Next Article in Journal
Association between Dietary Vitamin E Intake and Human Papillomavirus Infection among US Adults: A Cross-Sectional Study from National Health and Nutrition Examination Survey
Next Article in Special Issue
Nutritional Biomarkers for the Prediction of Response to Anti-TNF-α Therapy in Crohn’s Disease: New Tools for New Approaches
Previous Article in Journal
Maternal Obesity and Kawasaki Disease-like Vasculitis: A New Perspective on Cardiovascular Injury and Inflammatory Response in Offspring Male Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nutrition, Nutritional Status, Micronutrients Deficiency, and Disease Course of Inflammatory Bowel Disease

1
Gastroenterology Unit, Division of Gastroenterology, Hepatology, and Nutrition, Department of Life, Health and Environmental Sciences, University of L’Aquila, Piazzale Salvatore Tommasi 1, 67100 L’Aquila, Italy
2
Division of Gastroenterology, Galliera Hospital, 16128 Genoa, Italy
3
Diagnostic and Surgical Endoscopy Unit, San Salvatore Academic Hospital, 67100 L’Aquila, Italy
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(17), 3824; https://doi.org/10.3390/nu15173824
Submission received: 10 August 2023 / Revised: 25 August 2023 / Accepted: 28 August 2023 / Published: 31 August 2023

Abstract

:
During the disease course, most Inflammatory Bowel Disease patients present a condition of malnutrition, undernutrition, or even overnutrition. These conditions are mainly due to suboptimal nutritional intake, alterations in nutrient requirements and metabolism, malabsorption, and excessive gastrointestinal losses. A suboptimal nutritional status and low micronutrient serum levels can have a negative impact on both induction and maintenance of remission and on the quality of life of Inflammatory Bowel Disease patients. We performed a systematic review including all the studies evaluating the connection between nutrition, nutrition status (including undernutrition and overnutrition), micronutrient deficiency, and both disease course and therapeutic response in Inflammatory Bowel Disease patients. This systematic review was performed using PubMed/MEDLINE and Scopus. Four main clinical settings concerning the effect of nutrition on disease course in adult Inflammatory Bowel Disease patients were analyzed (induction of remission, maintenance of remission, risk of surgery, post-operative recurrence, and surgery-related complications). Four authors independently reviewed abstracts and manuscripts for eligibility. 6077 articles were found; 762 duplicated studies were removed. Out of 412 full texts analyzed, 227 were included in the review. The evidence summarized in this review showed that many nutritional aspects could be potential targets to induce a better control of symptoms, a deeper remission, and overall improve the quality of life of Inflammatory Bowel Disease patients.

1. Introduction

Inflammatory bowel diseases (IBD) are chronic inflammatory diseases, mediated by the immune system, which affect the gastrointestinal tract. The two main manifestations are Crohn’s Disease (CD) and ulcerative colitis (UC). CD may affect any area of the gastrointestinal tract and has a transmural involvement. UC generally occurs only in the rectum and colon and involves the mucosa and submucosa layers [1,2].
The etiology of IBD is not completely defined. Yet, several studies support the hypothesis that their onset is due to a combination and interplay of genetic factors, immune dysregulation and environmental triggers that can modify gut microbiome [3,4,5]. In this scenario, diet is a potential environmental trigger. The global increasing incidence of IBD seems to be associated with Western lifestyle and diet: a high intake of proteins and red meat can result in an increased production of bacterial metabolites, such as an increase of ammonia, indoles, phenols, and sulphides, and a decrease of short-chain fatty acids (SCFAs), which could all be involved in the development of IBD [6,7].
As a consequence, diet modifications have been considered therapeutic tools; for example, in pediatric IBD patients, enteral nutrition has been shown to be effective in inducing clinical remission, independently of the used formula [8,9,10].
Malnutrition, undernutrition and overnutrition seen in such patients are variable during the disease course [11] and due to suboptimal nutritional intake, alterations in nutrient requirements and metabolism, malabsorption, excessive gastrointestinal losses, and medication [12].
At the time of diagnosis, 60% of CD patients and 35% of UC patients are underweight, even if this proportion is lowered in the last years, reflecting the increased incidence of obesity [13,14]: 20–40% of adult patients with IBD are overweight (25 < body mass index (BMI) < 30 kg/m2), and an additional 15–40% are obese (BMI > 30 kg/m2) [15].
Obesity is associated with treatment failure (especially with anti-TNF drugs), risk of hospitalization, and lower endoscopic remission rates [16,17,18,19]; sarcopenia in overweight IBD patients (BMI ≥ 25 kg/m2) is the only significant predictor of the need for surgery (p = 0.002) [20]; nutritional deficits and low micronutrients serum levels can have a negative impact on both induction and maintenance of remission and on the quality of life of these patients [21,22]. Thus, the assessment of nutritional status in IBD patients is a crucial issue and current guidelines suggest that patients with IBD should be regularly screened for nutritional status, micronutrient deficiencies and bone mineral density [21,23].
Currently, there are limited data on the disease course and therapy response in cases of malnutrition in IBD, especially in the context of sarcopenia and undernutrition. Moreover, if micronutrient or vitamin supplementation (e.g., vitamin D supplementation) could be a potential therapeutic option or only an effect of disease activity it is still unclear [24,25].
The aim of this systematic review is trying to clarify the connection between nutrition, malnutrition (including overnutrition and undernutrition), micronutrient deficiency, and both disease course and therapeutic response in IBD patients.

