MASARYK UNIVERSITY FACULTY OF MEDICINE Modern approaches in diagnostics of inflammatory bowel disease in a pediatric population HABILITATION THESIS In Pediatrics (Collection of previously published scholarly works) Brno 2021 Petr Jabandžiev, MD, Ph.D. 2 I hereby declare that I wrote this habilitation thesis on my own, using the relevant resources listed in the references. .......................... Signature 3 Acknowledgment I would like to thank very sincerely all my mentors who stimulated my interest in research questions, namely Prof. Ondřej Slabý and Prof. Ajay Goel. Many thanks are due to my research collaborators and colleagues from the Department of Pediatrics, University Hospital Brno; Faculty of Medicine, Masaryk University; and Central European Institute of Technology. Last but not least, I would like to express my sincere thanks to my whole family, my always supportive wife Hana, and our beloved children. 4 Commentary Inflammatory bowel disease (IBD) is an umbrella term describing disorders that cause chronic inflammation of the gastrointestinal tract. IBD affects both children and adults, but children have their specificities. IBD is classically divided into Crohn’s disease and ulcerative colitis. In the current view, pediatric IBD encompasses a continuum of clinical categories, including typical ulcerative colitis, atypical ulcerative colitis, Crohn’s colitis, and Crohn’s disease. Where none of the above can be determined, we refer to inflammatory bowel disease unclassified. The incidence of IBD has been increasing in recent decades worldwide, and IBD has moved from being previously at the periphery of interest among gastroenterologists into focus as one of the most studied diseases. The pathogenesis of IBD involves an interaction of genetic and environmental variables that disrupt the relationship between the immune system and the gut microbiota. The clinical manifestations of IBD can be not only intestinal but also extraintestinal, and more aggressive forms of the disease are often present in children compared to adults. The course of IBD is unpredictable, with alternating periods of flare-up and remission. IBD in childhood can disrupt somatic and psychological development and has relevant social and economic consequences. Therefore, accurate and timely diagnosis and adequate therapy are essential. The presented habilitation thesis documents the author’s experience in managing pediatric patients with various gastroenterological problems, but at its core is a focus on IBD and description of modern diagnostic approaches. This habilitation thesis is conceived as a collection of 10 articles previously published by the author and his colleagues. It contains individual chapters dealing with the basic aspects of IBD. Where relevant, it is followed by commentaries introducing the topic of each publication, describing the current state of knowledge and how the author has contributed to knowledge in this field. The work is based on research activities at the authors’ workplaces, the Department of Pediatrics, University Hospital Brno; the Faculty of Medicine, Masaryk University; and Central European Institute of Technology. 5 Content of the work Acknowledgment ....................................................................................................................... 3 Commentary............................................................................................................................... 4 1. Introduction ............................................................................................................................ 7 2. Classification.......................................................................................................................... 9 2.1. Types of IBD ................................................................................................................... 9 2.2. Disease location............................................................................................................... 9 2.3. Age of onset................................................................................................................... 11 2.4. Authors’ contribution to the knowledge........................................................................ 11 3. Epidemiology ....................................................................................................................... 14 3.1. Authors’ contribution to the knowledge........................................................................ 15 4. Etiopathogenesis................................................................................................................... 17 4.1. Intestinal barrier and immune response......................................................................... 18 4.2. Environment .................................................................................................................. 18 4.3. Genetics ......................................................................................................................... 19 4.4. Authors’ contribution to the knowledge........................................................................ 19 5. Diagnostics........................................................................................................................... 22 5.1. Patient’s history, symptoms, and clinical examination ................................................. 23 5.2. Laboratory tests ............................................................................................................. 23 5.3. Endoscopic examination................................................................................................ 24 5.4. Histomorphologic examination ..................................................................................... 25 5.5. Imaging methods ........................................................................................................... 25 5.6. Examples of modern diagnostic methods potentially useful in clinical practice........... 25 5.7. Authors’ contribution to the knowledge........................................................................ 27 6. Differential diagnostics ........................................................................................................ 30 6.1. Very-early-onset IBD .................................................................................................... 31 6.2. Authors’ contribution to the knowledge........................................................................ 33 7. Therapy................................................................................................................................. 38 7.1. Medical therapy ............................................................................................................. 38 7.2. Nutritional therapy......................................................................................................... 40 7.3. Endoscopic and surgical therapy ................................................................................... 40 7.4. Authors’ contribution to the knowledge........................................................................ 40 8. Complications....................................................................................................................... 42 8.1. Complications from within the gastrointestinal tract .................................................... 42 6 8.2. Extraintestinal and systemic complications................................................................... 43 8.3. Authors’ contribution to the knowledge........................................................................ 43 9. Potential perspectives on further research............................................................................ 45 9.1. General perspectives on research in the field of IBD.................................................... 45 9.2. Author’s perspectives on research in the field of IBD .................................................. 46 10. Conclusions ........................................................................................................................ 52 List of abbreviations................................................................................................................. 53 List of Figures .......................................................................................................................... 55 List of Tables............................................................................................................................ 56 References ................................................................................................................................ 57 List of Annexes ........................................................................................................................ 80 7 1. Introduction Inflammatory bowel disease (IBD) is a term encompassing a range of chronic inflammatory conditions affecting the gastrointestinal tract and is traditionally subdivided into Crohn’s disease (CD) and ulcerative colitis (UC).1 CD is an inflammatory disease affecting any part of the gastrointestinal tract from the oral cavity to the rectum, but the most typical site of involvement is the terminal ileum. Clinical manifestations of CD in children are varied and may include fatigue, weight loss, abdominal pain, diarrhea, growth impairment, and delayed puberty.2 UC is characterized by inflammatory involvement of the colon. Typical symptoms depend on the extent of the disease and include crampy abdominal pain and diarrhea, often with blood.3 According to various sources, up to one-quarter of patients with IBD are diagnosed in childhood or adolescence.4, 5 The incidence of IBD has been increasing in recent decades in all populations (pediatric and adult) and worldwide.6, 7 The causes of this phenomenon are not clearly understood and this is probably one of the reasons why IBD has moved from the periphery of interest among gastroenterologists to one of the most studied diseases.8 The pathogenesis of IBD involves an interaction of genetic and environmental variables that disrupt the relationship between the immune system and the gut microbiota.9 There are significant differences between the child and adult populations, and patients diagnosed during childhood have differential characteristics (e.g., higher prevalence of CD, more extensive disease, and more frequent family history).10-13 In addition, the use of immunomodulators and biologic agents is greater in childhood‐onset patients, suggesting a more aggressive course of the disease.14 The course of IBD is unpredictable, with alternating periods of flare-up and remission. The variety of symptoms, both intestinal and extraintestinal; the presence of complications; often challenging diagnostic procedures; and the therapy itself, including potential side effects, make the disease challenging for young patients and their families. Proper somatic development may be impaired, while quality of life, social life, and education also are affected.15 The main elements of diagnostic workup include upper and lower endoscopy with mucosal biopsies and imaging procedures, and, in the differential diagnosis, it is necessary to exclude many IBD-mimicking diseases.16 Accurate and timely diagnosis and adequate therapy are essential, because diagnostic delay is associated with complicated disease course.17, 18 Therapeutic options include medication, nutritional therapy, and surgery.19-22 The goal of IBD treatment is achieving deep remission of the disease, which means elimination of the patient’s symptoms, normalization of laboratory parameters, induction of mucosal healing, restoration of normal growth, and prevention of 8 surgical complications.23 Thus, an interdisciplinary approach involving different health professions is essential in the management of patients with such complex medical problems.24 Moreover, the professionals caring for these patients must have a broad understanding of the topic because there is increasing interest in such additional treatment modalities as complementary and alternative medicine.25-27 9 2. Classification 2.1. Types of IBD Even though IBD in children and adults is similar in many ways, pediatric IBD (PIBD) often has atypical features making classification of PIBD challenging.11, 28, 29 Moreover, accurate phenotype classification of IBD is crucial for appropriate management and prognostication.30 Consensus-based criteria for the diagnosis of PIBD were published in 2005.31 These included a defined diagnostic work-up for new PIBD patient that enabled standardized diagnostics for CD, UC, and indeterminate colitis.31 This was an important achievement for both clinical practice and clinical research. In 2014, the revised Porto criteria were released and identified subtypes of PIBD as UC, atypical UC (atypical phenotypes such as macroscopic rectal sparing, isolated non-serpiginous gastric ulcers, normal crypt architecture, absence of chronicity in biopsies, or a cecal patch.), IBD unclassified (IBDU; inflammation is limited to the colon with features that make differentiation between UC and CD uncertain even after a complete workup), and CD.16 Consequently, the Paediatric IBD Porto Group of the European Society of Paediatric Gastroenterology, Hepatology and Nutrition (ESPGHAN) published the ‘‘PIBD-Classes’’ criteria that standardized the differentiation of PIBD into five categories: typical UC, atypical UC, IBDU, Crohn’s colitis, and CD.32 2.2. Disease location The previously used Montreal classification for disease characterization had several weaknesses regarding the classification of children.33, 34 For this reason, an international group of pediatric IBD experts developed evidence-based consensus recommendations for a pediatric modification of the Montreal classification (the so-called Paris classification) in an effort to facilitate research in PIBD and create uniform standards for defining IBD phenotypes.35 Table 1 and Table 2 summarize Paris classification for CD and UC, respectively.35 10 Table 1. Paris classification for Crohn’s disease. Age at diagnosis A1a: 0–<10 years A1b: 10–<17 years A2: 17–40 years A3: >40 years Location L1: distal 1/3 ileum ± limited cecal disease L2: colonic L3: ileocolonic L4a: upper disease proximal to ligament of Treitz  L4b: upper disease distal to ligament of Treitz and proximal to distal 1/3 ileum Behavior B1: nonstricturing nonpenetrating B2: stricturing B3: penetrating B2B3: both penetrating and stricturing disease, either at the same or different times p: perianal disease modifier Growth G0: no evidence of growth delay G1: growth delay Table 2. Paris classification for ulcerative colitis. Extent E1: ulcerative proctitis E2: left-sided UC (distal to splenic flexure) E3: extensive (hepatic flexure distally) E4: pancolitis (proximal to hepatic flexure) Severity S0: never severe S1: ever severe It should be added that this classification has more use in research than in clinical practice. Nevertheless, the Paris classification for PIBD could, for example, have a predictive value for long-term worse outcome.36 11 2.3. Age of onset PIBD patients can also be classified in terms of age. Montreal classification distinguished pediatric-onset IBD as cases diagnosed before 17 years of age and labeled them A1.33 In the Paris classification,35 A1 was further divided into A1a (diagnosed before 10 years of age) and A1b (diagnosed between 10 and 17 years of age). Among A1a cases, those diagnosed before 6 years of age are classified as very-early-onset IBD (VEO-IBD).37 Furthermore, IBD cases with onset by 2 years of age were classified as infantile-IBD and by 28 days of age as neonatal-onset IBDs, respectively.37, 38 Now, therefore, we have subgroups of pediatric-onset IBD (<17 years), early-onset IBD (<10 years), VEO-IBD (<6 years), infantile-onset IBD (<2 years), and neonatal-onset IBD (<28 days).37-39 IBD generally is regarded as a polygenic disease, and a genome-wide association study has revealed more than 240 known disease-associated genes.40 Pediatric-onset IBD seems to have a greater genetic component compared to adult-onset IBD due to lower cumulative exposure to environmental factors.41 In pediatrics, the name IBD is also used for a group of diseases that have similar symptomatology but are caused by rare genetic defects. Other diseases that involve the symptoms of IBD have a separate name, often based on molecular classification, and their manifestations are referred to as “IBD-like.”38, 39 Rare monogenic IBD and IBD-like syndromes therefore present the genetic extreme where a single highly pathogenic variant causes IBD symptoms. As such, pediatric-onset IBD represents a spectrum ranging from extreme monogenic variants (typically VEO-IBD) to adolescent complex variants (as in adults).41 Evaluating and managing children with VEO-IBD is a challenging field for pediatric gastroenterologists worldwide.37, 42 Specifics in diagnostics of VEO-IBD will be further described in another chapter. 