectangles represent interacting genes obtained from BIOGRID.
Figure 4. Genes of interest and their interacting genes. The green hexagonal nodes represent genes of interest, also marked with * after the gene name. The pink rectangles represent interacting genes obtained from BIOGRID.
Genes 14 00452 g004
Figure 5. TNF-independent pathways of non-response in gut epithelial cells. Genes with variants associated with response are outlined. (a) In the presence of aberrant TNF signaling in IBD, the TNFR1-associated pathway will induce excessive apoptosis or necroptosis. (b) During anti-TNF drug therapy, apoptosis or necroptosis (but also NF-κB pathways) may still be induced independent of TNF via TRIF-dependent TLR3 (or TLR4 [39]) signaling [2]. Deleterious changes in TRIF-dependent Toll-like receptor signaling may lead to molecular pathology of CD, which is less reliant on TNF expression alone, and, thus, resistant to TNF treatment.
Figure 5. TNF-independent pathways of non-response in gut epithelial cells. Genes with variants associated with response are outlined. (a) In the presence of aberrant TNF signaling in IBD, the TNFR1-associated pathway will induce excessive apoptosis or necroptosis. (b) During anti-TNF drug therapy, apoptosis or necroptosis (but also NF-κB pathways) may still be induced independent of TNF via TRIF-dependent TLR3 (or TLR4 [39]) signaling [2]. Deleterious changes in TRIF-dependent Toll-like receptor signaling may lead to molecular pathology of CD, which is less reliant on TNF expression alone, and, thus, resistant to TNF treatment.
Genes 14 00452 g005
Table 1. Summary of clinical data.
Table 1. Summary of clinical data.
Number of patients102
Average age at enrollment41.1
Average age at diagnosis27.4
SexMale43
Female59
Previously required
operative procedure
Yes58
No44
Disease locationL118
L232
L352
L42
Disease behaviorB135
B228
B335
B2 + B34
Perianal manifestations7
Concurrent
drug use
5-aminosalicylic acid47
Corticosteroids36
Azathioprine or
6-mercaptopurine
31
SmokingYes38
No64
Average IBDQ valueWeek 0152.42
Week 4169.48
Week 12173.36
Week 20175.08
Week 30175.43
Blood CRPWeek 019.48
Week 412.42
Week 1211.15
Week 2010.60
Week 3012.55
Table 2. Summary of pharmacogenetic anti-TNF signals.
Table 2. Summary of pharmacogenetic anti-TNF signals.
Disease 1SNP IDChrGeneStudyCorrelation (Genotype x Response)p Value, OR
PArs379427112PDE3A[21]G—nonresponsep = 0.0036
CD, AS, SpArs18006296TNF[22]G—responsep = 0.0007
CD, AS, SpA, RArs17997246TNF[22,23]C—responsep = 0.010
CD, AS, SpA, RArs17997246TNF[22,23]TT—responsep = 0.045
ASrs9179972IL18RAP[24]A—nonresponsep = 0.007
ASrs75562222MIF[24]G—nonresponsep = 0.019
ASrs18008961IL10[24]A—nonresponsep = 0.041
ASrs374069111ARFGAP2[24]A—nonresponsep = 0.002
ASrs10616221TNFRSF1B[24]G—nonresponsep = 0.048
IBD, RArs22759136IL17A[25,26]GG—nonresponse, GA or AA—nonresponsep = 0.040
RA, IBDrs22759136IL17A[25,26]GA or AA— nonresponsep = 0.050