2. Materials and Methods

This systematic review was performed using PubMed/MEDLINE and Scopus. For each of the relevant publications (previous review articles and included studies), reference sections were also screened for other applicable publications.
The research strategy for each clinical question is reported in the Supplementary Material (Table S1). We found 6077 articles; 762 duplicated studies were removed. Out of 412 full texts analyzed, 227 were included in the review (Figure S1).
No filters were used in the search strategy. The data of the last search was May 2023. The complete selection process is reported in the Supplementary Material (Table S2).
Four authors did a systematic literature search. Clinical questions and related outcomes of interest were identified using the PICO framework. Four main clinical settings concerning adult IBD patients were identified.
-
Induction of remission
-
Maintenance of remission
-
Risk of surgery
-
Postoperative recurrence (POR) and surgery-related complications

2.1. Selection of Studies and Data Extraction

Four authors (GS, SF, SM, and MM) independently reviewed abstracts and manuscripts for eligibility.
Conflicts were resolved by consensus, referring to the original articles. The selection was made according to the following criteria:

2.2. Inclusion CriteriaPatient Type: Adult Patients (age ≥ 18) with a Confirmed Diagnosis of IBD

-
Intervention: Nutritional management; Nutritional evaluation; serum evaluation or supplementation of micronutrients or albumin.
-
Outcome: evaluation of clinical relapse or disease activity (evaluated with disease activity score or loss of response to therapy); risk of surgery; POR and surgery-related complications
-
Study type: Meta-analysis, Randomized clinical trial (RCT), Non-randomized study of intervention (NRSI), cross-sectional study.

2.3. Exclusion Criteria

-
Paediatric patients
-
Non-human study
-
Lack of data concerning clinical response, risk of surgery, POR, and surgery-related complications.
Four reviewers (GS, SF, SM, and MM) independently reviewed the literature according to the above-predefined strategy and criteria and selected eligible studies; any disagreement was resolved by consensus or by recourse to a fifth author (MV).
Each reviewer extracted the data of interest in a pre-made template: title and reference details (first author, journal, year, country), study population characteristics (number of patients, gender, age, disease type (UC or CD), intervention details and outcome data (induction of remission, maintenance of remission, risk of surgery, POR, and surgery-related complications).
All data were recorded independently by the literature reviewers in separate databases and will be compared at the end of the reviewing process to limit selection bias. The database was also reviewed by another author (MV). Any disagreement was resolved by consensus or by recourse to the senior author.

3. Nutrition and Nutritional Status

If food is implicated in the pathogenesis of IBD is still not clear, but the impact of some nutrients on the behalf of the gastrointestinal tract has been suggested. For example, dietary fibre escape digestion in the small bowel and enter the colon where they are metabolized by gut bacteria which produce SCFAs, energy sources for colonocytes [26]. On the contrary red meat and other high-protein foods, contribute to sulphides formation which damages the mucus in the colon [27,28]. In the IBD population, 70% of patients report food-related symptom exacerbation, while a wide variety of foods are believed to be helpful [29]. This becomes a concern when patients drastically reduce or completely avoid important nutrients such as folic acid, calcium, vitamin B 12, and iron which represent the most frequent nutritional deficiency in IBD patients. This attitude, summed to disease duration, extent and severity, may put them at risk of developing nutritional deficiencies in the long term [30].

3.1. Nutrition and Exclusion Diet

Compared to recommended requirements, adults with IBD have an inadequate intake of energy, fibres, fat-soluble vitamins, folate and calcium [31]. Nutritional support for correcting deficiencies can be provided summarily in the form of parenteral nutrition (PN), enteral nutrition (EN) and specific diets.