2.4. Authors’ contribution to the knowledge My colleagues and I described a unique diagnosis of trichohepatoenteric syndrome (THES) (Annex 1). To our knowledge, this was the first case diagnosed in the Czech Republic. THES can have a clinical presentation similar to that of VEO-IBD and is often assigned to this group (or to VEO-IBD-like). The described patient had somatic retardation and woolly hair appearance and suffered from recurring episodes of watery mucous diarrhea, impaired liver functions, and failure to thrive (Figure 1).42 12 Figure 1. Clinical data of the patient with trichohepatoenteric syndrome. (A) Overall habitus of the patient. (B) Detail of the hairy part of the head. Brittle, easily breakable hair. (Ci) Hair analysis using light microscopy showing trichorrhexis nodosa. Magnification 40×. (Cii) Bristly split hair ends. Magnification 40×. The figure had been published in Jabandziev et al.42 Esophagogastroduodenoscopy and colonoscopy were performed at 4 years to rule out possible IBD, but only signs of nonspecific colitis were evident. Massive parallel sequencing 13 targeting a panel of primary immunodeficiency-related genes was used to examine the patient’s DNA. Next-generation sequencing (NGS) analysis revealed two heterozygous variants in the TTC37 gene. Nonsense p.Arg1201* and missense p.Leu1505Ser variants in exons 34 and 42, respectively, were evaluated as pathogenic based on in silico predictions, their rare occurrence in the general population, and the fact that both mutations had already been described in patients with THES. THES could be a life-threatening condition, particularly in children who develop liver disease or severe infection courses, and it must be considered in the differential diagnosis in children for whom a diagnosis of VEO-IBD is being considered.42 14 3. Epidemiology IBD has undoubtedly become a global disease.43, 44 Depending upon individual regions, we can find regions with low, medium, and high incidences.7 Differences in incidence could also be found by race45 and by age (children vs. adults).11 There is a general consensus within the gastroenterologist community that the incidence of IBD is increasing worldwide,7, 44 and especially in low-income countries.46, 47 With these trends, a significant burden on health systems and economic impacts can be expected.48-50 It is not easy to compare and standardize data from different parts of the world, however, due to heterogeneity among study designs. Diagnostic criteria can differ. Some studies use hospital records, while others use surveys and administrative data.51 The age limit is a vital inclusion criterion with a significant impact on reported incidence rates, as individual studies differ significantly in their definitions of childhood or upper age limit (15, 18, or 20 years respectively).6 Most data are retrospective; only a few prospective population-based studies have been conducted, those being particularly from developing countries. Furthermore, the incidence rates are often an extrapolation from one or more regions of a country.51 It cannot be ruled out that better diagnostics of IBD is at least partly responsible for the increase in incidence.52 In connection with the differences in incidence in individual countries, a so-called north–south gradient is discussed, with the highest incidence of IBD being found in the north.4 In adult patients, a positive association with a country’s wealth (as measured by gross domestic product) has been discussed.53, 54 It is also necessary to mention the fact that we do not have recent data from a large number of countries, and most countries lack accurate estimates for the incidence of pediatric IBD.51, 55 The most recent systematic review studying the incidence of IBD in the pediatric population worldwide was published in 2018,6 and it is interesting to compare that with the previous systematic review from 2011.4 The authors of the 2011 systematic review stated that the incidence of CD was increasing especially in some countries and, conversely, that the incidence of UC remained the same in most countries.4 In 2018, Sykora et al. reviewed 140 studies reporting data from 38 countries.6 The highest annual pediatric incidences of IBD were 23/100,000 person-years in Europe; 15.2/100,000 in North America; and 11.4/100,000 in Asia, the Middle East, and Oceania. The highest annual incidences of CD were 13.9/100,000 in North America and 12.3/100,000 in Europe. The highest annual incidences of UC were 15.0/100,000 in Europe and 10.6/100,000 in North America. The highest annual incidences of IBD-U were 3.6/100,000 in Europe and 2.1/100,000 in North America. Overall, 15 67% of CD, 46% of UC, and 11% of IBD-U studies reported increasing incidence in the trend analyses.6, 56 From a general perspective, it is not very clear how incidence trends will continue to evolve. It is questionable whether, for example, developed countries are still experiencing further increase in incidence. In a recently published systemic review and metaanalysis, Roberts et al. stated that the incidence of pediatric IBD continues to increase throughout Europe, with more robust evidence of a north–south than of an east–west gradient.57 3.1. Authors’ contribution to the knowledge Until recently, we did not have much information about the epidemiology of IBD in children within the Czech Republic. The first Czech national survey was conducted and published by Pozler et al. in 2006.58 This paper described how the incidence of CD had increased from 0.25/100,000 in 1990 to 1.25/100,000 in 2001. Schwarz et al.59 rather more recently (2017) published a 16-year prospective study of pediatric IBD patients in the Pilsen Region of the Czech Republic showing that a group of 170 pediatric patients (study period 2000–2015) represented an average incidence of IBD per 100,000/year of 10.0 (6.2 for CD, 2.8 for UC, and 1.0 for IBD-U).56, 59 Our study (Annex 2) aimed to determine the incidence and trends of IBD in the population of Czech children.56 The study included a 16-year period in the South Moravian Region. It characterized differences by sex and age and projected the incidence of IBD for future years. Overall, 358 pediatric patients with newly diagnosed IBD at the Department of Pediatrics, University Hospital Brno, were evaluated. Diagnosed were 192 children (53.6%) with CD, 123 (34.4%) with UC, and 43 (12.0%) with IBD-U. The incidence of IBD increased from 3.8 (CD 2.9, UC 0.9, and IBD-U 0.0) per 100,000/year in 2002 to 14.7 (CD 9.8, UC 4.0, and IBD-U 0.9) per 100,000/year in 2017 (p<0.001) (Figure 2). The overall IBD incidence per 100,000/year was 9.8 (95% confidence interval [CI]: 8.8–10.9). Constituent incidences per 100,000/year were CD 5.2 (95% CI: 4.5–6.0), UC 3.4 (95% CI: 2.8–4.0), and IBD-U 1.2 (95% CI: 0.9–1.6). IBD incidence was projected to reach 18.9 per 100,000/year in 2022 (Figure 2). The incidence of CD, UC, and U-IBD found in our study, including the proportions among them, were practically identical to the results from Schwarz et al.56, 59 Both studies showed that the overall incidence of IBD is increasing in Czech children, and especially the incidence of CD, while the trends for UC and IBD-U seem to be constant. 16 Despite some limitations, we would therefore suggest that our results are potentially similar to the incidence to be found in pediatric IBD patients across the Czech Republic.56 Figure 2. Incidence rates per 100,000/year among children (0–18 years of age) newly diagnosed with IBD (A) and CD (B) in South Moravian Region, 2002 to 2017 and 2018 to 2022. Black points represent actual data. Broken blue lines indicate trend over the observed period based on Poisson regression along with a 95% confidence interval. Blue points are future projections. The figure had been published in Jabandziev et al.56 These results therefore suggest that incidence rates of pediatric IBD and its subtypes in the Czech Republic are among the highest in the literature. Moreover, these data further emphasize the need to identify risk factors that contribute to the increasing incidence of IBD.56, 60 17 4. Etiopathogenesis Knowing IBD’s pathogenesis is key to understanding the causes of IBD, and a better understanding of these processes could lead to the development of novel IBD therapies.61 Despite significant achievements, we do not yet have a comprehensive theory describing all the pathogenetic processes leading to IBD. There is growing evidence, however, that the pathogenesis of IBD results from an interplay among host genetic susceptibility, environmental factors, immunological abnormalities, and intestinal barrier alteration (Figure 3).62, 63 Figure 3. Mechanisms involved in the pathogenesis of inflammatory bowel disease. Figure was prepared in accordance with Ramos et al.63 and created in collaboration with the Service Center for E-Learning at Masaryk University, Faculty of Informatics. 18 4.1. Intestinal barrier and immune response The mucosal barrier and its proper functioning, are essential in protecting against adhesion and invasion of luminal microorganisms.64 This is made possible by intact epithelium, synthesis of various antimicrobial peptides, and mucus layer formation.65 Damages to the intestinal mucosal barrier include, among others, defective production of antimicrobial peptides, changes in the thickness or composition of the intestinal mucus layer, changes in pattern recognition receptors, and defects in the autophagy process.66 The intestinal epithelium is in a functional equilibrium with the luminal contents. Disturbance of this equilibrium can lead to dysbiosis and further to the development of a pathological condition such as IBD.67, 68 In CD patients, for example, there have been described abnormal intestinal permeability; abnormities of tight junctions;69 and dysfunction of goblet cells, Paneth cells,70 and M cells.63 Upon encountering antigen and microbial products gaining access through the intestinal barrier, dendritic cells and other antigen-presenting cells initiate a cascade of proand anti-inflammatory signals.71 This results in the activation of various subsets of local and circulating lymphocytes migrating to effector sites where inflammation occurs.63 All these processes influence the innate response and production of various cytokines while affecting the function of effector and regulatory cells. Dysregulated immunological reactions in the gut that lead to imbalance in pro- and anti-inflammatory pathways involved in innate and adaptive immunity are regarded as fundamental to the development and persistence of inflammation in IBD.72 4.2. Environment The available evidence suggests that environmental exposures have variable effects on individuals with IBD.73 In a study by Elten et al., for example, greater exposure to residential greenspace during childhood was associated with reduced IBD risk, thus suggesting a novel avenue to IBD prevention in children.74 The same study group revealed associations between maternal and early-life exposures to air pollutants and risk of pediatric-onset IBD diagnosis.75 In a national case-control study in Sweden, higher cumulative exposure to systemic and particularly broad-spectrum antibiotic therapy was associated with greater risk of new-onset IBD and its subtypes.76 Moreover, greater adherence to a Mediterranean diet was associated with significantly lower risk of later-onset CD in two prospective studies by Khalili et al.77 Among environmental factors, diet is widely thought to play a crucial role in IBD 19 development. A positive explanation could be that dietary habits have an essential role in defining the composition of the human gut microbiota and patients suffering from IBD show a generalized decrease in bacterial biodiversity and reduction in specific taxa, including Firmicutes, Bacteroidetes, Lactobacillus, and Eubacterium.78 An umbrella review of available meta-analyses identified nine factors that increase the risk of IBD: smoking (CD), urban living (CD and IBD), appendectomy (CD), tonsillectomy (CD), antibiotic exposure (IBD), oral contraceptive use (IBD), consumption of soft drinks (UC), vitamin D deficiency (IBD), and non-Helicobacter pylori-like enterohepatic Helicobacter species (IBD). Furthermore, seven factors reduce the risk of IBD: physical activity (CD), breastfeeding (IBD), bed-sharing (CD), tea consumption (UC), high levels of folate (IBD), high levels of vitamin D (CD), and Helicobacter pylori infection (CD, UC, and IBD).79 Nevertheless, the impact of modifying specific environmental factors on risk of disease, risk of disease progression, and risk of relapse remains inadequately studied, there being only limited high-quality data available from interventional studies.80, 81 4.3. Genetics Studies of genetic context in IBD have gradually used various approaches reflecting advances in genetic testing (family and twin studies, linkage studies, candidate gene association studies, genome-wide association studies).82 Genome-wide association studies (GWAS) alone have revealed the majority of information related to the genetics of IBD, showing that complex genetic disorders like IBD are polygenic, being driven by multiple, common genetic polymorphisms.63, 83 To date, GWAS and post-GWAS deep resequencing studies have identified 240 IBD-associated loci.41, 84 Nevertheless, as many as 80–90% of GWASidentified loci are confined to noncoding variation that exerts its pathogenic effects through modulation of gene expression. Recent studies have focused upon small intranuclear molecules that can broadly regulate gene expression, such as epigenetic markers, microRNAs, and non-coding RNAs, all of which have been implicated in the pathogenesis of IBD through different pathways and will be discussed later.63, 85 4.4. Authors’ contribution to the knowledge The family of noncoding RNAs (ncRNAs) exhibits a variety of biological functions.86 Noncoding RNAs can be divided according to their function into two groups: housekeeping ncRNAs (e.g., tRNAs, rRNAs, snRNAs, snoRNAs) and regulatory ncRNAs. Transcripts 20 shorter than 200 nucleotides are termed short noncoding RNAs and transcripts exceeding 200 nucleotides are called long noncoding RNAs. Both groups are involved in regulating gene expression and operate on several levels depending on their types. The short ncRNAs, such as microRNAs (miRNAs), small interfering RNAs (siRNAs), and PIWI-interacting RNAs (piRNAs), are involved mostly in post-transcriptional regulation but also in many other specific processes such as transposon silencing or ribosomal RNA maturation.85, 87 The ncRNAs have emerged as potential biomarkers for several diseases, as these are generally stable and abundantly present in a variety of clinical specimens, including tissues and bodily fluids; are highly tissue-specific, cell type-specific, and condition-specific; and can be readily detected by routine and inexpensive laboratory techniques.85, 88, 89 Several miRNAs and specific miRNA signatures have been identified in IBD-associated tissues, including serum,90 intestinal mucosa,91, 92 and stools.93, 94 Among many other cellular processes, it has been shown that miRNAs play a significant role in intestinal immunity. In our article (Annex 3), we provided an overview of current knowledge on ncRNAs, their altered expression profiles in pediatric IBD patients, and how these are emerging as potentially valuable clinical biomarkers.85 Figure 4 depicts tissue miRNAs involved in the development of PIBD.85 21 Figure 4. Tissue miRNAs involved in the development of pediatric IBD. Abbreviations: TLR, toll-like receptor; TNF, tumor necrosis factor; ANCA, anti-neutrophil cytoplasmic antibodies; IFN, interferon; TIMP, tissue inhibitor of metalloproteinases; MMP, matrix metalloproteinase; Bcl2, B-cell lymphoma 2; BAX, BCL2 associated X; CCL, CC chemokine ligand; CCR, CC chemokine receptor; ompC, outer membrane protein C precursor. The figure is modified from Park et al.95 and had been published in Jabandziev et al.85 22 5. Diagnostics Diagnosis of IBD is based on the Revised Porto criteria.16, 31 In the Czech Republic, national recommendations based upon these criteria are available.96, 97 Accurate diagnosis should be based on a combination of history, physical and laboratory examination, esophagogastroduodenoscopy, and ileo-coloscopy with obtaining mucosal samples for histological examination and imaging of the small bowel, if indicated.16 It is necessary to exclude other diseases mimicking the symptoms of IBD.16 Thus, we should rule out possible intestinal infection, allergic diseases, and many others; determine the basic subtype of IBD (UC, CD, UIBD), extent of the disease (Paris classification),35 activity of the disease (using Pediatric Crohn’s Disease Activity Index [PCDAI]98-100 and the Pediatric Ulcerative Colitis Activity Index [PUCAI]);101, 102 and the behavior of the disease.96 Table 3 presents the overall comparative characteristics of CD and UC.103 Table 3. Multiple characteristics of Crohn’s disease and ulcerative colitis. Clinical features Crohn’s disease Ulcerative colitis Symptoms and signs Abdominal pain, diarrhea, weight loss, anorexia, growth failure Bloody diarrhea, abdominal pain Location Mouth to anus; involves all layers of the gut: mucosa to serosa; most common: ileocolonic Colon; involves only mucosa; most common: pancolitis Endoscopic findings Segmental distribution, aphthous ulcers, deep fissuring ulcers, cobblestoning, perianal disease, strictures, fistulas Diffuse and continuous erythema, friability, granularity, loss of vascular pattern from rectum to variable extent Histological findings Pathognomonic noncaseating granulomas; patchy cryptitis, crypt abscesses, ileitis Cryptitis, crypt abscesses, crypt architectural distortion, basal lymphocytosis, distal Paneth cell metaplasia Radiologic findings Rigid stenotic segments, skip areas, and sinus tracts or fistulas Dilatation of colon in toxic megacolon Figure was prepared in accordance with Oliveira et al.103 23 5.1. Patient’s history, symptoms, and clinical examination Only few aspects are given greater emphasis in the literature than the importance of a detailed evaluation of the patient’s history and a thorough clinical examination, including a careful examination of the perianal area. This is in contrast with the fact that this often is not done in routine clinical practice.104 IBD is manifested by a wide range of symptoms, both intestinal and extraintestinal.105 Although typical symptoms from gastrointestinal tract affection are usually at the forefront, IBD could first present as extra-intestinal manifestations.106 These include erythema nodosum, pyoderma gangrenosum, episcleritis, uveitis, arthritis, and others.107 Clinical manifestations of CD in children may include fatigue, weight loss, abdominal pain, diarrhea, anemia, growth failure, and delayed puberty.2 UC is characterized by inflammatory affection of the colon. Typical symptoms include crampy abdominal pain, diarrhea, and rectal bleeding. The clinical symptomatology also depends on the extent of the disease; according to the Paris classification,35 the disease can be divided into isolated proctitis, left-sided colitis, extended colitis, and pancolitis. Pancolitis is very common in children and affects about three-quarters of patients.108 5.2. Laboratory tests Baseline tests include erythrocyte sedimentation rate, complete blood count, liver enzymes, albumin, and C-reactive protein (CRP).16, 96 Use of serum antibodies remains complementary in clinical practice, and serology may have a role in assessing prognosis (e.g., p-ANCA, ASCA).109 Nevertheless, normal blood tests cannot exclude PIBD. Almost one-tenth of all IBD patients diagnosed in a study by Ashton et al. had normal values for all widely used blood tests, and even more than 20% of patients had normal CRP.110 In comparing widely used laboratory tests in UC and CD, normal inflammatory markers were more common in UC. UC was more likely to have abnormal liver enzymes and all normal results. CD was more likely to have abnormal ESR, CRP, hemoglobin, platelets, and albumin.110 In recent years, fecal calprotectin (FCP) testing has gained relatively high clinical popularity and widespread use.111 In a recent study by Walker et al., FCP testing of children with suspected IBD accurately distinguished IBD from a functional gut disorder, reducing secondary care referrals and associated utilization of diagnostic health care.112 Nevertheless, clinicians must also be aware of the potential methodological limitations of FCP examination in routine practice.113, 114 Evidence regarding the clinical use and value of FCP measurements 24 in various gastrointestinal disorders in children were reviewed in a recently published position paper from ESPGHAN. Currently, FCP’s main use lies in the diagnosis and monitoring of IBD and its differentiation from functional gastrointestinal disorders.115 Consensus statements have recently been published regarding prognostic laboratory (and clinical) factors for such important clinical questions, among others, as need for surgery and disease complications with regard to CD116 and UC.117 5.3. Endoscopic examination Endoscopy has an essential and irreplaceable role in the diagnosis and management of PIBD. It is used to differentiate IBD subtypes, monitor disease activity, assess response to therapy, and treat possible complications.118, 119 Various endoscopic scoring systems are used in both research and clinical practice,120 including, among others, Simple Endoscopic Score for Crohn's Disease (SES-CD),121 Ulcerative Colitis Endoscopic Index of Severity (UCEIS),122 and Rutgeerts score123 for predicting the postoperative recurrence in CD patients after ileocolonic resection. Revised Porto criteria recommend performing upper gastrointestinal endoscopy and ileo-colonoscopy with multiple biopsies for all suspected patients with PIBD, as well as small bowel imaging (unless typical UC is determined after endoscopy and histology) by magnetic resonance enterography or wireless capsule endoscopy.16 Currently, recommendations are generally available for endoscopy in pediatrics, including indications for diagnostic and therapeutic esophagogastroduodenoscopy and ileo-colonoscopy, stricture/stenosis endoscopic management, endoscopic solving of upper and lower gastrointestinal bleeding; and others.124, 125 Recommendations for training and ongoing skills maintenance in pediatric endoscopy have recently been published.126 In pediatrics, performing an endoscopic examination means, in most cases, undergoing general anesthesia. It is essential to determine the optimal examination strategy in the diagnosis of patients with chronic conditions such as non-bloody diarrhea and abdominal pain. In a study from de Vijver et al., evaluating symptoms and blood and stool markers in patients with non-bloody diarrhea was found to be the optimal test strategy that allowed for reserving diagnostic endoscopy only for children at high risk for IBD. This approach allowed minimizing exposure to endoscopy in children.127 25 5.4. Histomorphologic examination Collaboration with a pathologist is essential, and the pathologist is part of the multidisciplinary team caring for pediatric IBD patients. Very important is to share clinical data with the pathologist. Differential diagnosis between CD and UC can be particularly challenging, such as when there are atypical presentation patterns for UC and Crohn’s colitis.128 Extremely important is obtaining mucosal biopsies from all parts of the gastrointestinal tract in IBD patients, and missing biopsies from grossly normal tissue would have missed abnormal histology.129 It should be emphasized that diagnostic gastrointestinal biopsies in VEO-IBD patients differ from those occurring with older onset of PIBD and include broader differential diagnosis (e.g., autoimmune enteropathy).130 Supplemental (special) stains based upon a patient’s clinical history are needed in special situations and must take into account the local differential diagnostic situation, such as in low-resource countries.131, 132 5.5. Imaging methods The noninvasive nature of imaging makes it an ideal tool for serial assessment of disease activity and treatment response. The choice of imaging (bowel ultrasonography, computed tomography, magnetic resonance imaging, etc.) should be based on the specific advantages and disadvantages of each (ionizing radiation, availability, reproducibility, length of examination, cost).103 For example, while ultrasound is a widely used, safe, fast, and low-cost method both for children suspected of IBD and for monitoring children with IBD,133 the diagnostic accuracy of ultrasound in detecting intestinal inflammation remains inconclusive.134 5.6. Examples of modern diagnostic methods potentially useful in clinical practice Although we have a fairly wide range of tools at our disposal for adequate diagnosis of IBD, several alternative diagnostic techniques have been developed recently. Various noninvasive markers are currently available, but they have limitations and do not provide ideal utility.135 In pediatric and adult patients, we can often encounter diagnostic delays.17, 18 Thus, it would be highly advantageous to have tools to predict whether patients will develop IBD at all. For example, Torres et al. identified a panel of 51 protein biomarkers (e.g., complement cascade proteins, lysosomes, glycosaminoglycans) that were predictive of Crohn’s disease within 5 26 years. On the other hand, predictive factors for the development of UC could not be found.136 It is clear that further research efforts are needed in this field. Recently, several studies have investigated serum dipeptidyl peptidase-4 as a biomarker of IBD severity and also as a predictor of subclinical disease activity, but with conflicting results.137-139 Serum profiling and non-coding RNAs are just starting to become widely used but reveal great promise for future clinical practice. Non-coding RNAs and their role in IBD are among the author’s main scientific focus areas and are discussed elsewhere.85, 140 In any case, combining different serum biomarkers can be valuable in improving the performance of disease evaluation.141 Studies have recently revealed as potentially promising new fecal biomarkers of intestinal inflammation calgranulin-C (S100A12), tumor pyruvate kinase isoenzyme type M2 (TuM2-PK), and fecal osteoprotegerin (FOPG).142 Fecal calgranulin-C and FCP had excellent test characteristics to predict IBD and justify endoscopy in a study by Heida et al.143 Moreover, calgranulin C appears to be a more suitable maker for predicting mucosal healing in children with IBD.142 Other, less well-established fecal biomarkers include high mobility group box 1 (HMGB1), chitinase 3-like 1, defensins, matrix metalloproteinases, and human nucleic acid. Most of these still require further extensive evaluations and validation.135 New endoscopic methods can be expected to emerge and be applied in clinical practice. In the case of UC, for example, magnification colonoscopy, endocytoscopy, and confocal laser endomicroscopy (CLE) enable assessing histological inflammation without the need for biopsy.144 Concretely, CLE allows visualization of mucosal abnormalities and thus in vivo histology during ongoing endoscopic evaluation by identifying macroscopically normalappearing mucosa, assessing intestinal epithelial barrier function and vascular permeability, and characterizing potential mucosal lesions, including dysplastic lesions. In particular, CLE used in conventional endoscopy could facilitate the assessment of mucosal healing in IBD.145 In a pilot study by our study group, we evaluated CLE’s usefulness in diagnosing esophageal diseases and came to promising results.146 Massively parallel sequencing, also termed next-generation sequencing (NGS), is increasingly used in clinical practice. Its high capacity creates new opportunities for NGS’s clinical application for establishing disease diagnoses and prognoses and in making therapeutic decisions.42, 147 27 Targeted sequencing is the NGS assay most commonly used. Typically, it interrogates tens or hundreds of genes presumed to be associated with a particular clinical phenotype or group of diseases. Targeted NGS panels are today very time- and cost-effective. A specific type of targeted NGS assay, known as whole-exome sequencing (WES), analyzes all of a genome’s protein-coding regions. It is particularly beneficial in cases of disorders that are phenotypically heterogeneous and when it is difficult to select an appropriate panel of candidate causative genes. Despite its clear advantages, WES also has some limitations, such as that intronic and noncoding regions remain uncovered and sequencing quality (often expressed as sequencing depth) is insufficient for particular genes, precisely because the range of targeted regions is so very large. Because NGS technology produces huge amounts of data, the processing and storage of that information represents a considerable bioinformatic challenge.148 When properly selected and used, NGS technologies provide excellent tools that greatly expand the range of genes analyzed and can contribute importantly to ensuring adequate preventive and therapeutic procedures for patients at risk.39, 42, 147 This is evidenced by use of the methodology in our patients, as documented in the articles discussed in this work.42, 149 5.7. Authors’ contribution to the knowledge Despite advances in IBD diagnostics and therapy, there is still a need for biomarkers to accurately identify IBD patients and predict treatment response.85 This knowledge could ultimately lead to more individualized approaches to patients. The identification of patients at high risk for a complicated disease course could then be crucial.150, 151 In the article by Jabandziev et al. (Annex 4),140 we stratified therapeutically naive pediatric patients diagnosed with UC pancolitis according to the severity of their condition and prediction for standard treatment according to the specific expression of 10 candidate miRNAs that we identified in a previous review paper.85 The work was created during the author’s time in the laboratory of Prof. Ajay Goel, at the Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Los Angeles, CA, USA. The study enrolled therapeutically naïve, pediatric UC patients only with confirmed pancolitis. We examined formalin-fixed paraffin-embedded specimens of colonic tissue for expression of 10 selected candidate miRNAs. We performed receiver operating characteristic curve analysis, using area under the curve and a logistic regression model, to 28 evaluate the diagnostic and predictive power of the miRNA panels. The final analysis included 60 patients. As a control group, 18 children without macroscopic and microscopic signs of inflammatory bowel disease were examined. A combination of three candidate miRNAs (let-7i-5p, miR-223-3p, and miR-4284) enabled accurate detection of pediatric UC patients and controls (Figure 5). A panel of four candidate miRNAs (miR-375-3p, miR-146a- 5p, miR-223-3p, and miR-200b-3p) was associated with UC severity in pediatric patients, and another combination of three miRNAs (miR-21-5p, miR-192-5p and miR-194-5p) was associated with early relapse of the disease. Nine patients out of the total were diagnosed with primary sclerosing cholangitis simultaneously with ulcerative colitis. A panel of 6 candidate miRNAs (miR-142-3p, miR-146a-5p, miR-223-3p, let-7i-5p, miR-192-5p, and miR-194-5p) identified those patients with primary sclerosing cholangitis. Specific combinations of miRNAs show promise as tools for potential use in precise disease identification and severity and prognostic stratification in pediatric patients with ulcerative pancolitis.140 29 Figure 5. Diagnostic accuracy of 3-miRNA panel for identifying UC patients. (A) The adaptive LASSO model. (B) A correlation matrix displaying Spearman’s rank correlation coefficient for each pair of three selected miRNAs. (C) Principal component analysis illustrating the good separation of UC-patient group and control group. (D) ROC curves for detecting UC patients using the 3-miRNA panel. (E) A waterfall plot representing risk score of each patient. Red and blue columns indicate UC patients and controls, respectively. A heat map illustrating expression levels of the three candidate miRNAs expressed differentially between UC patients and controls. The figure had been published in Jabandziev et al.140 30 6. Differential diagnostics In the diagnostics of children with suspected IBD, it is necessary to differentiate a relatively wide range of diseases that may be similar in their symptoms to the manifestations of IBD.30 Patients’ history and specific clinical signs should always be taken into account. In thinking about differential diagnostics, it is necessary to consider (and also to exclude if possible) infectious causes, immune-inflammatory disorders, vascular ischemic disorders, drug-induced colitis, and many others.152 Possible IBD-mimicking diseases are summarized in Table 4.153, 154 Particularly very challenging in differential diagnosis can be to manage patients with VEO-IBD. Therefore, this topic is discussed in a following separate chapter. Table 4. Differential diagnostics of IBD-mimicking diseases. Infection bacterial Campylobacter jejuni, Salmonella sp., Shigella dysenteriae, Yersinia enterocolitica, enteroinvasive E. coli, Clostridium difficile etc. viral Norovirus, Rotavirus, Coronavirus, Adenovirus, Echovirus, Cytomegalovirus, Herpes simplex virus type 2 etc. parasitic Giardia lamblia, Entamoeba histolytica, etc. fungal Candida crusei, Candida glabrata etc. (especially in immunocompromised patients) toxins Vibrio cholerae (choleragen), Staphylococcus aureus (enterotoxin A-E), Clostridium difficile (toxin A, B) etc. Drugs (drug-induced colitis) microscopic colitis non-steroidal anti-inflammatory drugs, proton pumps inhibitors, statins, β-blockers, etc. macroscopic colitis chemotherapeutic drugs (e.g., taxane, platinum), immunomodulators (e.g., infliximab, adalimumab), laxatives, antibiotics etc. inflammatory colitis non-steroidal anti-inflammatory drugs, immune checkpoint inhibitors (nivolumab) etc. Vascular- ischemic disorders Henoch-Schönlein purpura, systemic vasculitis (dermatomyositis, systemic lupus erythematodes), granulomatosis with angiitis Immune- inflammatory (including monogenetic diseases) eosinophilic gastroenteritis, severe combined immunodeficiency syndrome, common variable immunodeficiency diseases, interleukin-10 signaling defects, chronic granulomatous disease, X-linked lymphoproliferative syndrome type 1,2, Wiskott-Aldrich syndrome, Omenn syndrome, congenital neutropenia, Behcet’s disease, autoimmune enteropathy, agammaglobulinemia, mevalonate kinase deficiency, etc. Other coeliac disease, lactose intolerance, irritable colon syndrome, glycogen storage disease type b, radiation colitis, intestinal lymphoma, Hermansky-Pudlak syndrome, trichohepatoenteric syndrome, sarcoidosis, laxative abuse, etc. Table was prepared in accordance with Kliegman et al.153 and Hamdeh et al.154 31 6.1. Very-early-onset IBD In VEO-IBD patients who do not respond to standard therapy, we must consider the possible presence of primary immunodeficiency. This possibility should be considered especially in children under 2 years of age. A phenotypic aide-memoire summarizing key findings to ensure that a careful clinical history for VEO-IBD and examination are made to narrow the search for an underlying monogenetic defect is: YOUNG AGE MATTERS MOST (YOUNG AGE onset, multiple family members and consanguinity, autoimmunity, thriving failure, treatment with conventional medication fails, endocrine concerns, recurrent infections or unexplained fever, severe perianal disease, macrophage activation syndrome and hemophagocytic lymphohistiocytosis, obstruction and atresia of intestine, skin lesions and dental and hair abnormalities, and tumors).38 Warning signs that could ideally lead to suspicion of monogenic VEO-IBD or IBD-like disease are summarized in Table 5.38 Table 5. Warning signs for suspecting monogenic IBD. Key points Comments Very early age onset of IBD-like immunopathology Likelihood increases with very early onset, particularly in those younger than 2 years of age at diagnosis Family history In particular, consanguinity, predominance of affected males in families, or multiple family members affected Atypical endoscopic or histological findings For example, extreme epithelial apoptosis or loss of germinal centers Resistance to conventional therapies Such as exclusive enteral nutrition, corticosteroids, and/or biological therapy Skin lesions, nail dystrophy, or hair abnormalities For example, epidermolysis bullosa, eczema, folliculitis, pyoderma or abscesses, woolly hair, or trichorrhexis nodosa Severe or very early onset perianal disease Fistulas and abscesses Lymphoid organ abnormalities For example, lymph node abscesses, splenomegaly Recurrent or atypical infections Intestinal and non-intestinal Hemophagocytic lymphohistiocytosis Induced by viral infections such as Epstein-Barr virus or cytomegalovirus, or macrophage activation syndrome Associated autoimmunity For example, arthritis, serositis, sclerosing cholangitis, anemia, and endocrine dysfunction, such as thyroiditis and type 1 diabetes mellitus Early development of tumors For example, non-Hodgkin lymphoma, skin tumors, hamartoma, thyroid tumors Table was prepared in accordance with Uhlig et al.38 32 Physical findings and comorbidities of which physicians should be aware in suspicion of monogenic VEO-IBD or VEO-IBD-like syndromes at the initial physical examination and during follow-up are depicted in Figure 6.155 Figure 6. Key indicators of monogenic IBD in clinical practice. In parentheses after the listed comorbidities and specific clinical findings are genes whose mutations are responsible for the disease manifestations. Figure was prepared in accordance with Nambu et al.155 and created in collaboration with the Service Center for E-Learning at Masaryk University, Faculty of Informatics. Certainly, a subset of the more aggressive, therapy-resistant VEO-IBDs, which have recently been regarded as associated with inborn errors of immunity (IEI), should be mentioned.28, 156 The updated IEI list from the International Union of Immunological Societies phenotypic classification includes 406 IEI disorders with 430 gene defects.157 To date, more than 50 monogenic defects associated with IBD or IBD-like phenotype have already been identified.158 Gastrointestinal manifestation of IEI is common and may precede the primary diagnosis of IEI.156, 158 From a pathophysiological point of view, VEO-IBD related to IEI manifests as a result of impaired barrier function of the intestinal epithelium, defects in bacterial killing by phagocytes, increased hyper- or autoimmune inflammatory pathways, or 33 impaired development and function of the adaptive immune system.42, 97, 156, 158 The subgroups of IEI-associated VEO-IBD are summarized in Table 6.28 Table 6. Inborn error of immunity-associated VEO-IBD groups • Genetic variants influencing the integrity of intestinal barrier • Genetic variants influencing bacterial recognition and clearance • Genetic variants in the IL-10-IL-10R pathway and related cytokine family members • Genetic variants impairing regulatory T cells • Genetic variants impairing development of the adaptive immune system • Genetic variants resulting in autoinflammatory disorders Table was prepared in accordance with Kelsen et al.28 Because the management of monogenic IBD is different from that for classical IBD, it is crucial to identify these patients. The Paediatric IBD Porto Group of ESPGHAN recently published a position paper covering indications, technologies (targeted panel, exome and genome sequencing), gene panel setup, cost-effectiveness of genetic screening, and requirements for the clinical care setting.39 It is clear from the above that the differential diagnosis of IBD and especially of the VEO-IBD group is highly complex and challenging. Therefore, it should ideally be performed in IBD centers having sufficient experience and capability to perform adequate diagnosis.159 6.2. Authors’ contribution to the knowledge Phosphomannomutase-2 deficiency (PMM2-CDG) Congenital disorders of glycosylation (CDG) comprise a large and heterogeneous group of more than 130 monogenic diseases.160 CDG is caused by defects in the synthesis of glycans and in the attachment of glycans to proteins and lipids. Clinical manifestation is multisystemic.161, 162 The N-linked protein glycosylation defect PMM2-CDG (phosphomannomutase-2 deficiency), previously known as CDG type Ia, was the first reported CDG and remains the most common CDG to date.160 PMM2-CDG patients can be divided into those with neurological and those with neurovisceral phenotype.163 Neurovisceral phenotype includes heart (pericardial