RA, CDrs76745512TNFRSF1A[23,27]AA—responsep = 0.040
RArs37618479TRAF1[28]GG—nonresponseOR = 7.4
RArs46126661NLRP3[29]T—nonresponsep = 0.025
IBD, RArs243056112IFNG[26,29]TA or AA— response, A—nonresponsep = 0.005
PED-IBDrs283687821BRWD1[4]GG—nonresponsep = 0.030
PED-IBDrs21889625IRF1-AS1[4]CC—nonresponsep = 0.028
PED-IBDrs69084256CDKAL1[4]CT or TT—nonresponsep = 0.049
PED-IBDrs23951856HLA-DQA1[4]GG—nonresponsep = 0.007
PED-IBDrs22418802ATG16L1[4,30]AA or AG—nonresponsep = 0.048
PED-IBDrs22418802ATG16L1[4,30]GG—responsep = 0.010
PED-IBDrs76242121ICOSLG[4]AA—nonresponsep = 0.045
MSrs180069312TNFRSF1A[31]G—nonresponseNA
CDrs18167024TLR2[26]CT or TT—responsep = 0.040
IBDrs38040994TLR2[26]CT or CC—responsep = 0.010
IBDrs119382284TLR2[26]AC or AA—nonresponsep = 0.040
IBDrs50307289TLR4[26]AG or AA—responsep = 0.010
IBDrs15549739TLR4[26]CT or CC—nonresponsep = 0.040
IBDrs1870843TLR9[26]CT—responsep = 0.040
IBDrs3521393TLR9[26]AA—nonresponsep = 0.040
IBDrs114659968LY96[26]CG or GG—responsep = 0.010
IBDrs722209417MAP3K14[26]CT–responsep = 0.050
IBDrs3615256TNF[26]AG—nonresponsep = 0.040
IBDrs414957012TNFRSF1A[26]TT—responsep = 0.030
IBDrs69271726TNFAIP3[26]CG or GG—nonresponsep = 0.030
IBDrs48483062near IL1B[26]AG or GG—responsep = 0.040
IBDrs104995637near IL6[26]CT or CC—responsep = 0.010
UCrs46964804TLR2[26]TT—nonresponsep = 0.040
UCrs25691905CD14[26]AG or AA—nonresponsep = 0.040
UCrs42519612IL1RN[26]CT or CC—nonresponsep = 0.040
CDrs22749101ITLN1[30]CC—responsep = 9.60 × 10−3
CDrs133611895IRGM[30]CC—responsep = 5.00 × 10−3
RArs109195631PTPRC[32]A—nonresponsep = 0.030
RArs120817651intergenic[33]A—nonresponsep = 7.39 × 10−4
RArs15322695PDZD2[33]G—nonresponsep = 7.37 × 10−4
RArs173012496EYA4[33]C—responsep = 5.67 × 10−5
RArs730564612intergenic[33]T—responsep = 1.47 × 10−4
RArs46948904TEC[33]C—responsep = 6.47 × 10−3
RArs135094811intergenic[33]A—nonresponsep = 8.64 × 10−3
RArs796231612LINC01619[33]G—nonresponsep = 2.05 × 10−2
RArs441159118LINC01387, LOC100130480[6]C—responsep = 5.14 × 10−5
RArs77670696LOC102723883[6]A—nonresponsep = 8.34 × 10−5
RArs46513701near PLA2G4A[6]A—responsep = 1.09 × 10−4
RArs181344311CNTN5[6]C—nonresponsep = 1.37 × 10−4
RArs14477223intergenic[6]C—responsep = 1.62 × 10−4
RArs15688857intergenic[6]A—responsep = 1.69 × 10−4
RArs121426231near PLA2G4A[6]A—responsep = 2.04 × 10−4
RArs237894514NUBPL[6]A—nonresponsep = 6.88 × 10−4
RArs64275281CD84[5]GA or AA—responsep = 8.00 × 10−8
CDrs46459831CASP9[10,34]C—nonresponsep = 0.040
CDrs7631101FASLG[34]C—responsep = 0.002
CDrs3969911FCGR3A[35]GG—responsep < 0.001
UCrs10048191IL23R[36]AA—responseOR = 1.27
UCrs22018411IL23R[36]GG—responseOR = 1.21
UCrs108896771IL23R[36]AA—responseOR = 1.26
UCrs112090321IL23R[36]AA—responseOR = 1.15
UCrs14959651IL23R[36]CC—responseOR = 1.29
UCrs75178471IL23R[36]GG—nonresponseOR = 0.76
UCrs104896291IL23R[36]CC—nonresponseOR = 0.81
UCrs114658041IL23R[36]GG—nonresponseOR = 0.77
UCrs13431511IL23R[36]AA—nonresponseOR = 0.85