3.1.1. Induction of Remission

Literature evidence seems to agree that steroids are more effective than EN in inducing remission in CD; anyway, both PN and EN used in combination with steroids, can improve the response rate to intravenous corticosteroid therapy [32,33,34,35,36,37]. Regarding UC, there is no evidence that EN alters the inflammatory response: therefore, it is not used in the treatment of active disease nor in the maintenance of remission in UC [38] (Table 1).
Oral diets proposed for active UC have been analyzed in the recent meta-analysis from Limketkai: symptom-guided exclusion diet, milk protein elimination, and gluten-free diet were considered. No one seems to be helpful for the induction of remission [48]. Recently, a UC Exclusion Diet (UCED), already proposed for a paediatric population and composed of limited animal fats and sulphated amino acids consumption, has been applied in a blinded, randomized controlled (CRAFT UC) including adults patients with active UC refractory to therapy; patients receiving UCED alone achieved higher remission and mucosal healing rates compared to those undergoing to faecal transplantation, with or without dietary modification [45,46].
Similarly, an open-label, pilot randomised trial shows that the CD Exclusion Diet (CDED), with or without partial EN, is effective for induction and maintenance of remission in biologic naive adults with mild-to-moderate CD [47]. Also, low refined carbohydrate diet and symptoms-guided diet outperformed controls for induction of remission in CD, even if with very low certainty of evidence [48].
Regarding the habit of reducing fibres in the acute phase, a prospective randomized controlled study on active CD concluded that consuming a low residue diet gave no advantage in terms of symptoms, hospitalization, surgery and nutritional status compared to a free Italian-style diet—rich in fibres [41]. Similar conclusions were more recently achieved in the DINE-CD trial on 191 CD patients with mild to moderate symptoms: leaving a Mediterranean diet in favor of the so-called Specific Carbohydrate Diet (SCD) eliminating all grains, sugars, milk products and most processed foods, gives no advantage in terms of clinical and biochemical remission [49].

3.1.2. Maintenance of Remission

Two meta-analyses focused on the use of EN in CD patients in clinical remission, both regarding partial EN, offering the possibility to add an oral daily diet. The type of enteral formulation (elemental, semi-elemental or polymeric) does not make a difference in terms of efficacy [33,34] (Table 1). The first, dated 2015, assessed that the combination of infliximab therapy plus a specialized EN is more effective in achieving and maintaining clinical remission compared with infliximab monotherapy. However, only 4 studies were included, all Japanese, the majority were retrospective and evaluating only the subjective clinical outcome [39]. More convincing evidence comes from another meta-analysis on 429 CD patients in remission and on maintaining therapy; 224 received EN and 205 received non-EN treatment as a control treatment. Patients receiving EN exhibited a higher frequency of clinical remission maintenance at 0.5–1 year and a significantly lower rate of clinical relapse at 0.5–2 years [40].
Fibre consumption seems to be beneficial also during remission. Two Japanese prospective, single-centre trials conclude that a semi-vegetarian diet, added to maintenance therapy, has a protective effect against relapse both in CD and UC [42,43]. In an open-label, parallel-group, multicentre, randomized clinical trial, UC patients in remission receiving only fermentable fibres, has the same relapse rate as those receiving mesalamine or mesalamine plus fibres [44].
It is not clear if reducing meat could be useful. While a prospective cohort study on 191 UC patients shows that a higher intake of meat may increase the risk of a relapse, the reduction of red meat as well as refined carbohydrates did not reduce the risk of relapse for CD [27,48]. The carrageenan-free diet, the anti-inflammatory diet and the milk protein elimination diet have no impact on the maintenance of remission in UC [48].

3.1.3. Risk of Surgery

A poor preoperative nutritional status is associated with an increased risk of postoperative complications [50]. If oral feeding is not sufficient, guidelines recommend that EN should be preferred over PN in malnourished patients and to postpone if possible surgery for 7–14 days to correct malnutrition; PN should be used as a supplement to EN if >60% of energy needs cannot be met via the enteral route or when an enteral administration is not possible (obstruction, anastomotic leak, fistula) [51].
Data arising from a meta-analysis suggest that CD patients receiving preoperative EN or PN are 74% less likely to have a postoperative complication compared to those receiving standard care without nutritional support; in particular, EN is significantly superior to standard of care without nutrition support in reducing post-operative complications, while PN has a trend toward being superior to standard [52]. However, other authors conducted systematic reviews on this topic and although all studies included presented encouraging results on nutritional support, both did not conduct a meta-analysis because of the heterogeneity of the studies, most classified as medium-low quality [53,54] (Table 2).