effusion and cardiomyopathy), liver (high serum transaminases), and gastrointestinal (chronic diarrhea, feeding tube, and gastroesophageal reflux) involvement.164, 165 In a retrospectively analyzed group of 96 French patients with PMM2-CDG, the presenting signs were mostly neurological (hypotonia, intellectual 34 disability, cerebellar syndrome) and observed in almost all the patients. In addition to neurological signs, a total of 38 patients exhibited visceral features including at least one of the following: feeding difficulty requiring nutritional support (n=23), cardiac features (n=20; pericarditis), hepato-gastrointestinal features (n=12; chronic diarrhea: 7, protein-losing enteropathy: 1, ascites: 3, liver failure: 1, portal hypertension: 1), kidney features (n=4), and hydrops fetalis (n=1).164 We described (Annex 5) an infant boy with PMM2-CDG and novel splicing mutation, who presented with pericardial effusion, typical dysmorphic facial features, inverted nipples, failure to thrive, and psychomotor retardation. Screening for CDG performed using isoelectric focusing of serum transferrin showed a typical PMM2-CDG pattern. Exome sequencing revealed one common pathogenic variant (c.691G > A/p.Val231Met) and one novel variant (c.447 C 3dupA) in the PMM2 gene. Both PMM2 variants were further confirmed by Sanger sequencing in both the proband and the parents’ DNA. The novel variant was predicted to result in loss of donor splice site. Analysis at mRNA level confirmed that it leads to exon five skipping (r.348_447del) and causes premature termination of translation to the protein (p.G117Kfs-4).149 Although CGD-MM2 is a relatively rare disorder, it can have gastrointestinal symptomatology and thus should be considered, especially in the differential diagnosis of patients with suspected VEO-IBD or VEO-IBD-like syndromes. MIRAGE syndrome A 2016 paper reported on a group of 11 patients with a newly identified syndrome that was termed MIRAGE.166 MIRAGE is an acronym for the significant findings of myelodysplasia, infection, restriction of growth, adrenal hypoplasia, genital phenotypes, and enteropathy. Gastrointestinal complications include chronic diarrhea and esophageal dysfunction.166 MIRAGE syndrome is an extremely rare disease. Several dozen patients have been described so far.167-170 We described the case report of a patient with MIRAGE syndrome (Annex 6).171 The author cared for this patient for a long time at our department in the intensive care unit and ambulatory care. The reported patient had a novel mutation in SAMD9 (c.2471 G>A, p.R824Q), manifesting with prominent gastrointestinal tract involvement and immunodeficiency. Among other major symptoms, he had difficulty swallowing, requiring percutaneous endoscopic gastrostomy, frequent gastrointestinal infections, and perianal 35 erosions. He suffered from repeated infections and periodic recurring fevers with elevation of inflammatory markers. At 26 months of age, he underwent hematopoietic stem cell transplantation that significantly improved hematological and immunological laboratory parameters. Nevertheless, he died at day 440 post-transplant due to sepsis. Even though it is a sporadic disease, SAMD9 mutations should be considered as a cause of enteropathy in pediatric patients, especially in combination with other described symptoms.171 Eosinophilic esophagitis The finding of eosinophilic infiltration accompanies inflammatory diseases of the gastrointestinal tract of various origins, and the differentiation of primary and secondary forms is crucial for further diagnosis and treatment.172 Primary eosinophilic gastrointestinal disorders (EGID) are a relatively new group of diseases. They can affect any part of the gastrointestinal tract and are characterized by mucosal infiltration of eosinophils in the absence of other causes for eosinophilia.173, 174 We can diagnose eosinophilic esophagitis, eosinophilic gastritis, eosinophilic gastroenteritis, eosinophilic enteritis, and eosinophilic colitis, or a combined involvement of different parts of the gastrointestinal tract.173 In populations of Western countries, eosinophilic esophagitis is by far the most common form.172 These diseases present with a broad spectrum of often non-specific symptoms and with differential diagnosis ranging from functional gastrointestinal diseases to severe organic disorders, including IBD. Generally, clinical manifestation depends upon the location of eosinophilic infiltration. The pathophysiology of EGID is still unclear. It seems that food and aeroallergens play an essential role. The endoscopic appearance of EGID is not specific. A combination of serious clinical suspicion and histopathologically proven dense eosinophilic infiltration of the intestine are essential for diagnosis.174 Unfortunately, with the exception of the esophagus, eosinophilic infiltration of the gastrointestinal wall is common.175 Furthermore, we can find eosinophils in mucosal specimens in the cases of many inflammatory diseases, such as celiac disease, IBD, and parasitic infections. Moreover, eosinophilic infiltration of the gastrointestinal tract may accompany hypereosinophilic syndrome, vasculitis, as well as post-medicamentous and malignant diseases.176 In our observational survey (Annex 7),177 we aimed to characterize features of eosinophilic esophagitis (EoE) diagnosed in five pediatric endoscopy centers and to describe local strategies for its treatment. This analysis focused on describing their general situation (age, gender, symptoms) and also aimed to investigate any possible linkage between age and 36 symptoms or length of diagnosis period. Demographic features; clinical symptoms; laboratory, endoscopic, and histopathological findings; and chosen treatment of patients were recorded and analyzed. We precisely described 33 new cases of children with EoE and their clinical characteristics. To our best knowledge, this was the first retrospective study on pediatric patients with EoE in the Czech Republic. We have proposed collecting long-term prospective observational data into a national EoE register of patients in the Czech Republic, as doing so would significantly improve our knowledge of this disease.177 Herpetic esophagitis Endoscopic examination of the upper gastrointestinal tract comprises an integral part of the diagnostics process in patients with suspected IBD.16, 125 Inflammation in upper gastrointestinal tract is described in approximately half of the children with IBD during the initial assessment. In a study by Castellaneta et al., the sites most frequently involved were the stomach (67%) followed by the esophagus (54%) and duodenum (22%).178 From the pathological point of view, among the manifestations of pediatric IBD in upper gastrointestinal tract include lymphocytic esophagitis, focally enhanced gastritis, duodenal inflammation, and epithelioid granulomas.179 In addition to IBD, differential diagnosis of ulcerative esophagitis must consider esophageal infections (herpes simplex, cytomegalovirus, varicella-zoster virus, candida, or various bacterial agents), trauma, esophageal burns, and Behcet’s disease.180, 181 We described (Annex 8)181 the case report of a 7-year-old immunocompetent boy with a suddenly occurring triad of symptoms: odynophagia, chest pain, and fever. In addition to other standard examinations, we performed esophagogastroduodenoscopy, where significant inflammatory changes transitioning to longitudinal ulcerations in the distal third of the esophagus were revealed.181 In addition to the standard sampling of esophageal tissue, biopsy material was also sent for bacteriological and mycological examination as well as for the detection of herpes simplex virus (HSV) and cytomegalovirus by polymerase chain reaction (PCR). The result of HSV DNA detection by PCR from the esophageal mucosa was positive. Herpesvirus infections, in general, can complicate the course of IBD. Immunosuppressive therapy appears to increase the risk of herpesvirus reactivation and predispose to more severe infections. While cytomegalovirus colitis and systemic Epstein-Barr virus reactivation are well known, HSV is rarely involved. When HSV infections do occur, they are usually confined to sites of local reactivation, such as mucosal surfaces (e.g., the oropharynx, 37 anogenital region, and eyes) and sites on the skin. However, case reports describing disseminated HSV infection with colonic involvement have been published.181, 182 The presented work demonstrates the necessity to consider the possible presence of HSV infection in differential diagnosis of endoscopic findings of gastrointestinal ulcerations.181 38 7. Therapy The goals of treatment include to relieve disease symptoms, achieve intestinal healing, reach growth potential, and optimize the quality of life while limiting drug toxicity.183 IBD therapy has come a long way in recent decades, and it will probably continue to undergo intensive development. There are national recommendations for the treatment of pediatric IBD patients,96, 97 as well as recent international recommendations for the treatment of both UC and CD.19-22 In an ideal situation, therapy of pediatric patients with IBD should be individualized, using a risk-stratification approach and predictors of poor outcome.184 Treatment modalities could be divided into several groups, specifically medical therapy, nutritional therapy, and endoscopic and surgical therapy. Experimental therapeutic modalities, such as fecal microbial transplantation, plasma exchange, using of mesenchymal stromal cells, and others, have also been studied.185-187 There also are some alternative approaches, which some patients require and about which the treating physician should be informed.188 7.1. Medical therapy Medical therapy has long been limited to so-called non-biological therapies (aminosalicylates, thiopurines, and steroids). Together with a better understanding of the etiopathogenesis of IBD, the development of biological therapy has been a significant advance in treatment.189 Currently, tumor necrosis factor-alpha inhibitors infliximab and adalimumab are approved for use in pediatric patients in the Czech Republic. The use of other agents, such as ustekinumab and vedolizumab, is only possible in the so-called “off label” regimen after approval by the relevant health insurance company.97 It can be expected, however, that other and novel therapies could enter the diagnostic repertoire of pediatric gastroenterologists. Generally, this occurs after experience has been gained with new drugs in the treatment of adult patients.190 The new biologics and small molecule drugs block immune cell communication or migration. Novel small molecules include Janus kinase (JAK) inhibitors (tofacitinib), small mothers against decapentaplegic homolog (SMAD)7 antisense oligonucleotides (mongersen), sphingosine-1-phosphate (S1P) receptor modulators (ozanimod, etrasimod), and phosphodiesterase (PDE)4 inhibitors (apremilast). Other novel biologics include anti-integrins (vedolizumab, natalizumab), anti-cytokines (ustekinumab, risankizumab, brazikumab, mirikizumab, guselkumab), and anti-mucosal vascular addressin cell adhesion molecule-1 (MAdCAM-1) (etrolizumab).183, 191 Some of these drugs are already part of clinical practice, while others are being tested in various phases of clinical trials. 39 Cases of children being treated with combinations of biological agents with relatively promising results have already been described in the literature. Dual biological therapy could thus potentially expand the range of treatment options.192 Figure 7 summarizes the different therapeutic agents and their site of action within the pathophysiology of IBD.191 Figure 7. Therapeutic targets of novel biologics and small molecules for the treatment of inflammatory bowel disease. Biologics and small molecules are indicated by red and green colors, respectively. AMP, adenosine monophosphate; cAMP, cyclic adenosine monophosphate; IEL, intraepithelial lymphocyte; Iκb, inhibitor of κB; IL, interleukin; JAK, Janus kinase; MAdCAM-1, mucosal vascular addressin cell adhesion molecule-1; NF-κB, nuclear factor-κB; P, phosphorylation; PDE, phosphodiesterase; S1P, sphingosine-1-phosphate; SMAD, small mothers against decapentaplegic homolog; STAT, signal transducer and activator of transcription; TGF-β, transforming growth factor-β; TGF-βR, TGF-β receptor; TNF-α, tumor necrosis factor-α; TNFR, TNF receptor. The figure was prepared in accordance with Na et al.191 and created in collaboration with the Service Center for E-Learning at Masaryk University, Faculty of Informatics. 40 7.2. Nutritional therapy In pediatrics, nutritional therapy can be used for two reasons: to improve the nutritional status of both UC and CD patients or as a means to induce remission in patients with a defined type of CD (exclusive enteral nutrition).193 Recently, evidence has been published on the possibility of combining enteral nutrition and a special diet in the treatment of CD (Crohn’s Disease Exclusion Diet, CDED), suggesting very promising results.194, 195 At our department, we have relatively good experience with this treatment modality. In the context of IBD treatment, the effect of other diets has also been investigated, but with inconclusive results.196- 199 7.3. Endoscopic and surgical therapy Endoscopic examination and follow-up is a common modality in managing patients with IBD.125 Endoscopic instrumentation allows us, for example, to dilate possible stenoses.200 Surgical treatment has become a common part of comprehensive IBD treatment. In UC, this involves mainly subtotal or total colectomy with the construction of ileo-anal pouch anastomosis.201 In CD, it is often an ileocecal resection performed because of irreducible inflammatory activity in this area, or resolution of symptomatic stenosis of the ileocecal valve.202 The role of the surgeon in managing complications of perianal involvement (i.e., fistulas, abscesses) is indispensable.203, 204 7.4. Authors’ contribution to the knowledge Ileocecal resection is an integral part of treating selected pediatric and adult patients with CD,201, 205 despite the fact that, concurrently with the development of biological therapies, the need for surgery is decreasing.206 The issue of predicting postoperative disease recurrence, however, was not entirely clear. The aim of the study by Poredska et al. (Annex 9)207 was to determine whether the histological activity of CD in resection margins after ileocecal resection is associated with early endoscopic recurrence of the disease. This study was conducted with adult patients, but some of these patients had been previously treated at the at the authors’ workplace (i.e., the Department of Pediatrics). The resection line was always up in macroscopically healthy tissue. Ileocecal resected specimens were histologically examined for the presence of microscopic signs of CD, including both resection margins, that is, the small and large intestine. At 6 months postoperatively, patients underwent a follow-up 41 colonoscopy during which endoscopic recurrence was assessed according to the Rudgeerts score.208 We investigated whether histological findings in the resection margins correlated with endoscopic recurrence of CD. Furthermore, the effects of preoperative therapy and other risk factors associated with endoscopic recurrence were evaluated. Endoscopic recurrence of CD at 6 months after surgery was observed in 23 patients out of a total of 107 enrolled. Microscopic evidence of CD in the resection margins was associated with significantly higher endoscopic recurrence in the anastomosis (56.5% versus 4.8%, p<0.001). Duration of disease from diagnosis to surgery (p=0.006) and length of resected bowel (p=0.019) were significantly longer in patients with proven endoscopic recurrence. Thus, it was shown that microscopic evidence of CD in the resection line in cases of ileocecal resection was significantly associated with a higher risk of early postoperative endoscopic recurrence. The results of a recent meta-analysis, where our work was included, showed that positive resection margins and myenteric plexitis and granulomas in the resection margins significantly increased the risk for postoperative recurrence of CD.209 This is consistent with the results of our study.207 42 8. Complications This chapter will discuss complications, both within and outside the digestive tract, that are so-called extraintestinal complications. It should be said that complications include here, from a somewhat broader point of view, manifestations that may be present as initial manifestations of the disease in individual patients (e.g., perianal fistulation). In the following text, the issue of potential adverse effects of medicament therapy will not be discussed further. 