CDrs10616241TNFRSF1B[27]A—T haplotype—nonresponsep = 0.010
CDrs33971TNFRSF1B[27]A—T haplotype—nonresponsep = 0.010
CDrs102103022ATG16L1[10]T—responsep = 8.11 × 10−4
CDrs11436342IL1B[37]C—nonresponsep = 0.027
CDrs9092536LTA[38]Homozygotes for the LTA NcoI-TNFc-aa13L-aa26 haplotype 1-1-1-1—nonresponsep = 0.007
1 PA = psoriatic arthritis, RA = rheumatoid arthritis, IBD = inflammatory bowel disease, CD = Crohn’s disease, MS = multiple sclerosis, AS = ankylosing spondylitis, SpA = spondyloarthritis.
Table 3. SNPs associated with either biological or clinical response to ADA treatment in CD patients.
Table 3. SNPs associated with either biological or clinical response to ADA treatment in CD patients.
GeneSNP IDChrp ValueNonresponse Association
Timeframe (Week)
Our Analyzed Nonresponse GenotypeReported
Nonresponse
Genotype
IL17Ars227591369.73 × 10−320AA or AGGG in RA,
AA or AG in IBD
MIFrs755622224.00 × 10−34CC or CGG in AS
ARFGAP2rs3740691111.24 × 10−54GGA in AS
TLR2rs469648041.48 × 10−320AATT in UC
ICOSLG (B7RP1)rs762421212.11 × 10−34AA or AGAA in PED-IBD
CD14rs256919058.00 × 10−3
2.60 × 10−2
12
30
AA or AG
AG or GG
AA or AG in UC
TNFars36152562.79 × 10−34AA or AGAG in IBD
CDKAL1rs690842561.73 × 10−230CT or TTCT or TT in PED-IBD
TNFAIP3rs692717263.87 × 10−24CCCG or GG in IBD
TNFRSF1Ars767455123.94 × 10−230CCCC in CD and RA
IL1RNrs425196123.93 × 10−230CCCC or CT in UC
TNFars180062966.15 × 10−34GGAA in CD, AS, SpA
TNFRSF1Ars1800693122.90 × 10−24CCC in MS
TNFRSF1Brs106162213.25 × 10−212GGGG in AS
PTPRCrs1091956313.27 × 10−212AA or AGAA in RA
TLR2rs1193822849.82 × 10−320CCAA or AC in IBD
IRGMrs1336118954.45 × 10−212CC or CTTT in CD
TLR4rs155497391.36 × 10−230CC or CTCC or CT in IBD
IL10rs180089611.22 × 10−24CC or CTT in AS
ITLN1rs227491014.79 × 10−24CT or TTTT in CD
IFNGrs2430561123.73 × 10−24AAA in RA
TT in IBD
TLR9rs35213931.98 × 10−220AAAA in IBD
TRAF1rs376184792.68 × 10−212AAGG in RA
PDE3Ars3794271124.40 × 10−24TTG in PA
TLR2rs380409943.89 × 10−220CCTT in IBD
NLRP3rs461266613.00 × 10−220CCT in RA
near IL1Brs484830622.23 × 10−24GGAA in IBD
MAP3K14rs7222094173.59 × 10−24CCCT in IBD
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Jezernik, G.; Gorenjak, M.; Potočnik, U. MIF Variant rs755622 Is Associated with Severe Crohn’s Disease and Better Response to Anti-TNF Adalimumab Therapy. Genes 2023, 14, 452. https://doi.org/10.3390/genes14020452

AMA Style

Jezernik G, Gorenjak M, Potočnik U. MIF Variant rs755622 Is Associated with Severe Crohn’s Disease and Better Response to Anti-TNF Adalimumab Therapy. Genes. 2023; 14(2):452. https://doi.org/10.3390/genes14020452

Chicago/Turabian Style

Jezernik, Gregor, Mario Gorenjak, and Uroš Potočnik. 2023. "MIF Variant rs755622 Is Associated with Severe Crohn’s Disease and Better Response to Anti-TNF Adalimumab Therapy" Genes 14, no. 2: 452. https://doi.org/10.3390/genes14020452

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

Article Metrics

Back to TopTop