3.1.4. Post-Operative Recurrence and Surgery-Related Complications

Two post-surgical scenarios in which nutrition can be pivotal are pouchitis and short bowel syndrome, but quality studies are lacking. Regarding pouchitis, the literature reports several dietary interventions in small numbers of patients, and some of these studies were retrospective and without controls.
We only found a prospective study testing exclusive EN given for 28 days to 7 patients with chronic antibiotic-dependent pouchitis, resulting in a symptomatic improvement but no reduction in endoscopic or histological signs of inflammation [55].
Consumption of fibres has been considered in patients with a pouch, either as adjunctive fibre supplementation (e.g., inulin), or as a fibre-rich diet. A randomized, double-blind, placebo-control crossover study on 20 patients with J-pouch, revealed that dietary inulin supplementation leads to a reduction of endoscopic and histologic inflammation of pouch mucosa [56]. A prospective cross-sectional study compared the dietary intake of 80 pouch patients to that of 80 healthy volunteer controls; within the study population, it compared those with a normal pouch to those with pouchitis. Pouch patients consumed significantly higher servings of fats and oils compared to healthy controls; patients with pouchitis consume fewer fruit servings and antioxidants than patients with a normal pouch [57]. Godny and colleagues suggest that consuming ≥1.5 fruit servings/day would be associated with a reduced risk of developing pouchitis in the following year [58].
Short bowel syndrome is a malabsorption syndrome consequence of multiple intestinal resections, whose severity and clinical presentation depending on the site and the extension of the resection, the health of the remaining mucosa and its ability to compensate. Intestinal failure resulting, can be severe, moderate or mild, requiring respectively PN, EN or oral supplements/dietary adjustments [60]. However, literature evidence in this field is limited, due to the extreme variety of presentations and the scarcity of IBD-specific works. ESPEN guidelines on chronic intestinal failure in adults recommend that patients with short bowel syndrome with a preserved colon consume a high-carbohydrate-low-fat diet; instead, the fat: carbohydrate ratio seems of less importance in patients without a colon, although the grade of evidence is low [59], being literature about this topic discordant [61,62]. A meta-analysis of clinical trials showed that the treatment with growth hormone and glutamine combined with a modified high-carbohydrate-low-fat diet was effective without any major adverse effects in patients with short bowel syndrome [63].

3.1.5. Main Evidence and Clinical Implications

-
PN and EN used in combination with steroids, can improve the response rate to intravenous corticosteroid therapy.
-
CDED is effective for induction and maintenance of remission in mild-to-moderate, biologic naive CD.
-
Only a low refined carbohydrate diet and a symptoms-guided diet outperformed controls for induction of remission in CD, even if with very low certainty of evidence.
-
In CD patients in maintenance therapy, adding EN offers better results in maintaining clinical remission. In CD and UC patients in maintenance therapy high fibre diets have a protective effect against relapse.
-
Preoperative EN or PN reduce the risk of postoperative complication in CD patients.
-
Exclusive EN in patients with chronic antibiotic dependent pouchitis can improve symptoms but not endoscopic or histological signs of inflammation; fibre intake can reduce endoscopic and histologic inflammation of pouch mucosa and reduce the risk of pouchitis.

3.2. Sarcopenia

Sarcopenia is defined by a loss in muscle mass and lean body mass that leads to functional changes and decreased strength [64].
The European Working Group on Sarcopenia in Older People (EWGSOP) developed 3 criteria for its definition and diagnosis: low muscle mass, low muscle strength, low physical performance (being necessary for the diagnosis the first criterion, plus one or both of the other two criteria) [65].
During disease flares, both the reduced caloric intake and the mucosa inflammation can impair nutrient absorption and determine weight loss [21].

3.2.1. Clinical Disease Course and Response to Therapies

Most of the studies comparing the course of CD in sarcopenic patients versus non-sarcopenic, showed a worse disease course in the first group, in terms of the need for corticosteroids, the occurrence of complications, fecal calprotectin (FC) value, and endoscopic recurrence [66,67,68,69,70,71,72,73]; in most of the studies such difference was statistically significant, while for just two of them a trend to a worse course emerged [74,75].

3.2.2. Risk of Surgery

In a meta-analysis published in 2017 including eight non-randomized studies, the authors showed that 46.2% (409/885) of IBD patients presented a condition of sarcopenia, with a higher prevalence in CD compared to UC (571 and 341 patients, respectively) [76]. This meta-analysis reported a non-significant difference between the sarcopenic and non-sarcopenic groups in terms of the need for surgery (unadjusted OR: 1.826; 95% CI 0.913–3.654; p = 0.089; I2 = 54.62%, p = 0.051) and post-operative complications (unadjusted OR: 3.265; 95% CI 0.575–18.557; p = 0.182; I2 = 88.46%, p < 0.001); however, pooled adjusted data for significant covariates among three included studies showed that sarcopenic group presented higher prevalence of surgery (adjusted OR: 2.665; 95% CI 1.121–6.336; p = 0.027; I2 = 33.94%, p = 0.220) and post-operative complications (adjusted OR = 6.097; 95% CI 1.756–21.175; p = 0.004; I2 = 0.0%, p = 0.637) [76]. Such a higher incidence of surgery in sarcopenic IBD patients is also reported by two observational studies, where higher odds of requiring surgery within 1 year were assessed also in those on biological therapy (OR, 2.36; 95% CI, 1.06–5.26, p = 0.04) [77,78].
Regarding CD, some authors [66,75,79,80,81] reported a higher incidence of surgery and shorter surgery-free survival in sarcopenic patients compared to non-sarcopenic, while others do not [73,82,83,84]. Bamba et al. concluded that sarcopenia is a risk factor for surgery in active CD patients, in univariate and multivariate analysis (OR 0.313; p = 0.006, OR 0.318; p = 0.015) [80].
Concerning UC patients, we found three retrospective studies including hospitalized patients with acute severe ulcerative colitis (ASUC). The need for rescue therapy and colectomy was higher for sarcopenic than non-sarcopenic patients (p < 0.001 and p = 0.02 for rescue therapy; p = 0.001 and p = 0.16 for colectomy) [85,86]. Moreover, sarcopenia results an independent risk factor for rescue therapy (OR 4.079, p < 0.001; OR 3.401, p = 0.03), urgent surgery (p = 0.001 and OR 2.999, 95% CI 1.285–6.997; p = 0.01) and post-operative complications (p = 0.003; OR 4.157, 95% CI 1.364–12.667, p = 0.012, respectively) in univariate and multivariate analysis [85,87].