8.1. Complications from within the gastrointestinal tract Strictures can develop practically anywhere in the gastrointestinal tract, and especially in patients with CD. These lesions may be asymptomatic or may require some type of intervention (dilatation or surgical resection).210 Gastrointestinal fistulas are abnormal connections between the bowel and neighboring organs. The occurrence of CD in childhood is associated with more aggressive development of perianal fistulas. Fistulas can occur in almost one-third of patients within 5–7 years after diagnosis.211 Fistula types include perianal, entero-cutaneous, entero-enteric, enteromesenteric, recto-vaginal, and entero-vesical.212 Perianal fistulas are most common and commonly cause severe infections, fecal incontinence, perianal discharge, negative selfimage, and social isolation. Perianal fistulas significantly reduce the quality of life of patients and are relatively difficult to treat,213 but the introduction of anti-TNF has improved patients outcomes.183, 214 Toxic megacolon (TM) is one of the most feared complications of IBD, especially UC, and should be considered in these patients.215 It is characterized by a combination of systemic toxicity (fever, tachycardia, leukocytosis, altered mental status) and segmental or total dilatation of the colon.216 TM is fraught with high morbidity and mortality and can require surgical treatment. Various infections, especially Clostridium difficile, may also be a causative factor in the development of TM.21, 212 Other complications include primary sclerosing cholangitis, orofacial granulomatosis, and cholecystolithiasis.217-220 A feared long-run complication is malignancy, inasmuch as the onset of IBD in childhood is associated with increased risk of any cancer, and especially of gastrointestinal cancers, both in childhood and later in life.221 43 8.2. Extraintestinal and systemic complications Approximately one-third of children with CD and as many as 10% of children with UC suffer from growth impairment. The cause of growth failure is multifactorial and includes, for example, decreased appetite and decreased peroral intake, increased metabolic demand, malabsorption due to mucosal inflammation, growth hormone resistance due to inflammation, and use of corticosteroids. Therefore, thorough monitoring of linear growth is extremely important, and overall treatment effort should be directed to restoring normal growth.12, 222 Along with the above factors, even lower physical activity can lead to lower bone mineral density,223 as a higher incidence of low bone mineral density in pediatric IBD patients has been observed in children with CD. A higher rate of bone fracture in children with IBD was reported to follow corticosteroid treatment.224 Patients with IBD also are at risk for deficiencies of various micronutrients, such as iron, folate, vitamin B12, and vitamin D.12 Psychosocial influences are not negligible, as children with IBD can have higher rates of depressive and anxiety disorders.225 Various body systems can be affected, including musculoskeletal (arthritis, ankylosing spondylitis),226 integumentary system (erythema nodosum, pyoderma gangrenosum),227 ophthalmological (iritis, episcleritis),228 urogenital (calcium oxalate stones),229 hematological (anemia, thromboembolism)230, 231 and immunological (infections).212, 232, 233 8.3. Authors’ contribution to the knowledge In general, IBD and its treatment are associated with significant morbidity and highly probable hospitalization.234, 235 Hospitalization alone increases the risk of infectious complications.236 IBD patients, and especially those with more severe disease courses, often require corticosteroids, biologics, and/or immunosuppressive agents. Underlying active disease and treatment with immune-suppressing therapy contributes to an increased risk of serious and opportunistic infections in these patients.235, 237 Moreover, beyond immunological dysregulations commonly underlying IBD, risk factors for infections in children with IBD are represented by rare monogenic disorders, including primary immunodeficiencies, poor nutritional status, and, above all, in comparison to adults, more frequent need for early and/or combined immunosuppressive and biologic therapies.232, 233 Sepsis is a syndrome shaped by pathogen factors and host factors (e.g., sex, race and other genetic determinants, age, comorbidities, environment) with characteristics that evolve over time.238, 239 What differentiates sepsis from infection is an aberrant or dysregulated host response and the presence of organ dysfunction. The clinical and biological phenotype of 44 sepsis can be modified by preexisting acute illness, long-standing comorbidities, medication, and interventions.240 Despite significant advances in research and treatment strategies, sepsis remains among the most threatening conditions in intensive care units for children and adults.241, 242 Considerable variabilities in the course of sepsis can be caused by, among other things, variants of the genes encoding the individual components of the immune system.243 Only limited data was available describing the influence of single nucleotide polymorphisms and their combinations on the risk for development and severity of the course of sepsis in the pediatric population.244 In our study (Annex 10), we revealed statistically significant differences in the structure of single nucleotide polymorphism combinations between the group of patients with septic conditions and the control group, as well as between the group of patients with the most severe conditions (severe sepsis, septic shock, and multiorgan dysfunction syndrome) and the control group. This approach has made it possible to describe low-, medium- and high-risk combinations of genetic polymorphisms in the development and subsequent severity of septic conditions. The study described a total of 23 pediatric patients who died due to a septic condition. One of these patients was treated for Crohn’s disease and died due to septic shock.244 Septic complications are relatively rare in pediatric patients with IBD, but the possibility for their occurrence should be kept in mind.245 45 9. Potential perspectives on further research 9.1. General perspectives on research in the field of IBD Currently, research in the field of IBD is very intensive and the interest in this issue is constantly increasing. Barash et al. mapped IBD research from the MEDLINE/PubMed database for the 25-year period 1992 and 2016. A total of 18,653 relevant publications were classified as IBD-related and further analyzed. The annual number of publications increased almost 4-fold (from 354 to 1361) during the studied period.246 Despite recent advances in research, however, the complexity of IBD still creates enormous challenges for researchers.247 Current research is not fully able to satisfactorily address important research questions.248 Further and deeper understanding of the pathophysiological processes in IBD would help to discover relevant biomarkers that could be used in both diagnosis and treatment.81 A promising concept could relate to functional integration of those -omes relevant to the pathogenesis of IBD, such as the exposome, genome, epigenome, microbiome, and others, forming the so-called IBD interactome.249, 250 This could be defined as a disease network within which dysregulation of individual -omes causes intestinal inflammation mediated by dysfunctional molecular networks controlling all biological events (Figure 8).250 Figure 8. Functional integration of -omes relevant to the pathogenesis of the IBD-forming IBD interactome. The hypothetical disease network on the right shows the various molecular hubs (white nodes) of the network, which is regulated by central regulatory hubs (gray nodes). The figure was prepared in accordance with Souza et al.250 and created in collaboration with the Service Center for E-Learning at Masaryk University, Faculty of Informatics. 46 In step with a better understanding of the pathogenesis of IBD, the portfolio of therapeutic options continues to expand. Nevertheless, new therapeutics can be costly, and patients may stop responding to therapies or not respond to them in the first place. Therefore, there is a growing need for more personalized therapy based on insights into the biology of the underlying disease and a drive to change our approach from reactive treatment driven by disease complications to proactive care to prevent disease sequelae.72 This thinking embraces a clear desire to make medicine as individualized – or, more precisely, as personalized – as possible. Personalized medicine involves identifying patients at high risk of progression and complications and better characterizing patients who may respond preferentially to specific treatments.251 In an even narrower sense, we have recently encountered the term precision medicine. The concepts of personalized medicine and precision medicine are very similar, but precision medicine also includes a multidisciplinary, data-driven approach to support better clinical decision-making through a clear understanding of the molecular basis of an individual’s disease.72 Precision medicine in this sense means tailoring treatment to the individual patient and incorporating different data-driven (and multi-omics) approaches to support accurate clinical decision-making. In the case of IBD, precision medicine would be of significant benefit, as it would allow for early treatment that is both effective and appropriate for the individual.72 In summary, there is a need to understand and predict the natural course of IBD-disease susceptibility, activity, and behavior; to predict disease progression and response to therapy; and to optimize current and develop new molecular technologies.252 Precision medicine could provide the ways to do just that. To achieve this, prospective longitudinal cohort studies are needed to identify and validate precision medicine biomarkers for predicting disease progression and for predicting and monitoring treatment response. Methodological harmonization across studies together with the development of standardized methods and infrastructure are key to achieving this goal.252 It should be borne in mind that the overall situation can change quite a lot. After all, who among caregivers and patients could have guessed the substantial changes to the routine management of IBD that we experienced during the unexpected coronavirus disease 2019 (COVID-19) pandemic?253 9.2. Author’s perspectives on research in the field of IBD Microbial dysbiosis and microRNA Together with colleagues from the Department of Pathology, University Hospital Brno and Central European Institute of Technology, we obtained grant funds from the Czech Health 47 Research Council for carrying out our research project entitled: Study on microbial dysbiosis and microRNA deregulation as a basis of the therapy individualization in pediatric patients with inflammatory bowel disease. Work on the project is already fully underway. As part of this research, we will work with our biobank, where we already have material from more than 100 pediatric patients with newly diagnosed IBD. The specific aims of the project are to: - identify miRNAs specific for IBD patients by global profiling of miRNA expression in the tissues and stools of IBD patients compared to healthy control subjects and miRNAs specific for different IBD subtypes (classes) by comparing miRNA expression profiles in therapeutically naïve patients; - establish an miRNA predictive panel to identify patients with good and bad response to a standard therapeutic regimen; - validate and analytically characterize candidate diagnostic, prognostic, and predictive miRNAs on an independent cohort of IBD patients; - analyze microbiome composition in stool and tissue biopsy samples and identify specific changes in microbiome composition in IBD patients compared to healthy controls and in patients with different IBD subtypes; - compare levels of candidate diagnostic, prognostic, and predictive miRNAs in tissue, stool and plasma and correlate these with specific changes in the composition of the intestinal microbiome; and - propose a prospective clinical trial verifying the clinical benefit of identified miRNAs in improving the diagnostic and therapeutic approach to pediatric patients with IBD. A schematic representation of the study design is depicted in Figure 9. 48 Figure 9. Schematic representation of the study design. CD – Crohn’s disease; UC – ulcerative colitis; HC – healthy control. Brain alterations in pediatric patients with IBD Central nervous system (CNS) involvement has been reported in pediatric and adult IBD patients, with both neurological and psychiatric phenomena.254-259 To date, there are not many studies investigating directly affection of the CNS in CD, especially in children. Research on possible cerebral involvement in IBD generally and CD specifically has been largely marginalized and failed to capitalize on recent developments in magnetic resonance imaging (MRI). In a cross-sectional pilot study, we searched for eventual macrostructural, microstructural, and functional brain affection in children with CD early after the disease’s onset. Nine pediatric CD patients within 2 years of disease development and at the same time with their disease in remission (according to FCP and PCDAI) and nine healthy controls underwent structural, diffusion-weighted imaging and resting-state functional MRI acquisition in combination with extensive neuropsychological testing. While no differences in cortical thickness between CD patients and healthy controls were found, alterations were detected in diffusion tensor imaging parameters over vast cortical regions essential for regulation of the autonomous nervous system, sensorimotor processing, cognition, and behavior. These alternations were accompanied by generally increased functional and structural connectivity 49 (Figure 10). Although still requiring further validation in longitudinal projects enrolling more significant numbers of subjects, this study sets out possible directions for further research. The discussed data are from an article that is now under review at the journal Frontiers in Pediatrics. In the future, we would like to seek grant funding to conduct a prospective study on microstructural changes in the brain of pediatric patients after the diagnosis of IBD. Figure 10. Results of parcellated analysis comparing Crohn’s disease patients and healthy controls. (A) fractional anisotropy, (B) mean diffusivity, (C) degree centrality, (D) amplitude of lowfrequency fluctuations (no significant results), (E) structural connectivity, and (F) partial functional connectivity regularized using ridge regression. Absence of significant results (subcortical outcomes for mean diffusivity amplitude of low frequency fluctuations and structural connectivity) is marked with “×”. Paneth cells and interleukin 17 Toward better understanding IBD pathophysiology, we investigated, in cooperation with colleagues from the Institute of Molecular Genetics of the Czech Academy of Sciences (IMG CAS), the role of interleukin 17 (IL-17) on Paneth cells (PCs). Paneth cells are a highly specialized cell type with many functions in intestinal physiology.260 Recently, there is increasing evidence for a potential role of PCs in the development of ileal CD.261 As 50 mentioned above, microbiota composition regulation is essential for intestinal homeostasis when it is known that IL-17 regulates antimicrobial peptide production on epithelial surfaces.262 The effect of IL-17 on PCs is nevertheless still unclear. We showed that PCs express high levels of surface IL-17 receptor (IL-17R), and targeted ablation of IL-17R in PCs decreases the cellularity of ileal PCs and the expression of their enteric α-defensins. Mice with PCs lacking IL-17R showed upregulated inflammatory pathways and higher severity of induced ileitis. These changes were associated with lower gut microbiota diversity, capable of inflicting death upon its transfer to genetically susceptible mice. Strikingly, a sub-cohort of pediatric patients with newly diagnosed Crohn’s disease displayed a low number of ileal PCs, high ileitis severity score, and diminished serum levels of IL-17, thereby resembling the phenotype of mice with IL-17R-deficient PCs. Our study identified IL-17 signaling in PCs as an essential contributor to ileal homeostasis acting via the prevention of dysbiosis (Figure 11). The discussed data are from an article that is now under review at the journal Cell Reports. We will continue our cooperation with IMG CAS on research in this field. 51 Figure 11. Patients with CD manifest low ileum PC numbers, which correlate with low serum IL-17 and terminal ileitis. (A–C) Immunofluorescence microscopy analysis of frozen ileum biopsies from CD and nonIBD patients. Sections were stained with lysozyme (PCs, green) and DAPI (nuclei, blue). (A) Graph shows average numbers of PCs in patient/biopsy per crypt. (B) The PC number shows bimodal distribution in CD patients. The number of PCs per crypt is plotted arbitrarily for non-IBD and CD individuals, together with the best normal (dashed curve) and bimodal normal (full line) fits to the data, respectively. All samples in the CD-PCLow group of CD patients lie below the first percentile of non-IBD individuals and CD-PCNorm patients (horizontal dashed line). Non-IBD n=5, CD-PCNorm n=11, and CD-PCLow n=5. (C) Representative images of ileum crypts from non-IBD patients (left), CD-PCNorm (middle), and CD-PCLow patients (right). Scale bar represents 10 μm. (D) Level of IL-17A measured by ELISA in the serum of patients analyzed and stratified in B (sera were not available from 3 patients in CD-PCNorm and 1 patient in the CD-PCLow cohort). Data shown was tested by Student’s t-test. (E) Representative ilea hematoxylin and eosin staining (left) and histopathology scoring (right) from biopsies of patients analyzed and stratified in B. Scale bar represents 50 μm. Data in (A), (D), and (E) have been tested by Student’s t-test. p-values >0.05 are shown. *, p<0.05; ****, p<0.0001. Horizontal lines show mean±SEM. 52 10. Conclusions Comprehensive management of IBD patients, especially in children, is a major challenge for gastroenterologists and other health care professionals worldwide. In addition to its complexity, however, it represents a fascinating and dynamically changing field of medicine to which it is worth devoting one’s efforts. The approach to diagnosis, follow-up, and management of IBD has undergone a major transformation in recent decades and can be expected to continue dynamically to evolve. Together with colleagues, we intend to continue research in this area. We would be delighted if our findings would, at least in part, contribute to better care for pediatric IBD patients. 53 List of abbreviations CD Crohn’s disease CDED Crohn’s disease exclusion diet CDG congenital disorders of glycosylation CLE confocal laser endomicroscopy CRP C-reactive protein EGID eosinophilic gastrointestinal disorders EoE eosinophilic esophagitis ESPGHAN European Society of Paediatric Gastroenterology, Hepatology and Nutrition FCP fecal calprotectin FOPG fecal osteoprotegerin GWAS genome-wide association studies HMGB1 high mobility group box 1 HSV herpes simplex virus IBD inflammatory bowel disease IBDU inflammatory bowel disease unclassified IEI inborn errors of immunity IL-17 interleukin 17 JAK Janus kinase MAdCAM-1 mucosal vascular addressin cell adhesion molecule-1 miRNAs microRNAs MRI magnetic resonance imaging ncRNAs noncoding RNAs NGS next-generation sequencing PCDAI Pediatric Crohn’s Disease Activity Index PCR polymerase chain reaction PCs Paneth cells PDE phosphodiesterase PIBD pediatric inflammatory bowel disease piRNAs PIWI-interacting RNAs 54 PMM2 phosphomannomutase 2 PUCAI Pediatric Ulcerative Colitis Activity Index RNA ribonucleic acid S1P sphingosine-1-phosphate siRNAs small interfering RNAs SMAD small mothers against decapentaplegic homolog THES trichohepatoenteric syndrome TM toxic megacolon TuM2-PK tumor pyruvate kinase isoenzyme type M2 UC ulcerative colitis VEO-IBD very-early-onset inflammatory bowel disease WES whole-exome sequencing 55 List of Figures Figure 1. Clinical data of the patient with trichohepatoenteric syndrome. ............................. 12 Figure 2. Incidence rates per 100,000/year among children (0–18 years of age) newly diagnosed with IBD (A) and CD (B) in South Moravian Region, 2002 to 2017 and 2018 to 2022.......................................................................................................................................... 16 Figure 3. Mechanisms involved in the pathogenesis of inflammatory bowel disease. ........... 17 Figure 4. Tissue miRNAs involved in the development of pediatric IBD. ............................. 21 Figure 5. Diagnostic accuracy of 3-miRNA panel for identifying UC patients...................... 29 Figure 6. Key indicators of monogenic IBD in clinical practice............................................. 32 Figure 7. Therapeutic targets of novel biologics and small molecules for the treatment of inflammatory bowel disease..................................................................................................... 39 Figure 8. Functional integration of -omes relevant to the pathogenesis of the IBD-forming IBD interactome. ...................................................................................................................... 45 Figure 9. Schematic representation of the study design.......................................................... 48 Figure 10. Results of parcellated analysis comparing Crohn’s disease patients and healthy controls..................................................................................................................................... 