3.2.3. Post-Operative Recurrence and Surgery-Related Complications

Other recent studies not included in the above mentioned meta-analysis reported a higher incidence of post-operative complications and a higher risk of post-surgery re-hospitalization in sarcopenic IBD patients [78,88,89,90], except one [91].
Considering CD, five studies showed a worse post-operative course (evaluated through various outcomes) in sarcopenic patients [72,84,92,93,94], while two studies did not confirm such differences [74,95].
Concerning UC, sarcopenia seems to be an independent risk factor for surgical site infections after ileal pouch-anal anastomosis (IPAA) in univariate and multivariate analysis (OR 5.85; p = 0.008 and OR 4.91; p = 0.03, respectively) [96].
In conclusion, sarcopenia seems to be a relevant predictor of a worse disease course. It is important to evaluate the presence of sarcopenia, especially in patients with severe disease, the elderly, and patients with risk-factors for surgery. In these groups, sarcopenia can increase the need for rescue therapy, the need surgery or urgent surgery, and even post-surgery complications.

3.2.4. Main Evidence and Clinical Implications

-
Sarcopenia in CD patients may result in a slight increase of worse clinical, biochemical, and endoscopic outcomes.
-
In IBD patients, in CD patients and ASUC setting sarcopenia results in a slight increase in the need for surgery, shorter surgery-free time or colectomy.
-
In IBD patients, in CD patients and after the IPAA setting, sarcopenia is a risk factor for post-operative complications.

3.3. Obesity

In the last decades, overweight (BMI > 25 kg/m2) and obesity (BMI > 30 kg/m2) increased not only in the general population but also among IBD patients [97]. In a time-trend analysis involving 10,282 CD patients (from 1991 to 2008) and including 40 RCT, the mean BMI increased from 20.8 to 27.0 [98]. Obesity and overweight were reported respectively in 18% and 38% of IBD patients, with a higher percentage of obesity in CD than in UC [99].
Only few studies analyzed the impact of obesity on the course of UC, while more data are available in CD patients. Anyhow, results are often contrasting and non-conclusive [100]. Obesity may be associated with a worse prognosis in CD in terms of perianal complications, disease activity, hospitalization, time to the first surgery and more aggressive medical treatment [101]. This could be probably due to the ability of visceral fat to produce cytokines and thus promote inflammation [102].

3.3.1. Clinical Disease Course and Response to Therapies

Obesity and overweight seem to negatively impact the efficacy of medications [103].
Obese patients treated with thiopurines and anti-TNF could have worse outcomes compared to non-obese patients, due to subtherapeutic dosing: a retrospective study among 1494 IBD patients (634 UC, 860 CD) found a lower dosing per kilogram of AZA, 6-MP, methotrexate and anti-TNF in the obese subgroup (p < 0.0001) [104].
A retrospective analysis conducted on CD patients revealed that during the first year of therapy with IFX, clinical flare, loss of response and need for surgery are improved by BMI increase, but extreme values (BMI < 18.5 kg/m2 and BMI > 30 kg/m2) are associated with worse outcomes [17]. Moreover, obese patients starting IFX may also have a higher risk of flare (HR 3.03, p < 0.001), with an earlier flare the greater the BMI, both in CD (HR = 1.06 per 1 kg/m2 increase, p = 0.02) and in UC (HR = 1.30 per 1 kg/m2 increase, p = 0.01) [19]. Nevertheless, the literature data are not univocal.
From an analysis of pooled data among 1014 CD patients exploring prognostic factors to IFX therapy persistence, emerges that the difference in the prevalence of obesity between American and European patients did not influence long-term (>5 years) use of IFX [105]. For some authors, obesity could be a risk factor for a shorter time to dose escalation in ADA patients (p = 0.013), but not for IFX (p = 0.164) [16]. Another pooled analysis among IBD patients treated with IFX (723 CD, 484 UC) explored the influence of BMI on clinical remission, clinical response, and mucosal healing; it concluded that obesity is not associated with an inferior response to IFX (OR 0.94, p = 0.97; OR 0.84, p = 0.45; OR 1.13, p = 0.95, respectively) [106].