49 Figure 11. Patients with CD manifest low ileum PC numbers, which correlate with low serum IL-17 and terminal ileitis.......................................................................................................... 51 56 List of Tables Table 1. Paris classification for Crohn’s disease..................................................................... 10 Table 2. Paris classification for ulcerative colitis.................................................................... 10 Table 3. Multiple characteristics of Crohn’s disease and ulcerative colitis. ........................... 22 Table 4. Differential diagnostics of IBD-mimicking diseases. ............................................... 30 Table 5. Warning signs for suspecting monogenic IBD.......................................................... 31 Table 6. Inborn error of immunity-associated VEO-IBD groups............................................ 33 57 References 1. Mulder DJ, Noble AJ, Justinich CJ, Duffin JM. A tale of two diseases: the history of inflammatory bowel disease. J Crohns Colitis. 2014;8(5):341-8. doi:10.1016/j.crohns.2013.09.009 2. Lee YA, Chun P, Hwang EH, Mun SW, Lee YJ, Park JH. Clinical Features and Extraintestinal Manifestations of Crohn Disease in Children. Pediatr Gastroenterol Hepatol Nutr. 2016;19(4):236-242. doi:10.5223/pghn.2016.19.4.236 3. Dhaliwal J, Walters TD, Mack DR, et al. Phenotypic Variation in Paediatric Inflammatory Bowel Disease by Age: A Multicentre Prospective Inception Cohort Study of the Canadian Children IBD Network. J Crohns Colitis. 2020;14(4):445-454. doi:10.1093/ecco-jcc/jjz106 4. Benchimol EI, Fortinsky KJ, Gozdyra P, Van den Heuvel M, Van Limbergen J, Griffiths AM. Epidemiology of pediatric inflammatory bowel disease: a systematic review of international trends. Inflamm Bowel Dis. 2011;17(1):423-39. doi:10.1002/ibd.21349 5. Ghione S, Sarter H, Fumery M, et al. Dramatic Increase in Incidence of Ulcerative Colitis and Crohn's Disease (1988-2011): A Population-Based Study of French Adolescents. Am J Gastroenterol. 2018;113(2):265-272. doi:10.1038/ajg.2017.228 6. Sýkora J, Pomahačová R, Kreslová M, Cvalínová D, Štych P, Schwarz J. Current global trends in the incidence of pediatric-onset inflammatory bowel disease. World J Gastroenterol. 2018;24(25):2741-2763. doi:10.3748/wjg.v24.i25.2741 7. Ng SC, Shi HY, Hamidi N, et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet. 2018;390(10114):2769-2778. doi:10.1016/S0140-6736(17)32448-0 8. Scott FI, Rubin DT, Kugathasan S, et al. Challenges in IBD Research: Pragmatic Clinical Research. Inflamm Bowel Dis. 2019;25(Supplement_2):S40-S47. doi:10.1093/ibd/izz085 9. Graham DB, Xavier RJ. Pathway paradigms revealed from the genetics of inflammatory bowel disease. Nature. 2020;578(7796):527-539. doi:10.1038/s41586-020- 2025-2 10. Santos MPC, Gomes C, Torres J. Familial and ethnic risk in inflammatory bowel disease. Ann Gastroenterol. 2018;31(1):14-23. doi:10.20524/aog.2017.0208 11. Kelsen J, Baldassano RN. Inflammatory bowel disease: the difference between children and adults. Inflamm Bowel Dis. 2008;14 Suppl 2:S9-11. doi:10.1002/ibd.20560 58 12. Rosen MJ, Dhawan A, Saeed SA. Inflammatory Bowel Disease in Children and Adolescents. JAMA Pediatr. 2015;169(11):1053-60. doi:10.1001/jamapediatrics.2015.1982 13. Duricova D, Burisch J, Jess T, Gower-Rousseau C, Lakatos PL, ECCO-EpiCom. Agerelated differences in presentation and course of inflammatory bowel disease: an update on the population-based literature. J Crohns Colitis. 2014;8(11):1351-61. doi:10.1016/j.crohns.2014.05.006 14. Chaparro M, Garre A, Ricart E, et al. Differences between childhood- and adulthoodonset inflammatory bowel disease: the CAROUSEL study from GETECCU. Aliment Pharmacol Ther. 2019;49(4):419-428. doi:10.1111/apt.15114 15. Krohn K, Pfeifer M, Manzey P, Koletzko S. [Inflammatory bowel diseases-the biopsychosocial reality in childhood and adolescence]. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz. 2020;63(7):839-845. doi:10.1007/s00103-020- 03166-z 16. Levine A, Koletzko S, Turner D, et al. ESPGHAN revised porto criteria for the diagnosis of inflammatory bowel disease in children and adolescents. J Pediatr Gastroenterol Nutr. 2014;58(6):795-806. doi:10.1097/MPG.0000000000000239 17. Ricciuto A, Mack DR, Huynh HQ, et al. Diagnostic Delay Is Associated With Complicated Disease and Growth Impairment in Paediatric Crohn's Disease. J Crohns Colitis. 2021;15(3):419-431. doi:10.1093/ecco-jcc/jjaa197 18. Schoepfer A, Santos J, Fournier N, et al. Systematic Analysis of the Impact of Diagnostic Delay on Bowel Damage in Paediatric Versus Adult Onset Crohn's Disease. J Crohns Colitis. 2019;13(10):1334-1342. doi:10.1093/ecco-jcc/jjz065 19. van Rheenen PF, Aloi M, Assa A, et al. The Medical Management of Paediatric Crohn's Disease: an ECCO-ESPGHAN Guideline Update. J Crohns Colitis. 2020;doi:10.1093/ecco-jcc/jjaa161 20. Turner D, Ruemmele FM, Orlanski-Meyer E, et al. Management of Paediatric Ulcerative Colitis, Part 1: Ambulatory Care-An Evidence-based Guideline From European Crohn's and Colitis Organization and European Society of Paediatric Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr. 2018;67(2):257-291. doi:10.1097/MPG.0000000000002035 21. Turner D, Ruemmele FM, Orlanski-Meyer E, et al. Management of Paediatric Ulcerative Colitis, Part 2: Acute Severe Colitis-An Evidence-based Consensus Guideline From the European Crohn's and Colitis Organization and the European Society of Paediatric 59 Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr. 2018;67(2):292- 310. doi:10.1097/MPG.0000000000002036 22. Ruemmele FM, Veres G, Kolho KL, et al. Consensus guidelines of ECCO/ESPGHAN on the medical management of pediatric Crohn's disease. J Crohns Colitis. 2014;8(10):1179- 207. doi:10.1016/j.crohns.2014.04.005 23. Guariso G, Gasparetto M. Treating children with inflammatory bowel disease: Current and new perspectives. World J Gastroenterol. 2017;23(30):5469-5485. doi:10.3748/wjg.v23.i30.5469 24. Wren AA, Maddux MH. Integrated Multidisciplinary Treatment for Pediatric Inflammatory Bowel Disease. Children (Basel). 2021;8(2)doi:10.3390/children8020169 25. McCann LJ, Newell SJ. Survey of paediatric complementary and alternative medicine use in health and chronic illness. Arch Dis Child. 2006;91(2):173-4. doi:10.1136/adc.2004.052514 26. Singh UP, Singh NP, Busbee B, et al. Alternative medicines as emerging therapies for inflammatory bowel diseases. Int Rev Immunol. 2012;31(1):66-84. doi:10.3109/08830185.2011.642909 27. Lin SC, Cheifetz AS. The Use of Complementary and Alternative Medicine in Patients With Inflammatory Bowel Disease. Gastroenterol Hepatol (N Y). 2018;14(7):415-425. 28. Kelsen JR, Russo P, Sullivan KE. Early-Onset Inflammatory Bowel Disease. Immunol Allergy Clin North Am. 2019;39(1):63-79. doi:10.1016/j.iac.2018.08.008 29. Lev-Tzion R, Turner D. Is pediatric IBD treatment different than in adults? Minerva Gastroenterol Dietol. 2012;58(2):137-50. 30. Tontini GE, Vecchi M, Pastorelli L, Neurath MF, Neumann H. Differential diagnosis in inflammatory bowel disease colitis: state of the art and future perspectives. World J Gastroenterol. 2015;21(1):21-46. doi:10.3748/wjg.v21.i1.21 31. IBD Working Group of the European Society for Paediatric Gastroenterology HpaN. Inflammatory bowel disease in children and adolescents: recommendations for diagnosis--the Porto criteria. J Pediatr Gastroenterol Nutr. Jul 2005;41(1):1-7. 32. Birimberg-Schwartz L, Zucker DM, Akriv A, et al. Development and Validation of Diagnostic Criteria for IBD Subtypes Including IBD-unclassified in Children: a Multicentre Study From the Pediatric IBD Porto Group of ESPGHAN. J Crohns Colitis. 2017;11(9):1078- 1084. doi:10.1093/ecco-jcc/jjx053 33. Silverberg MS, Satsangi J, Ahmad T, et al. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: report of a Working Party of the 60 2005 Montreal World Congress of Gastroenterology. Can J Gastroenterol. 2005;19 Suppl A:5A-36A. doi:10.1155/2005/269076 34. Satsangi J, Silverberg MS, Vermeire S, Colombel JF. The Montreal classification of inflammatory bowel disease: controversies, consensus, and implications. Gut. 2006;55(6):749-53. doi:10.1136/gut.2005.082909 35. Levine A, Griffiths A, Markowitz J, et al. Pediatric modification of the Montreal classification for inflammatory bowel disease: the Paris classification. Inflamm Bowel Dis. 2011;17(6):1314-21. doi:10.1002/ibd.21493 36. Assa A, Rinawi F, Shamir R. The Long-Term Predictive Properties of the Paris Classification in Paediatric Inflammatory Bowel Disease Patients. J Crohns Colitis. 2018;12(1):39-47. doi:10.1093/ecco-jcc/jjx125 37. Arai K. Very Early-Onset Inflammatory Bowel Disease: A Challenging Field for Pediatric Gastroenterologists. Pediatr Gastroenterol Hepatol Nutr. 2020;23(5):411-422. doi:10.5223/pghn.2020.23.5.411 38. Uhlig HH, Schwerd T, Koletzko S, et al. The diagnostic approach to monogenic very early onset inflammatory bowel disease. Gastroenterology. 2014;147(5):990-1007.e3. doi:10.1053/j.gastro.2014.07.023 39. Uhlig HH, Charbit-Henrion F, Kotlarz D, et al. Clinical Genomics for the Diagnosis of Monogenic Forms of Inflammatory Bowel Disease: A Position Paper From the Paediatric IBD Porto Group of European Society of Paediatric Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr. 2021;72(3):456-473. doi:10.1097/MPG.0000000000003017 40. de Lange KM, Moutsianas L, Lee JC, et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat Genet. 2017;49(2):256-261. doi:10.1038/ng.3760 41. Jezernik G, Mičetić-Turk D, Potočnik U. Molecular Genetic Architecture of Monogenic Pediatric IBD Differs from Complex Pediatric and Adult IBD. J Pers Med. 2020;10(4)doi:10.3390/jpm10040243 42. Jabandziev P, Hlavackova E, Bily V, et al. Trichohepatoenteric syndrome in a patient with TTC37 mutations - case report. Gastroent Hepatol. 2020;74(6):481-487. doi:10.48095/ccgh20201 43. Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat Rev Gastroenterol Hepatol. 2015;12(12):720-7. doi:10.1038/nrgastro.2015.150 61 44. Kaplan GG, Windsor JW. The four epidemiological stages in the global evolution of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol. 2021;18(1):56-66. doi:10.1038/s41575-020-00360-x 45. Bernstein CN, Rawsthorne P, Cheang M, Blanchard JF. A population-based case control study of potential risk factors for IBD. Am J Gastroenterol. 2006;101(5):993-1002. doi:10.1111/j.1572-0241.2006.00381.x 46. Rajbhandari R, Blakemore S, Gupta N, et al. Crohn's disease in low and lower-middle income countries: A scoping review. World J Gastroenterol. 2020;26(43):6891-6908. doi:10.3748/wjg.v26.i43.6891 47. Kamm MA. Rapid changes in epidemiology of inflammatory bowel disease. Lancet. 2018;390(10114):2741-2742. doi:10.1016/S0140-6736(17)32669-7 48. Coward S, Clement F, Benchimol EI, et al. Past and Future Burden of Inflammatory Bowel Diseases Based on Modeling of Population-Based Data. Gastroenterology. 2019;156(5):1345-1353.e4. doi:10.1053/j.gastro.2019.01.002 49. Principi M, Labarile N, Bianchi FP, et al. The Cost of Inflammatory Bowel Disease Management Matches with Clinical Course: A Single Outpatient Centre Analysis. Int J Environ Res Public Health. 2020;17(12)doi:10.3390/ijerph17124549 50. El-Matary W, Kuenzig ME, Singh H, et al. Disease-Associated Costs in Children With Inflammatory Bowel Disease: A Systematic Review. Inflamm Bowel Dis. 2020;26(2):206- 215. doi:10.1093/ibd/izz120 51. Eszter Müller K, Laszlo Lakatos P, Papp M, Veres G. Incidence and paris classification of pediatric inflammatory bowel disease. Gastroenterol Res Pract. 2014;2014:904307. doi:10.1155/2014/904307 52. Collaborators GIBD. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol. 2020;5(1):17-30. doi:10.1016/S2468-1253(19)30333-4 53. Burisch J, Pedersen N, Čuković-Čavka S, et al. East-West gradient in the incidence of inflammatory bowel disease in Europe: the ECCO-EpiCom inception cohort. Gut. 2014;63(4):588-97. doi:10.1136/gutjnl-2013-304636 54. Hradský O. Rizikové faktory vzniku a průběhu zánětlivých střevních onemocnění u dětí. Univerzita Karlova; 2017. 55. Safarpour AR, Mehrabi M, Keshtkar A, Edjtehadi F, Bagheri Lankarani K. Systematic review and meta-analysis of the incidence and prevalence and 30-year trend of inflammatory 62 bowel diseases in Asia: a study protocol. BMJ Open. 2019;9(11):e031854. doi:10.1136/bmjopen-2019-031854 56. Jabandziev P, Pinkasova T, Kunovsky L, et al. Regional Incidence of Inflammatory Bowel Disease in a Czech Pediatric Population: 16 Years of Experience (2002-2017). J Pediatr Gastroenterol Nutr. 2020;70(5):586-592. doi:10.1097/MPG.0000000000002660 57. Roberts SE, Thorne K, Thapar N, et al. A Systematic Review and Meta-analysis of Paediatric Inflammatory Bowel Disease Incidence and Prevalence Across Europe. J Crohns Colitis. 2020;14(8):1119-1148. doi:10.1093/ecco-jcc/jjaa037 58. Pozler O, Maly J, Bonova O, et al. Incidence of Crohn disease in the Czech Republic in the years 1990 to 2001 and assessment of pediatric population with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2006;42(2):186-9. doi:10.1097/01.mpg.0000189328.47150.bc 59. Schwarz J, Sýkora J, Cvalínová D, et al. Inflammatory bowel disease incidence in Czech children: A regional prospective study, 2000-2015. World J Gastroenterol. 2017;23(22):4090-4101. doi:10.3748/wjg.v23.i22.4090 60. Scott FI, Rubin DT, Kugathasan S, et al. Challenges in IBD Research: Pragmatic Clinical Research. Inflamm Bowel Dis. 2019;25(Suppl 2):S40-S47. doi:10.1093/ibd/izz085 61. Lee SH, Kwon JE, Cho ML. Immunological pathogenesis of inflammatory bowel disease. Intest Res. 2018;16(1):26-42. doi:10.5217/ir.2018.16.1.26 62. Guan Q. A Comprehensive Review and Update on the Pathogenesis of Inflammatory Bowel Disease. J Immunol Res. 2019;2019:7247238. doi:10.1155/2019/7247238 63. Ramos GP, Papadakis KA. Mechanisms of Disease: Inflammatory Bowel Diseases. Mayo Clin Proc. 2019;94(1):155-165. doi:10.1016/j.mayocp.2018.09.013 64. Chichlowski M, Hale LP. Bacterial-mucosal interactions in inflammatory bowel disease: an alliance gone bad. Am J Physiol Gastrointest Liver Physiol. 2008;295(6):G1139- 49. doi:10.1152/ajpgi.90516.2008 65. Antoni L, Nuding S, Weller D, et al. Human colonic mucus is a reservoir for antimicrobial peptides. J Crohns Colitis. 2013;7(12):e652-64. doi:10.1016/j.crohns.2013.05.006 66. Antoni L, Nuding S, Wehkamp J, Stange EF. Intestinal barrier in inflammatory bowel disease. World J Gastroenterol. 2014;20(5):1165-79. doi:10.3748/wjg.v20.i5.1165 67. Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018;11(1):1-10. doi:10.1007/s12328-017-0813-5 63 68. Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020;30(6):492-506. doi:10.1038/s41422-020-0332-7 69. Capaldo CT, Powell DN, Kalman D. Layered defense: how mucus and tight junctions seal the intestinal barrier. J Mol Med (Berl). 2017;95(9):927-934. doi:10.1007/s00109-017- 1557-x 70. Okumura R, Takeda K. Roles of intestinal epithelial cells in the maintenance of gut homeostasis. Exp Mol Med. 2017;49(5):e338. doi:10.1038/emm.2017.20 71. Mann ER, Li X. Intestinal antigen-presenting cells in mucosal immune homeostasis: crosstalk between dendritic cells, macrophages and B-cells. World J Gastroenterol. 2014;20(29):9653-64. doi:10.3748/wjg.v20.i29.9653 72. Borg-Bartolo SP, Boyapati RK, Satsangi J, Kalla R. Precision medicine in inflammatory bowel disease: concept, progress and challenges. F1000Res. 2020;9doi:10.12688/f1000research.20928.1 73. Ananthakrishnan AN, Bernstein CN, Iliopoulos D, et al. Environmental triggers in IBD: a review of progress and evidence. Nat Rev Gastroenterol Hepatol. 2018;15(1):39-49. doi:10.1038/nrgastro.2017.136 74. Elten M, Benchimol EI, Fell DB, et al. Residential Greenspace in Childhood Reduces Risk of Pediatric Inflammatory Bowel Disease: A Population-Based Cohort Study. Am J Gastroenterol. 2021;116(2):347-353. doi:10.14309/ajg.0000000000000990 75. Elten M, Benchimol EI, Fell DB, et al. Ambient air pollution and the risk of pediatriconset inflammatory bowel disease: A population-based cohort study. Environ Int. 2020;138:105676. doi:10.1016/j.envint.2020.105676 76. Nguyen LH, Örtqvist AK, Cao Y, et al. Antibiotic use and the development of inflammatory bowel disease: a national case-control study in Sweden. Lancet Gastroenterol Hepatol. 2020;5(11):986-995. doi:10.1016/S2468-1253(20)30267-3 77. Khalili H, Håkansson N, Chan SS, et al. Adherence to a Mediterranean diet is associated with a lower risk of later-onset Crohn's disease: results from two large prospective cohort studies. Gut. 2020;69(9):1637-1644. doi:10.1136/gutjnl-2019-319505 78. Mentella MC, Scaldaferri F, Pizzoferrato M, Gasbarrini A, Miggiano GAD. Nutrition, IBD and Gut Microbiota: A Review. Nutrients. 2020;12(4)doi:10.3390/nu12040944 79. Piovani D, Danese S, Peyrin-Biroulet L, Nikolopoulos GK, Lytras T, Bonovas S. Environmental Risk Factors for Inflammatory Bowel Diseases: An Umbrella Review of Metaanalyses. Gastroenterology. 2019;157(3):647-659.e4. doi:10.1053/j.gastro.2019.04.016 64 80. Abegunde AT, Muhammad BH, Bhatti O, Ali T. Environmental risk factors for inflammatory bowel diseases: Evidence based literature review. World J Gastroenterol. 2016;22(27):6296-317. doi:10.3748/wjg.v22.i27.6296 81. Olivera P, Danese S, Jay N, Natoli G, Peyrin-Biroulet L. Big data in IBD: a look into the future. Nat Rev Gastroenterol Hepatol. 2019;16(5):312-321. doi:10.1038/s41575-019- 0102-5 82. Liu JZ, Anderson CA. Genetic studies of Crohn's disease: past, present and future. Best Pract Res Clin Gastroenterol. 2014;28(3):373-86. doi:10.1016/j.bpg.2014.04.009 83. Liu JZ, van Sommeren S, Huang H, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet. 2015;47(9):979-986. doi:10.1038/ng.3359 84. Hu S, Uniken Venema WT, Westra HJ, et al. Inflammation status modulates the effect of host genetic variation on intestinal gene expression in inflammatory bowel disease. Nat Commun. 2021;12(1):1122. doi:10.1038/s41467-021-21458-z 85. Jabandziev P, Bohosova J, Pinkasova T, Kunovsky L, Slaby O, Goel A. The Emerging Role of Noncoding RNAs in Pediatric Inflammatory Bowel Disease. Inflamm Bowel Dis. 2020;26(7):985-993. doi:10.1093/ibd/izaa009 86. Zhang P, Wu W, Chen Q, Chen M. Non-Coding RNAs and their Integrated Networks. J Integr Bioinform. 2019;16(3)doi:10.1515/jib-2019-0027 87. Martens-Uzunova ES, Olvedy M, Jenster G. Beyond microRNA--novel RNAs derived from small non-coding RNA and their implication in cancer. Cancer Lett. 2013;340(2):201- 11. doi:10.1016/j.canlet.2012.11.058 88. Anastasiadou E, Jacob LS, Slack FJ. Non-coding RNA networks in cancer. Nat Rev Cancer. 2018;18(1):5-18. doi:10.1038/nrc.2017.99 89. Salviano-Silva A, Lobo-Alves SC, Almeida RC, Malheiros D, Petzl-Erler ML. Besides Pathology: Long Non-Coding RNA in Cell and Tissue Homeostasis. Noncoding RNA. 2018;4(1)doi:10.3390/ncrna4010003 90. Iborra M, Bernuzzi F, Correale C, et al. Identification of serum and tissue micro-RNA expression profiles in different stages of inflammatory bowel disease. Clin Exp Immunol. 2013;173(2):250-8. doi:10.1111/cei.12104 91. Wu LY, Ma XP, Shi Y, et al. Alterations in microRNA expression profiles in inflamed and noninflamed ascending colon mucosae of patients with active Crohn's disease. J Gastroenterol Hepatol. 2017;32(10):1706-1715. doi:10.1111/jgh.13778 65 92. Zahm AM, Hand NJ, Tsoucas DM, Le Guen CL, Baldassano RN, Friedman JR. Rectal microRNAs are perturbed in pediatric inflammatory bowel disease of the colon. J Crohns Colitis. 2014;8(9):1108-17. doi:10.1016/j.crohns.2014.02.012 93. Verdier J, Breunig IR, Ohse MC, et al. Faecal Micro-RNAs in Inflammatory Bowel Diseases. J Crohns Colitis. 2020;14(1):110-117. doi:10.1093/ecco-jcc/jjz120 94. Dragoni G, Innocenti T, Galli A. Biomarkers of Inflammation in Inflammatory Bowel Disease: How Long before Abandoning Single-Marker Approaches? Dig Dis. 2021;39(3):190-203. doi:10.1159/000511641 95. Park JH, Peyrin-Biroulet L, Eisenhut M, Shin JI. IBD immunopathogenesis: A comprehensive review of inflammatory molecules. Autoimmun Rev. 2017;16(4):416-426. doi:10.1016/j.autrev.2017.02.013 96. Adamcova M, Bajer M, Bajerova K, et al. Czech Working Group for Paediatric Gastroenterology and Nutrition guidelines for diagnostics and treatment of inflammatory bowel diseases in children. Ces-slov Pediat. 2012;67(4)(2) 97. Bronsky J, Berankova K, Cerna Z, et al. Czech Working Group for Paediatric Gastroenterology and Nutrition guidelines for diagnostics and treatment of inflammatory bowel diseases in children – 1st edition update. Gastroent Hepatol. 2017;71(1):11-18. doi:10.14735/amgh20171 98. Hyams JS, Ferry GD, Mandel FS, et al. Development and validation of a pediatric Crohn's disease activity index. J Pediatr Gastroenterol Nutr. May 1991;12(4):439-47. 99. Hyams J, Markowitz J, Otley A, et al. Evaluation of the pediatric crohn disease activity index: a prospective multicenter experience. J Pediatr Gastroenterol Nutr. 2005;41(4):416-21. doi:10.1097/01.mpg.0000183350.46795.42 100. Turner D, Levine A, Walters TD, et al. Which PCDAI Version Best Reflects Intestinal Inflammation in Pediatric Crohn Disease? J Pediatr Gastroenterol Nutr. 2017;64(2):254-260. doi:10.1097/MPG.0000000000001227 101. Turner D, Hyams J, Markowitz J, et al. Appraisal of the pediatric ulcerative colitis activity index (PUCAI). Inflamm Bowel Dis. 2009;15(8):1218-23. doi:10.1002/ibd.20867 102. Dotson JL, Crandall WV, Zhang P, et al. Feasibility and validity of the pediatric ulcerative colitis activity index in routine clinical practice. J Pediatr Gastroenterol Nutr. Feb 2015;60(2):200-4. doi:10.1097/MPG.0000000000000568 103. Oliveira SB, Monteiro IM. Diagnosis and management of inflammatory bowel disease in children. BMJ. 2017;357:j2083. doi:10.1136/bmj.j2083 104. Zbořil V. Idiopatické střevní záněty. Mladá fronta; 2018. 66 105. Greuter T, Vavricka SR. Extraintestinal manifestations in inflammatory bowel disease - epidemiology, genetics, and pathogenesis. Expert Rev Gastroenterol Hepatol. 2019;13(4):307-317. doi:10.1080/17474124.2019.1574569 106. Yu YR, Rodriguez JR. Clinical presentation of Crohn's, ulcerative colitis, and indeterminate colitis: Symptoms, extraintestinal manifestations, and disease phenotypes. Semin Pediatr Surg. 2017;26(6):349-355. doi:10.1053/j.sempedsurg.2017.10.003 107. Jang HJ, Kang B, Choe BH. The difference in extraintestinal manifestations of inflammatory bowel disease for children and adults. Transl Pediatr. J2019;8(1):4-15. doi:10.21037/tp.2019.01.06 108. Aloi M, D'Arcangelo G, Pofi F, et al. Presenting features and disease course of pediatric ulcerative colitis. J Crohns Colitis. 2013;7(11):e509-15. doi:10.1016/j.crohns.2013.03.007 109. Birimberg-Schwartz L, Wilson DC, Kolho KL, et al. pANCA and ASCA in Children with IBD-Unclassified, Crohn's Colitis, and Ulcerative Colitis-A Longitudinal Report from the IBD Porto Group of ESPGHAN. Inflamm Bowel Dis. 2016;22(8):1908-14. doi:10.1097/MIB.0000000000000784 110. Ashton JJ, Borca F, Mossotto E, Phan HTT, Ennis S, Beattie RM. Analysis and Hierarchical Clustering of Blood Results Before Diagnosis in Pediatric Inflammatory Bowel Disease. Inflamm Bowel Dis. 2020;26(3):469-475. doi:10.1093/ibd/izy369 111. Ricciuto A, Griffiths AM. Clinical value of fecal calprotectin. Crit Rev Clin Lab Sci. 2019;56(5):307-320. doi:10.1080/10408363.2019.1619159 112. Walker GJ, Chanchlani N, Thomas A, et al. Primary care faecal calprotectin testing in children with suspected inflammatory bowel disease: a diagnostic accuracy study. Arch Dis Child. 2020;105(10):957-963. doi:10.1136/archdischild-2019-317823 113. D'Amico F, Rubin DT, Kotze PG, et al. International consensus on methodological issues in standardization of fecal calprotectin measurement in inflammatory bowel diseases. United European Gastroenterol J. 2021;9(4):451-460. doi:10.1002/ueg2.12069 114. D'Amico F, Nancey S, Danese S, Peyrin-Biroulet L. A Practical Guide for Faecal Calprotectin Measurement: Myths and Realities. J Crohns Colitis. 2021;15(1):152-161. doi:10.1093/ecco-jcc/jjaa093 115. Koninckx CR, Donat E, Benninga MA, et al. The Use of Fecal Calprotectin Testing in Paediatric Disorders: A Position Paper of the European Society for Paediatric Gastroenterology and Nutrition Gastroenterology Committee. J Pediatr Gastroenterol Nutr. 2021;72(4):617-640. doi:10.1097/MPG.0000000000003046 67 116. Ricciuto A, Aardoom M, Orlanski-Meyer E, et al. Predicting Outcomes in Pediatric Crohn's Disease for Management Optimization: Systematic Review and Consensus Statements From the Pediatric Inflammatory Bowel Disease-Ahead Program. Gastroenterology. 2021;160(1):403-436.e26. doi:10.1053/j.gastro.2020.07.065 117. Orlanski-Meyer E, Aardoom M, Ricciuto A, et al. Predicting Outcomes in Pediatric Ulcerative Colitis for Management Optimization: Systematic Review and Consensus Statements From the Pediatric Inflammatory Bowel Disease-Ahead Program. Gastroenterology. 2021;160(1):378-402.e22. doi:10.1053/j.gastro.2020.07.066 118. Spiceland CM, Lodhia N. Endoscopy in inflammatory bowel disease: Role in diagnosis, management, and treatment. World J Gastroenterol. 2018;24(35):4014-4020. doi:10.3748/wjg.v24.i35.4014 119. Bharadwaj S, Narula N, Tandon P, Yaghoobi M. Role of endoscopy in inflammatory bowel disease. Gastroenterol Rep (Oxf). 2018;6(2):75-82. doi:10.1093/gastro/goy006 120. Limdi JK, Picco M, Farraye FA. A review of endoscopic scoring systems and their importance in a treat-to-target approach in inflammatory bowel disease (with videos). Gastrointest Endosc. 2020;91(4):733-745. doi:10.1016/j.gie.2019.11.032 121. Daperno M, D'Haens G, Van Assche G, et al. Development and validation of a new, simplified endoscopic activity score for Crohn's disease: the SES-CD. Gastrointest Endosc. 2004;60(4):505-12. doi:10.1016/s0016-5107(04)01878-4 122. Travis SP, Schnell D, Krzeski P, et al. Developing an instrument to assess the endoscopic severity of ulcerative colitis: the Ulcerative Colitis Endoscopic Index of Severity (UCEIS). Gut. 2012;61(4):535-42. doi:10.1136/gutjnl-2011-300486 123. Rutgeerts P, Geboes K, Vantrappen G, Beyls J, Kerremans R, Hiele M. Predictability of the postoperative course of Crohn's disease. Gastroenterology. 1990;99(4):956-63. doi:10.1016/0016-5085(90)90613-6 124. Thomson M, Tringali A, Dumonceau JM, et al. Paediatric Gastrointestinal Endoscopy: European Society for Paediatric Gastroenterology Hepatology and Nutrition and European Society of Gastrointestinal Endoscopy Guidelines. J Pediatr Gastroenterol Nutr. 2017;64(1):133-153. doi:10.1097/MPG.0000000000001408 125. Oliva S, Thomson M, de Ridder L, et al. Endoscopy in Pediatric Inflammatory Bowel Disease: A Position Paper on Behalf of the Porto IBD Group of the European Society for Pediatric Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr. 2018;67(3):414-430. doi:10.1097/MPG.0000000000002092 68 126. Broekaert I, Tzivinikos C, Narula P, et al. European Society for Paediatric Gastroenterology, Hepatology and Nutrition Position Paper on Training in Paediatric Endoscopy. J Pediatr Gastroenterol Nutr. 2020;70(1):127-140. doi:10.1097/MPG.0000000000002496 127. Van de Vijver E, Heida A, Ioannou S, et al. Test Strategies to Predict Inflammatory Bowel Disease Among Children With Nonbloody Diarrhea. Pediatrics. 2020;146(2)doi:10.1542/peds.2019-2235 128. Sahn B, De Matos V, Stein R, et al. Histological features of ileitis differentiating pediatric Crohn disease from ulcerative colitis with backwash ileitis. Dig Liver Dis. 2018;50(2):147-153. doi:10.1016/j.dld.2017.10.006 129. Glass J, Alcalá HE, Tobin M. The Value of Obtaining Colonic Mucosal Biopsies of Grossly Normal Tissue in Pediatric Patients. J Pediatr Gastroenterol Nutr. 2021;72(5):677- 682. doi:10.1097/MPG.0000000000003038 130. Conrad MA, Carreon CK, Dawany N, Russo P, Kelsen JR. Distinct Histopathological Features at Diagnosis of Very Early Onset Inflammatory Bowel Disease. J Crohns Colitis. 2019;13(5):615-625. doi:10.1093/ecco-jcc/jjy212 131. Magro F, Langner C, Driessen A, et al. European consensus on the histopathology of inflammatory bowel disease. J Crohns Colitis. 2013;7(10):827-51. doi:10.1016/j.crohns.2013.06.001 132. Engel PJH, Fiehn AK, Munck LK, Kristensson M. The subtypes of microscopic colitis from a pathologist's perspective: past, present and future. Ann Transl Med. 2018;6(3):69. doi:10.21037/atm.2017.03.16 133. Schreiber-Dietrich D, Chiorean L, Cui XW, et al. Particularities of Crohn's disease in pediatric patients: current status and perspectives regarding imaging modalities. Expert Rev Gastroenterol Hepatol. 2015;9(10):1313-25. doi:10.1586/17474124.2015.1083420 134. van Wassenaer EA, de Voogd FAE, van Rijn RR, et al. Diagnostic Accuracy of Transabdominal Ultrasound in Detecting Intestinal Inflammation in Paediatric IBD Patients-a Systematic Review. J Crohns Colitis. 2019;13(12):1501-1509. doi:10.1093/ecco-jcc/jjz085 135. Duvoisin G, Lopez RN, Day AS, Lemberg DA, Gearry RB, Leach ST. Novel Biomarkers and the Future Potential of Biomarkers in Inflammatory Bowel Disease. Mediators Inflamm. 2017;2017:1936315. doi:10.1155/2017/1936315 136. Torres J, Petralia F, Sato T, et al. Serum Biomarkers Identify Patients Who Will Develop Inflammatory Bowel Diseases Up to 5 Years Before Diagnosis. Gastroenterology. 2020;159(1):96-104. doi:10.1053/j.gastro.2020.03.007 69 137. Pinto-Lopes P, Afonso J, Pinto-Lopes R, et al. Serum Dipeptidyl Peptidase 4: A Predictor of Disease Activity and Prognosis in Inflammatory Bowel Disease. Inflamm Bowel Dis. 2020;26(11):1707-1719. doi:10.1093/ibd/izz319 138. Pinto-Lopes P, Melo F, Afonso J, et al. Fecal Dipeptidyl Peptidase-4: An Emergent Biomarker in Inflammatory Bowel Disease. Clin Transl Gastroenterol. 2021;12(3):e00320. doi:10.14309/ctg.0000000000000320 139. Perry C, Kapur N, Barrett TA. DPP-4 as a Novel Biomarker for Inflammatory Bowel Disease: Is It Ready for Clinical Use? Inflamm Bowel Dis. 2020;26(11):1720-1721. doi:10.1093/ibd/izz320 140. Jabandziev P, Kakisaka T, Bohosova J, et al. MicroRNAs in Colon Tissue of Pediatric Ulcerative Pancolitis Patients Allow Detection and Prognostic Stratification. J Clin Med. 2021;10(6). doi:10.3390/jcm10061325 141. Chen P, Zhou G, Lin J, et al. Serum Biomarkers for Inflammatory Bowel Disease. Front Med (Lausanne). 2020;7:123. doi:10.3389/fmed.2020.00123 142. Leach ST, Day AS, Messenger R, et al. Fecal Markers of Inflammation and Disease Activity in Pediatric Crohn Disease: Results from the ImageKids Study. J Pediatr Gastroenterol Nutr. 2020;70(5):580-585. doi:10.1097/MPG.0000000000002615 143. Heida A, Van de Vijver E, van Ravenzwaaij D, et al. Predicting inflammatory bowel disease in children with abdominal pain and diarrhoea: calgranulin-C versus calprotectin stool tests. Arch Dis Child. 2018;103(6):565-571. doi:10.1136/archdischild-2017-314081 144. Naganuma M, Hosoe N, Kanai T, Ogata H. Recent trends in diagnostic techniques for inflammatory bowel disease. Korean J Intern Med. 2015;30(3):271-8. doi:10.3904/kjim.2015.30.3.271 145. Buchner AM. Confocal Laser Endomicroscopy in the Evaluation of Inflammatory Bowel Disease. Inflamm Bowel Dis. 2019;25(8):1302-1312. doi:10.1093/ibd/izz021 146. Kunovský L, Kala Z, Kroupa R, et al. Confocal laser endomicroscopy in the diagnostics of esophageal diseases: a pilot study. Vnitr Lek. 2020;66(5):62-68. 147. Goodwin S, McPherson JD, McCombie WR. Coming of age: ten years of nextgeneration sequencing technologies. Nat Rev Genet. 2016;17(6):333-51. doi:10.1038/nrg.2016.49 148. Slatko BE, Gardner AF, Ausubel FM. Overview of Next-Generation Sequencing Technologies. Curr Protoc Mol Biol. 2018;122(1):e59. doi:10.1002/cpmb.59 149. Slaba K, Noskova H, Vesela P, et al. Novel Splicing Variant in the. Front Genet. 2020;11:561054. doi:10.3389/fgene.2020.561054 70 150. Siegel CA, Bernstein CN. Identifying Patients With Inflammatory Bowel Diseases at High vs Low Risk of Complications. Clin Gastroenterol Hepatol. 2020;18(6):1261-1267. doi:10.1016/j.cgh.2019.11.034 151. Naviglio S, Lacorte D, Lucafò M, et al. Causes of Treatment Failure in Children With Inflammatory Bowel Disease Treated With Infliximab: A Pharmacokinetic Study. J Pediatr Gastroenterol Nutr. 2019;68(1):37-44. doi:10.1097/MPG.0000000000002112 152. Gecse KB, Vermeire S. Differential diagnosis of inflammatory bowel disease: imitations and complications. Lancet Gastroenterol Hepatol. 2018;3(9):644-653. doi:10.1016/S2468-1253(18)30159-6 153. Kliegman R. Nelson Textbook of Pediatrics. vol 21. Elsevier; 2020. 154. Hamdeh S, Micic D, Hanauer S. Drug-Induced Colitis. Clin Gastroenterol Hepatol. 2020; 30:S1542-3565(20)30614-5. doi:10.1016/j.cgh.2020.04.069 155. Nambu R, Muise AM. Advanced Understanding of Monogenic Inflammatory Bowel Disease. Front Pediatr. 2020;8:618918. doi:10.3389/fped.2020.618918 156. Kelsen JR, Sullivan KE, Rabizadeh S, et al. NASPGHAN Position Paper on The Evaluation and Management for Patients with Very Early-Onset Inflammatory Bowel Disease (VEO-IBD). J Pediatr Gastroenterol Nutr. 2019;doi:10.1097/MPG.0000000000002567 157. Bousfiha A, Jeddane L, Picard C, et al. Human Inborn Errors of Immunity: 2019 Update of the IUIS Phenotypical Classification. J Clin Immunol. 2020;40(1):66-81. doi:10.1007/s10875-020-00758-x 158. Shim JO. Recent Advance in Very Early Onset Inflammatory Bowel Disease. Pediatr Gastroenterol Hepatol Nutr. 2019;22(1):41-49. doi:10.5223/pghn.2019.22.1.41 159. Nambu R, Warner N, Mulder DJ, et al. A Systematic Review of Monogenic Inflammatory Bowel Disease. Clin Gastroenterol Hepatol. 2021; 18:S1542-3565(21)003311. doi:10.1016/j.cgh.2021.03.021 160. Chang IJ, He M, Lam CT. Congenital disorders of glycosylation. Ann Transl Med. 2018;6(24):477. doi:10.21037/atm.2018.10.45 161. Francisco R, Marques-da-Silva D, Brasil S, et al. The challenge of CDG diagnosis. Mol Genet Metab. 2019;126(1):1-5. doi:10.1016/j.ymgme.2018.11.003 162. Bogdańska A, Lipiński P, Szymańska-Rożek P, et al. Clinical, biochemical and molecular phenotype of congenital disorders of glycosylation: long-term follow-up. Orphanet J Rare Dis. 2021;16(1):17. doi:10.1186/s13023-020-01657-5 71 163. Witters P, Honzik T, Bauchart E, et al. Long-term follow-up in PMM2-CDG: are we ready to start treatment trials? Genet Med. 2019;21(5):1181-1188. doi:10.1038/s41436-018- 0301-4 164. Schiff M, Roda C, Monin ML, et al. Clinical, laboratory and molecular findings and long-term follow-up data in 96 French patients with PMM2-CDG (phosphomannomutase 2congenital disorder of glycosylation) and review of the literature. J Med Genet. 2017;54(12):843-851. doi:10.1136/jmedgenet-2017-104903 165. Francisco R, Pascoal C, Marques-da-Silva D, et al. New Insights into Immunological Involvement in Congenital Disorders of Glycosylation (CDG) from a People-Centric Approach. J Clin Med. 2020;9(7)doi:10.3390/jcm9072092 166. Narumi S, Amano N, Ishii T, et al. SAMD9 mutations cause a novel multisystem disorder, MIRAGE syndrome, and are associated with loss of chromosome 7. Nat Genet. 2016;48(7):792-7. doi:10.1038/ng.3569 167. Shima H, Hayashi M, Tachibana T, et al. MIRAGE syndrome is a rare cause of 46,XY DSD born SGA without adrenal insufficiency. PLoS One. 2018;13(11):e0206184. doi:10.1371/journal.pone.0206184 168. Perisa MP, Rose MJ, Varga E, Kamboj MK, Spencer JD, Bajwa RPS. A novel SAMD9 variant identified in patient with MIRAGE syndrome: Further defining syndromic phenotype and review of previous cases. Pediatr Blood Cancer. 2019;66(7):e27726. doi:10.1002/pbc.27726 169. Onuma S, Wada T, Araki R, et al. MIRAGE syndrome caused by a novel missense variant (p.Ala1479Ser) in the. Hum Genome Var. 2020;7:4. doi:10.1038/s41439-020-0091-5 170. Ishiwa S, Kamei K, Tanase-Nakao K, et al. A girl with MIRAGE syndrome who developed steroid-resistant nephrotic syndrome: a case report. BMC Nephrol. 2020;21(1):340. doi:10.1186/s12882-020-02011-4 171. Formankova R, Kanderova V, Rackova M, et al. Novel SAMD9 Mutation in a Patient With Immunodeficiency, Neutropenia, Impaired Anti-CMV Response, and Severe Gastrointestinal Involvement. Front Immunol. 2019;10:2194. doi:10.3389/fimmu.2019.02194 172. Furuta GT, Katzka DA. Eosinophilic Esophagitis. N Engl J Med. Oct 2015;373(17):1640-8. doi:10.1056/NEJMra1502863 173. Zhang M, Li Y. Eosinophilic gastroenteritis: A state-of-the-art review. J Gastroenterol Hepatol. 2017;32(1):64-72. doi:10.1111/jgh.13463 72 174. Hlouskova E, Bajerova K, Pecl J, Jabandziev P, Jezova M, Tuma J. Eosinophilic enteritis - case report of a rare manifestation and review of updates. Gastroent Hepatol. 2020;74(6):492-496. doi:10.48095/ccgh2020492 175. Matsushita T, Maruyama R, Ishikawa N, et al. The number and distribution of eosinophils in the adult human gastrointestinal tract: a study and comparison of racial and environmental factors. Am J Surg Pathol. 2015;39(4):521-7. doi:10.1097/PAS.0000000000000370 176. Mehta P, Furuta GT. Eosinophils in Gastrointestinal Disorders: Eosinophilic Gastrointestinal Diseases, Celiac Disease, Inflammatory Bowel Diseases, and Parasitic Infections. Immunol Allergy Clin North Am. 2015;35(3):413-37. doi:10.1016/j.iac.2015.04.003 177. Pecl J, Karaskova E, Kunovsky L, et al. Eosinophilic esophagitis - 10 years of experience in five Czech pediatric endoscopy centers. Gastroent Hepatol. 2020;74(6):469- 480. doi:10.48095/ccgh2020469 178. Castellaneta SP, Afzal NA, Greenberg M, et al. Diagnostic role of upper gastrointestinal endoscopy in pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2004;39(3):257-61. doi:10.1097/00005176-200409000-00006 179. Abuquteish D, Putra J. Upper gastrointestinal tract involvement of pediatric inflammatory bowel disease: A pathological review. World J Gastroenterol. 2019;25(16):1928-1935. doi:10.3748/wjg.v25.i16.1928 180. Galbraith JC, Shafran SD. Herpes simplex esophagitis in the immunocompetent patient: report of four cases and review. Clin Infect Dis. 1992;14(4):894-901. doi:10.1093/clinids/14.4.894 181. Jabandziev P, Jouza M, Pecl J, et al. Herpetic esophagitis in a 7-year-old immunocompetent patient. Gastroent Hepatol. 2020;74(3):233-237. doi:10.14735/amgh2020233 182. Phadke VK, Friedman-Moraco RJ, Quigley BC, Farris AB, Norvell JP. Concomitant herpes simplex virus colitis and hepatitis in a man with ulcerative colitis: Case report and review of the literature. Medicine (Baltimore). 2016;95(42):e5082. doi:10.1097/MD.0000000000005082 183. Breton J, Kastl A, Conrad MA, Baldassano RN. Positioning Biologic Therapies in the Management of Pediatric Inflammatory Bowel Disease. Gastroenterol Hepatol (N Y). 2020;16(8):400-414. 73 184. Atreya R, Neurath MF, Siegmund B. Personalizing Treatment in IBD: Hype or Reality in 2020? Can We Predict Response to Anti-TNF? Front Med (Lausanne). 