3.3.2. Risk of Surgery

A review including 22 articles showed an association between visceral adiposity and both the risk of complex CD (OR 26.1; 95% CI 2–75.4, p = 0.02), and POR (RR 2.1; 95% CI 1.5–3; p = 0.012). However, data about visceral adiposity on postsurgical complications and the efficacy of medical therapy are conflicting [107].
A meta-analysis including 7 studies (5 retrospective and 2 prospective–5 with IBD patients and 2 with CD patients only) comparing obese and non-obese patients, showed no differences in the risk of perianal disease (RR = 0.97, p = 0.81) and use of anti-TNF (RR= 0.89, p = 0.26) or immunomodulators (RR = 0.96, p = 0.43); moreover, obese patients have a lower risk of hospitalization (RR 0.84, p = 0.003) and surgery (RR 0.82, p = 0.003). However, the authors hypothesized that obesity could be an indirect sign of remission [108].
Similar results were achieved in a cross-sectional study among 846 CD patients, concluding that there was no association between obesity and perianal disease, stricturing disease, or surgery (OR = 0.71; 95% CI 0.39–1.28), while obesity was associated with a lower risk of penetrating disease (OR = 0.56; 95% CI 0.31–0.99) [109].
A retrospective study on 148 CD patients, comparing patients with BMI < 18.5 kg/m2 with those with a BMI > 25 kg/m2, showed that the latters have a significantly shorter time to first surgery (252 versus 24 months, respectively; p = 0.043) [110]. Other two retrospective studies evaluating visceral adipose tissue by CT scan in CD patients, concluded that high visceral adipose subjects have an increased risk of surgery (OR = 2.02; p = 0.006) [111] and of complicated disease (stricturing/penetrating disease and previous surgeries) (OR 26.1; 95% CI 2.0–754; p = 0.02) [67].

3.3.3. Post-Operative Recurrence and Surgery-Related Complications

Concerning the prognostic impact of obesity on IBD patients undergoing surgery, more data are available and quite concordant. There are two recent meta-analyses including a total of 16,933 IBD patients confirming that obese patients are at higher risk of post-surgical complication (OR = 1.45; CI 95% 1.24–1.69) [112] (OR = 1.33; CI 95% 1.04–1.70) [113]. These results seem to be confirmed even in another recent meta-analysis including a more heterogeneous population—not only IBD—undergoing colectomy and IPAA (OR 2.27; CI 95% 1.42–3.61) [114] and in three more studies not included in the above-mentioned meta-analyses [115,116,117].
A post-hoc analysis of POCER study including 44 CD patients concluded that visceral adiposity is a risk factor for endoscopic post-operative recurrence [70]. A population-based study among 143 CD patients who underwent elective ileocolectomy and studied by preoperative CT scan, found that the visceral/subcutaneous fat ratio is a more reliable predictor of post-operative morbidity than conventional adiposity markers such as BMI (p = 0.03) [101].

3.3.4. Main Evidence and Clinical Implications

-
Obesity may worsen the effectiveness of IBD therapies, but the evidence is very uncertain.
-
Data concerning obesity and the risk of surgery are contrasting and the quality of evidence is low.
-
The only meta-analysis concludes that obesity does not increase the risk of perianal disease, stricture disease, surgery, or the use of immunomodulators/anti-TNF therapies.
-
Obesity increases post-operative complications in IBD patients and in CD is a risk factor for endoscopic post-operative recurrence.

3.4. Albuminemia

Chronic inflammation of the mucosa determines both malabsorption and intestinal protein losses, resulting in hypoalbuminemia.

3.4.1. Clinical Disease Course and Response to Therapies

In IBD, hypoalbuminemia correlates with more relapses, with secondary loss of response to infliximab [118,119] and with increased clearance of infliximab and golimumab [120,121].
Low albumin levels are associated with longer hospitalizations and with higher rates of surgery (OR 2.54; 95% CI 1.15–3.93) [122].
In CD patients, the higher the albumin levels, the higher the clinical remission rate after a dose escalation of infliximab [123]; while hypoalbuminemia is associated with a worse outcome, a greater risk of relapse and interrupting anti-TNF therapy for albumin serum levels < 3.5 g/dL (p = 0.0274) [124,125,126].
Albumin levels are directly proportional to adalimumab levels and inversely proportional to anti-adalimumab antibodies [127]. Moreover, values of albuminemia < 3 g/dL correlate with worse outcomes in patients with enterocutaneous fistulas and with a higher probability of abdominal abscesses during infliximab therapy [128,129].
Hypoalbuminemia is a negative prognostic factor also for UC, associated with a more severe course [130], a greater use of steroids, a worse endoscopic response/remission and primary failure to infliximab [131,132].