2020;7:517. doi:10.3389/fmed.2020.00517 185. Claytor JD, El-Nachef N. Fecal microbial transplant for inflammatory bowel disease. Curr Opin Clin Nutr Metab Care. 2020;23(5):355-360. doi:10.1097/MCO.0000000000000676 186. Selset Aandahl G, Jacobsen CD, Rode L, Rathe Oedegaard E. Preliminary experience with plasma exchange in patients with ulcerative colitis. Transfus Sci. 2000;22(3):155-60. doi:10.1016/s0955-3886(00)00039-4 187. Ciccocioppo R, Baumgart DC, Dos Santos CC, Galipeau J, Klersy C, Orlando G. Perspectives of the International Society for Cell & Gene Therapy Gastrointestinal Scientific Committee on the Intravenous Use of Mesenchymal Stromal Cells in Inflammatory Bowel Disease (PeMeGi). Cytotherapy. 2019;21(8):824-839. doi:10.1016/j.jcyt.2019.05.003 188. Langhorst J, Wulfert H, Lauche R, et al. Systematic review of complementary and alternative medicine treatments in inflammatory bowel diseases. J Crohns Colitis. 2015;9(1):86-106. doi:10.1093/ecco-jcc/jju007 189. Antonioli L, Fornai M, Romano B, Pellegrini C, Blandizzi C. Editorial: IBD Management-Novel Targets and Therapeutic Perspectives. Front Pharmacol. 2020;11:448. doi:10.3389/fphar.2020.00448 190. Ungaro RC, Aggarwal S, Topaloglu O, Lee WJ, Clark R, Colombel JF. Systematic review and meta-analysis: efficacy and safety of early biologic treatment in adult and paediatric patients with Crohn's disease. Aliment Pharmacol Ther. 2020;51(9):831-842. doi:10.1111/apt.15685 191. Na SY, Moon W. Perspectives on Current and Novel Treatments for Inflammatory Bowel Disease. Gut Liver. 2019;13(6):604-616. doi:10.5009/gnl19019 192. Olbjørn C, Rove JB, Jahnsen J. Combination of Biological Agents in Moderate to Severe Pediatric Inflammatory Bowel Disease: A Case Series and Review of the Literature. Paediatr Drugs. 2020;22(4):409-416. doi:10.1007/s40272-020-00396-1 193. Verburgt CM, Ghiboub M, Benninga MA, de Jonge WJ, Van Limbergen JE. Nutritional Therapy Strategies in Pediatric Crohn's Disease. Nutrients. 2021;13(1)doi:10.3390/nu13010212 194. Levine A, Wine E, Assa A, et al. Crohn's Disease Exclusion Diet Plus Partial Enteral Nutrition Induces Sustained Remission in a Randomized Controlled Trial. Gastroenterology. 2019;157(2):440-450.e8. doi:10.1053/j.gastro.2019.04.021 74 195. Sigall Boneh R, Sarbagili Shabat C, Yanai H, et al. Dietary Therapy With the Crohn's Disease Exclusion Diet is a Successful Strategy for Induction of Remission in Children and Adults Failing Biological Therapy. J Crohns Colitis. 2017;11(10):1205-1212. doi:10.1093/ecco-jcc/jjx071 196. Zhan YL, Zhan YA, Dai SX. Is a low FODMAP diet beneficial for patients with inflammatory bowel disease? A meta-analysis and systematic review. Clin Nutr. 2018;37(1):123-129. doi:10.1016/j.clnu.2017.05.019 197. Vrdoljak J, Vilović M, Živković PM, et al. Mediterranean Diet Adherence and Dietary Attitudes in Patients with Inflammatory Bowel Disease. Nutrients. 2020;12(11)doi:10.3390/nu12113429 198. Strisciuglio C, Cenni S, Serra MR, et al. Effectiveness of Mediterranean Diet's Adherence in children with Inflammatory Bowel Diseases. Nutrients. 2020;12(10)doi:10.3390/nu12103206 199. Cucinotta U, Romano C, Dipasquale V. Diet and Nutrition in Pediatric Inflammatory Bowel Diseases. Nutrients. 2021;13(2)doi:10.3390/nu13020655 200. Bessissow T, Reinglas J, Aruljothy A, Lakatos PL, Van Assche G. Endoscopic management of Crohn's strictures. World J Gastroenterol. 2018;24(17):1859-1867. doi:10.3748/wjg.v24.i17.1859 201. Kelay A, Tullie L, Stanton M. Surgery and paediatric inflammatory bowel disease. Transl Pediatr. 2019;8(5):436-448. doi:10.21037/tp.2019.09.01 202. Amil-Dias J, Kolacek S, Turner D, et al. Surgical Management of Crohn Disease in Children: Guidelines From the Paediatric IBD Porto Group of ESPGHAN. J Pediatr Gastroenterol Nutr. 2017;64(5):818-835. doi:10.1097/MPG.0000000000001562 203. Forsdick VK, Tan Tanny SP, King SK. Medical and surgical management of pediatric perianal crohn's disease: A systematic review. J Pediatr Surg. 2019;54(12):2554-2558. doi:10.1016/j.jpedsurg.2019.08.036 204. Mutanen A, Pakarinen MP. Perianal Crohn's Disease in Children and Adolescents. Eur J Pediatr Surg. 2020;30(5):395-400. doi:10.1055/s-0040-1716724 205. Meima-van Praag EM, Buskens CJ, Hompes R, Bemelman WA. Surgical management of Crohn's disease: a state of the art review. Int J Colorectal Dis. 2021;36(6):1133-1145. doi:10.1007/s00384-021-03857-2 206. Ashton JJ, Borca F, Mossotto E, et al. Increased prevalence of anti-TNF therapy in paediatric inflammatory bowel disease is associated with a decline in surgical resections during childhood. Aliment Pharmacol Ther. 2019;49(4):398-407. doi:10.1111/apt.15094 75 207. Poredska K, Kunovsky L, Marek F, et al. The Influence of Microscopic Inflammation at Resection Margins on Early Postoperative Endoscopic Recurrence After Ileocaecal Resection for Crohn's Disease. J Crohns Colitis. 2020;14(3):361-368. doi:10.1093/ecco- jcc/jjz153 208. Marteau P, Laharie D, Colombel JF, et al. Interobserver Variation Study of the Rutgeerts Score to Assess Endoscopic Recurrence after Surgery for Crohn's Disease. J Crohns Colitis. 2016;10(9):1001-5. doi:10.1093/ecco-jcc/jjw082 209. Tandon P, Malhi G, Abdali D, et al. Active Margins, Plexitis, and Granulomas Increase Postoperative Crohn's Recurrence: Systematic Review and Meta-analysis. Clin Gastroenterol Hepatol. 2021;19(3):451-462. doi:10.1016/j.cgh.2020.08.014 210. Kim S. Surgery in Pediatric Crohn's Disease: Indications, Timing and Post-Operative Management. Pediatr Gastroenterol Hepatol Nutr. 2017;20(1):14-21. doi:10.5223/pghn.2017.20.1.14 211. Adler J, Dong S, Eder SJ, Dombkowski KJ, System IPILH. Perianal Crohn Disease in a Large Multicenter Pediatric Collaborative. J Pediatr Gastroenterol Nutr. 2017;64(5):e117e124. doi:10.1097/MPG.0000000000001447 212. Rowe W. Complications of Inflammatory Bowel Disease. https://emedicine.medscape.com/article/1918545-overview#a1 213. Adler J, Jary HK, Eder SJ, et al. Identifying perianal fistula complications in pediatric patients with Crohn's disease using administrative claims. PLoS One. 2019;14(8):e0219893. doi:10.1371/journal.pone.0219893 214. Lee T, Kamm MA, Bell S, et al. Long-term outcomes of perianal fistulizing Crohn's disease in the biologic era. JGH Open. 2021;5(2):235-241. doi:10.1002/jgh3.12475 215. Benchimol EI, Turner D, Mann EH, et al. Toxic megacolon in children with inflammatory bowel disease: clinical and radiographic characteristics. Am J Gastroenterol. 2008;103(6):1524-31. doi:10.1111/j.1572-0241.2008.01807.x 216. Desai J, Elnaggar M, Hanfy AA, Doshi R. Toxic Megacolon: Background, Pathophysiology, Management Challenges and Solutions. Clin Exp Gastroenterol. 2020;13:203-210. doi:10.2147/CEG.S200760 217. Ricciuto A, Hansen BE, Ngo B, et al. Primary Sclerosing Cholangitis in Children With Inflammatory Bowel Diseases Is Associated With Milder Clinical Activity But More Frequent Subclinical Inflammation and Growth Impairment. Clin Gastroenterol Hepatol. 2020;18(7):1509-1517.e7. doi:10.1016/j.cgh.2019.08.048 76 218. Lazzerini M, Bramuzzo M, Ventura A. Association between orofacial granulomatosis and Crohn's disease in children: systematic review. World J Gastroenterol. 2014;20(23):7497- 504. doi:10.3748/wjg.v20.i23.7497 219. Gavioli CFB, Florezi GP, Dabronzo MLD, Jiménez MR, Nico MMS, Lourenço SV. Orofacial Granulomatosis and Crohn Disease: Coincidence or Pattern? A Systematic Review. Dermatology. 2021:1-6. doi:10.1159/000513446 220. Kucharska M, Daniluk U, Kwiatek-Średzińska KA, et al. Hepatobiliary manifestations of inflammatory bowel disease in children. Clin Exp Hepatol. 2019;5(3):203-209. doi:10.5114/ceh.2019.87632 221. Olén O, Askling J, Sachs MC, et al. Childhood onset inflammatory bowel disease and risk of cancer: a Swedish nationwide cohort study 1964-2014. BMJ. 2017;358:j3951. doi:10.1136/bmj.j3951 222. Ishige T. Growth failure in pediatric onset inflammatory bowel disease: mechanisms, epidemiology, and management. Transl Pediatr. 2019;8(1):16-22. doi:10.21037/tp.2018.12.04 223. Pappa H, Thayu M, Sylvester F, Leonard M, Zemel B, Gordon C. Skeletal health of children and adolescents with inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2011;53(1):11-25. doi:10.1097/MPG.0b013e31821988a3 224. Yang HR. Updates on bone health in children with gastrointestinal diseases. Ann Pediatr Endocrinol Metab. Mar 2020;25(1):10-14. doi:10.6065/apem.2020.25.1.10 225. Qualter P, Rouncefield-Swales A, Bray L, et al. Depression, anxiety, and loneliness among adolescents and young adults with IBD in the UK: the role of disease severity, age of onset, and embarrassment of the condition. Qual Life Res. 2021;30(2):497-506. doi:10.1007/s11136-020-02653-9 226. Ossum AM, Palm Ø, Lunder AK, 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(1):96-104. doi:10.1093/ecco-jcc/jjx126 227. Diaconescu S, Strat S, Balan GG, et al. Dermatological Manifestations in Pediatric Inflammatory Bowel Disease. Medicina (Kaunas). 2020;56(9)doi:10.3390/medicina56090425 228. Ottaviano G, Salvatore S, Salvatoni A, Martelossi S, Ventura A, Naviglio S. Ocular Manifestations of Paediatric Inflammatory Bowel Disease: A Systematic Review and Metaanalysis. J Crohns Colitis. 2018;12(7):870-879. doi:10.1093/ecco-jcc/jjy029 77 229. Bianchi L, Gaiani F, Bizzarri B, et al. Renal lithiasis and inflammatory bowel diseases, an update on pediatric population. Acta Biomed. 2018;89(9-S):76-80. doi:10.23750/abm.v89i9-S.7908 230. Goyal A, Zheng Y, Albenberg LG, et al. Anemia in Children With Inflammatory Bowel Disease: A Position Paper by the IBD Committee of the North American Society of Pediatric Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol Nutr. 2020;71(4):563-582. doi:10.1097/MPG.0000000000002885 231. McKie K, McLoughlin RJ, Hirsh MP, Cleary MA, Aidlen JT. Risk Factors for Venous Thromboembolism in Children and Young Adults With Inflammatory Bowel Disease. J Surg Res. 2019;243:173-179. doi:10.1016/j.jss.2019.04.087 232. Alvisi P, Dipasquale V, Barabino A, et al. Infections and malignancies risks related to TNF-α-blocking agents in pediatric inflammatory bowel diseases. Expert Rev Gastroenterol Hepatol. 2019;13(10):957-961. doi:10.1080/17474124.2019.1663173 233. Dipasquale V, Romano C. Pharmacological treatments and infectious diseases in pediatric inflammatory bowel disease. Expert Rev Gastroenterol Hepatol. 2018;12(3):237- 247. doi:10.1080/17474124.2018.1391091 234. Cavalcante SS, Mota E, Silva LR, Teixeira LF, Cavalcante LB. Risk factors for developing nosocomial infections among pediatric patients. Pediatr Infect Dis J. 2006;25(5):438-45. doi:10.1097/01.inf.0000217377.54597.92 235. Holmer A, Singh S. Overall and comparative safety of biologic and immunosuppressive therapy in inflammatory bowel diseases. Expert Rev Clin Immunol. 2019;15(9):969-979. doi:10.1080/1744666X.2019.1646127 236. Rodríguez-Acelas AL, de Abreu Almeida M, Engelman B, Cañon-Montañez W. Risk factors for health care-associated infection in hospitalized adults: Systematic review and meta-analysis. Am J Infect Control. 2017;45(12):e149-e156. doi:10.1016/j.ajic.2017.08.016 237. Singh S, Facciorusso A, Dulai PS, Jairath V, Sandborn WJ. Comparative Risk of Serious Infections With Biologic and/or Immunosuppressive Therapy in Patients With Inflammatory Bowel Diseases: A Systematic Review and Meta-Analysis. Clin Gastroenterol Hepatol. 2020;18(1):69-81.e3. doi:10.1016/j.cgh.2019.02.044 238. Chicco D, Jurman G. Survival prediction of patients with sepsis from age, sex, and septic episode number alone. Sci Rep. 2020;10(1):17156. doi:10.1038/s41598-020-73558-3 239. Font MD, Thyagarajan B, Khanna AK. Sepsis and Septic Shock - Basics of diagnosis, pathophysiology and clinical decision making. Med Clin North Am. 2020;104(4):573-585. doi:10.1016/j.mcna.2020.02.011 78 240. Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016;315(8):801-10. doi:10.1001/jama.2016.0287 241. Cruz AT, Lane RD, Balamuth F, et al. Updates on pediatric sepsis. J Am Coll Emerg Physicians Open. 2020;1(5):981-993. doi:10.1002/emp2.12173 242. Fleischmann-Struzek C, Mellhammar L, Rose N, et al. Incidence and mortality of hospital- and ICU-treated sepsis: results from an updated and expanded systematic review and meta-analysis. Intensive Care Med. 2020;46(8):1552-1562. doi:10.1007/s00134-020-06151-x 243. Lu H, Wen D, Wang X, et al. Host genetic variants in sepsis risk: a field synopsis and meta-analysis. Crit Care. 2019;23(1):26. doi:10.1186/s13054-019-2313-0 244. Jabandziev P, Smerek M, Michalek J, Fedora M, Kosinova L, Hubacek JA. Multiple gene-to-gene interactions in children with sepsis: a combination of five gene variants predicts outcome of life-threatening sepsis. Crit Care. 2014;18(1):R1. doi:10.1186/cc13174 245. Satoh K, Okuyama M, Furuya T, Irie Y, Nakae H. Severe Sepsis Caused by Bacteria That Entered via the Intestinal Tract: A Case of Crohn's Disease in a Child. Cureus. 2020;12(8):e9822. doi:10.7759/cureus.9822 246. Barash Y, Klang E, Tau N, et al. Evolution of Inflammatory Bowel Disease Research From a Bird's-Eye Perspective: A Text-Mining Analysis of Publication Trends and Topics. Inflamm Bowel Dis. 2021;27(3):434-439. doi:10.1093/ibd/izaa091 247. Fiocchi C. Inflammatory Bowel Disease: Complexity and Variability Need Integration. Front Med (Lausanne). 2018;5:75. doi:10.3389/fmed.2018.00075 248. Dhyani M, Joshi N, Bemelman WA, et al. Challenges in IBD Research: Novel Technologies. Inflamm Bowel Dis. 02019;25(Suppl 2):S24-S30. doi:10.1093/ibd/izz077 249. Kumar M, Garand M, Al Khodor S. Integrating omics for a better understanding of Inflammatory Bowel Disease: a step towards personalized medicine. J Transl Med. 2019;17(1):419. doi:10.1186/s12967-019-02174-1 250. de Souza HSP. Etiopathogenesis of inflammatory bowel disease: today and tomorrow. Curr Opin Gastroenterol. 2017;33(4):222-229. doi:10.1097/MOG.0000000000000364 251. Flamant M, Roblin X. Inflammatory bowel disease: towards a personalized medicine. Therap Adv Gastroenterol. 2018;11:1756283X17745029. doi:10.1177/1756283X17745029 252. Denson LA, Curran M, McGovern DPB, et al. Challenges in IBD Research: Precision Medicine. Inflamm Bowel Dis. 2019;25(Suppl 2):S31-S39. doi:10.1093/ibd/izz078 79 253. D'Amico F, Rahier JF, Leone S, Peyrin-Biroulet L, Danese S. Views of patients with inflammatory bowel disease on the COVID-19 pandemic: a global survey. Lancet Gastroenterol Hepatol. 2020;5(7):631-632. doi:10.1016/S2468-1253(20)30151-5 254. Ben-Or O, Zelnik N, Shaoul R, Pacht A, Lerner A. The neurologic profile of children and adolescents with inflammatory bowel disease. J Child Neurol. 2015;30(5):551-7. doi:10.1177/0883073814521296 255. Szigethy EM, Youk AO, Benhayon D, et al. Depression subtypes in pediatric inflammatory bowel disease. J Pediatr Gastroenterol Nutr. 2014;58(5):574-81. doi:10.1097/MPG.0000000000000262 256. Morís G. Inflammatory bowel disease: an increased risk factor for neurologic complications. World J Gastroenterol. 2014;20(5):1228-37. doi:10.3748/wjg.v20.i5.1228 257. García-Alanís M, Quiroz-Casian L, Castañeda-González H, et al. Prevalence of mental disorder and impact on quality of life in inflammatory bowel disease. Gastroenterol Hepatol. 2021;44(3):206-213. doi:10.1016/j.gastrohep.2020.06.025 258. Zhang B, Wang HE, Bai YM, et al. Inflammatory bowel disease is associated with higher dementia risk: a nationwide longitudinal study. Gut. 2021;70(1):85-91. doi:10.1136/gutjnl-2020-320789 259. Bhamre R, Sawrav S, Adarkar S, Sakaria R, J Bhatia S. Psychiatric comorbidities in patients with inflammatory bowel disease. Indian J Gastroenterol. 2018;37(4):307-312. doi:10.1007/s12664-018-0870-9 260. Gassler N. Paneth cells in intestinal physiology and pathophysiology. World J Gastrointest Pathophysiol. 2017;8(4):150-160. doi:10.4291/wjgp.v8.i4.150 261. Wehkamp J, Stange EF. An Update Review on the Paneth Cell as Key to Ileal Crohn's Disease. Front Immunol. 2020;11:646. doi:10.3389/fimmu.2020.00646 262. Valeri M, Raffatellu M. Cytokines IL-17 and IL-22 in the host response to infection. Pathog Dis. 2016;74(9)doi:10.1093/femspd/ftw111 80 List of Annexes (Selected publications arranged in order of appearance in the text) Annex 1 Jabandziev P, Hlavackova E, Bily V, Karaskova E, Kunovsky L, Ravcukova B, Grombirikova H, Kozumplikova R, Buckova H, Slaba K, Pinkasova T, Jouza M, Pecl J, Jezova, M, Curtisova V, Freiberger T. Trichohepatoenteric syndrome in a patient with TTC37 mutations - case report. Gastroent Hepatol. 2020; 74 (6), 481–487. Annex 2 Jabandziev P, Pinkasova T, Kunovsky L, Papez J, Jouza M, Karlinova B, Novackova M, Urik M, Aulicka S, Slaby O, Bohosova, J, Bajerova K, Bajer M, Goel A. Regional Incidence of Inflammatory Bowel Disease in a Czech Pediatric Population: 16 Years of Experience (2002–2017). J Pediatr Gastroenterol Nutr. 2020; 70 (5), 586–592. Annex 3 Jabandziev P, Bohosova J, Pinkasova, T, Kunovsky L, Slaby O, Goel A. The Emerging Role of Noncoding RNAs in Pediatric Inflammatory Bowel Disease. Inflamm Bowel Dis. 2020; 26 (7), 985–993. Annex 4 Jabandziev P, Kakisaka T, Bohosova J, Pinkasova T, Kunovsky L, Slaby O, Goel A. MicroRNAs in Colon Tissue of Pediatric Ulcerative Pancolitis Patients Allow Detection and Prognostic Stratification. J Clin Med. 2021; 10 (6), 1325. Annex 5 Slaba K, Noskova H, Vesela P, Tuckova J, Jicinska H, Honzik T, Hansikova H, Kleiblova P, Stourac P, Jabandziev P, Slaby O, Prochazkova D. Novel Splicing Variant in the. Front Genet. 2020; 11, 561054. 81 Annex 6 Formankova R, Kanderova V, Rackova M, Svaton M, Brdicka T, Riha P, Keslova P, Mejstrikova E, Zaliova M, Freiberger T, Grombirikova H, Zemanova Z, Vlkova M, Fencl F, Copova I, Bronsky J, Jabandziev P, Sedlacek P, Soukalova J, Zapletal O, Stary J, Trka J, Kalina T, Skvarova Kramarzova K, Hlavackova E, Litzman J, Fronkova E. Novel SAMD9 Mutation in a Patient with Immunodeficiency, Neutropenia, Impaired Anti-CMV Response, and Severe Gastrointestinal Involvement. Front Immunol. 2019; 10:2194. Annex 7 Pecl J, Karaskova E, Kunovsky L, Jimramovsky F, Schneiderova H, Pinkasova, T, Veverkova M, Jouza M, Hlouskova E, Bajerova K, Latalova V, Veghova-Velganova M, Geryk M, Sulakova A, Toukalkova L, Jaksic D, Zimen M, Jezova M, Urik M, Wiesnerova M, Jabandziev P., Eosinophilic esophagitis - 10 years of experience in five Czech pediatric endoscopy centers. Gastroent Hepatol. 2020; 74 (6), 469–480. Annex 8 Jabandziev P, Jouza M, Pecl J, Urik M, Papez J, Pinkasova T, Slaba K, Trna J, Kyclova J, Vaculova J, Kunovsky L. Herpetic esophagitis in a 7-year-old immunocompetent patient. Gastroent Hepatol. 2020; 74 (3), 233-237. Annex 9 Poredska K, Kunovsky L, Marek F, Kala Z, Prochazka V, Dolina J, Zboril V, Kovalcikova P, Pavlik T, Jabandziev P, Pavlovsky Z, Vlazny J, Mitas L. The Influence of Microscopic Inflammation at Resection Margins on Early Postoperative Endoscopic Recurrence After Ileocaecal Resection for Crohn’s Disease. J Crohns Colitis. 2020; 14 (3), 361–368. Annex 10 Jabandziev P, Smerek M, Michalek J, Fedora M, Kosinova L, Hubacek JA. Multiple geneto-gene interactions in children with sepsis: a combination of five gene variants predicts outcome of life-threatening sepsis. Crit Care. 2014; 18 (1), R1. 82 Annex 1 83 84 85 86 87 88 89 Annex 2 90 91 92 93 94 95 96 Annex 3 97 98 99 100 101 102 103 104 105 Annex 4 106 107 108 109 110 111 112 113 114 115 116 Annex 5 117 118 119 120 121 122 123 Annex 6 124 125 126 127 128 129 130 131 Annex 7 132 133 134 135 136 137 138 139 140 141 142 143 Annex 8 144 145 146 147 148 Annex 9 149 150 151 152 153 154 155 156 Annex 10 157 158 159 160 161 162 163 164