3.4.2. Risk of Surgery

In UC patients with hypoalbuminemia, a higher likelihood of colectomy has been shown [131,133,134]; albumin values > 3.5 g/dL are an independent factor of colectomy-free survival with OR 3.03 (95% CI, 1.12–8.22; p = 0.029) [135], while values < 3 g/dL (HR, 2.67; 95% CI, 1.20–5.92) are associated with an increased risk of colectomy, and values < 2.45 g/dL on admission represent a significant independent predictor of colectomy (OR 6.097, 95% CI 1.8310–20.3047) [136,137]. Increasing CPR/albumin ratio and platelet/albumin ratio are suggested by some authors as predictors of rescue therapy failure and, as a consequence, of surgery [138,139,140,141,142,143,144].

3.4.3. Post-Operative Recurrence and Surgery-Related Complications

Literature evidence demonstrates a higher incidence of post-surgical complications, mortality, anastomotic leaks (OR 2.8; 95% CI 1.3–5.1; p = 0.03), post-operative ileus, and a higher prevalence of incisional hernia (HR 2.02, p = 0.002) in IBD patients with hypoalbuminemia (OR 2.72; p = 0.03) [145,146,147,148,149].
In particular, for UC patients a greater risk of portal-mesenteric thrombosis and infectious complications have been reported, as well as a higher rate of post-colectomy reoperation and IPAA failure [150,151,152,153,154,155].
For CD patients, low serum albumin levels seem to correlate with a high risk of post-operative complications with ORs ranging from 1.207 to 2.232, especially septic complications [156,157,158,159,160,161,162,163,164,165]. Even abscess drainage, whether surgical or percutaneous, has a higher risk of complications in such patients (OR 0.921; 95% CI 0.89–0.96) [166].
Post-surgery endoscopic recurrences after initiating anti-TNF therapy are more frequent for albumin values < 3.3 g/dL with an OR of 34.10 (95% CI, 1.72–28.04) [167].

3.4.4. Main Evidence and Clinical Implications

All studies showed that albumin plays a fundamental role in the disease course, response to drugs, surgery rates, and even more concerning the post-surgical complications both in CD and UC patients. Therefore, albumin is a relevant factor to be kept in mind in the management of IBD patients.

4. Anemia and Micronutrients

The chronic inflammatory status and the impaired absorption of nutrients due to bowel damage leads to a possible deficiency of vitamins and micronutrients that are crucial for the overall well-being [11]. If the low serum levels of these microelements are a cause or an effect of disease activity remains unclear, and if supplementation of these elements could be a potential therapeutic target is not well defined. However, the evidence is sufficiently solid in showing that active disease is linked to low serum micronutrient levels (Table 3).
The micronutrients and vitamins most involved in IBD are iron, selenium, zinc, copper, manganese, vitamin D, vitamin B12 and folic acid, vitamins A, E, C, K, B1 and B6. Micronutrient and vitamin deficiencies may be linked only to IBD or also to the concomitant presence of other autoimmune diseases such as autoimmune chronic atrophic gastritis (leading to malabsorption of iron and vitamin B12) and celiac disease (responsible for malabsorption of iron and folic acid) [192].

4.1. Anemia

Anemia is a common complication of IBD. According to the World Health Organization (WHO) criteria, anemia is defined as a hemoglobin (HgB) level less than 13 g/dL in men and 12 g/dL in non-pregnant females [193].
The most common causes of anemia in IBD are iron deficiency, folic acid or vitamin B12 deficiency, chronic disease anemia due to inflammation, and combined causes. Certainly, it is crucial to classify the etiology of anemia in IBD to select the correct treatment [194,195].
Pure iron deficiency anemia is defined in the case of ferritin serum levels < 30 μg/L and the normal value of CRP; chronic disease anemia is defined by ferritin serum level > 100 μg/L and high levels of CRP. Combined anemia is defined by ferritin serum level < 100 μg/L and high levels of CRP [196].
Anemia affects the quality of life, cognitive functions, the ability to work, hospitalization, and healthcare costs [197]; if low levels of HgB could affect the disease course of IBD is not well known.

4.1.1. Clinical Disease Course and Response to Therapies

A retrospective study on CD patients showed that anemia is a predictive factor for loss of response to anti-TNF [198]; moreover, anemic CD patients had two-times greater odds ratio of severe hospitalization compared with non-anemic patients (OR 1.49, 95% CI: 1.37–1.61) [168].
In UC patients, anemia is a marker of disease severity (e.g., Truelove and Witts index) [199,200], a predictor of relapse in patients treated with 5-ASA, and a predictor of re-admission to hospital, as shown in two large nationwide studies [169,170]. Furthermore, anemia seems to be a predictor of colectomy [171].
Two prospective studies demonstrated that anemia is associated with the extension of the disease, treatment escalation, and disease outcomes (including hospitalization and surgery) in both CD and UC, therefore, its evaluation can be helpful in stratifying disease severity [201,202].

4.1.2. Post-Operative Recurrence and Surgery-Related Complications

Seven retrospective studies analyzed incidence and risk factors for post-operative complications in CD patients undergoing bowel resection, showing that pre-operative anemia was associated with a higher risk of post-operative morbidity and mortality [172,173,174], higher risk of sepsis [160], surgical site infection [175] and post-surgical complications [176] such as prolonged postoperative ileus [177].

4.1.3. Main Evidence and Clinical Implications

-
Current evidence suggests that anemia could be associated with high severity of IBD.
-
All studies showed a worse disease course or higher risk of post-surgical complications in IBD anemic patients.
-
Preoperative correction of anemia may improve surgical outcomes.

4.2. Iron

Iron deficiency (ID) is one of the worldwide most common disorders, affecting about 50% of IBD patients [195,203,204], with a prevalence ranging from 26.5% to 62.5% and representing the most common micronutrient deficiency of IBD [195,205,206,207].
In IBD patients, the diagnostic criteria for ID depend on the severity of inflammation: in remission, serum ferritin < 30 µg/L and transferrin saturation index (TSAT) < 16% are indicative of ID, while during the acute phase of the disease (CRP > 5 mg/L and/or FC > 150 mg/kg), ID is defined as ferritin < 100 µg/L.
Iron is an essential trace element involved in many cellular processes including oxygen transport, mitochondrial electron transport, gene regulation and DNA synthesis, and its deficit can manifest through a court of very heterogeneous extra-bowel symptoms including chronic fatigue, sleep disorders, agitation, decreased physical and cognitive performance, immune system impairment, significantly affecting the patient’s wellbeing [178].
Female patients and severe disease activity patients are at higher risk, due to menstrual losses in premenopausal women and to bloody diarrhoea. Interestingly, patients with ID without anaemia presented health-related quality of life (HRQoL) questionnaires with lower overall scores [178,208].

4.2.1. Clinical Disease Course and Response to Therapies

ID in the absence of anaemia negatively impacts the normal perception of HRQoL in patients with IBD, so its early diagnosis and correction could be a valuable target in the treatment of these patients [178,179,180]. Few studies have examined the effects of treating ID in patients with IBD without anaemia using different formulations of intravenous iron and in all of them, there was a significant improvement in patient’ symptoms and HRQoL, independently of disease activity [181,182].
Recently a multicentre, prospective, observational study by Eliadou and colleagues, including 98 IBD patients with ID without anaemia, showed that 1 month after a single 500 mg dose of intravenous ferric carboxymaltose (FCM) there was a significant increase of serum ferritin, serum iron, TSAT, and an improvement of symptoms [209]. Moreover, improvements in the mean EQ-5D scores among CD (p < 0.01) and UC (p < 0.05) patients were observed [182].
Another single-centre prospective study, including 84 CD patients and 24 healthy volunteers showed that decreased serum iron and total iron binding capacity correlate negatively with CDAI scores (r = −0.513, r = −0.409, both p < 0.01), representing independent risk factors for serious disease in a logistic regression analysis, with a sensitivity of 32.7% and a specificity of 100% (AUC = 0.812, p < 0.01); a cut-off value of serum iron of 5.25 µmol/L was used to distinguish moderate from mild/remission groups (AUC = 0.729, p = 0.001; sensitivity: 48.1%; specificity 93.5%) [181].
There are no definitive recommendations for the treatment of iron deficiency without anaemia (IDWA). Oral intake is not tolerated in up to 20% of patients and could expose patients to adverse effects perceived as symptoms worsening [205,210,211,212,213]. Intravenous iron is more effective, has an excellent security profile and has a reduced risk of adverse events, resulting in better tolerance [214,215].

4.2.2. Main Evidence and Clinical Implications

Iron supply in patients with IBD and IDWA improves the symptoms related to ID and HRQoL.

4.3. Vitamin B12 and Folic Acid

Vitamin B12 and folate deficiencies are common in patients with IBD. Folate deficiency is due to a combination of factors: poor diet, malabsorption, an increased requirement due to the increased granulocytes and other inflammatory cells, severe inflammation, resection, enteric fistulas and the use of drugs such as sulfasalazine and methotrexate [216,217].
As shown in a recent meta-analysis, the folate level in IBD patients was significantly lower compared to healthy groups; however, a lower concentration of folate was found in UC but not in CD patients [218].
Vitamin B12 deficiency is more common in CD patients compared to UC, with a prevalence of 33% and 16% respectively [219].
In CD patients, prior intestinal surgery is an independent risk factor for low serum concentrations of vitamin B12 [220]. A meta-analysis by Battat et al. identified that an ileal resection longer than 20 cm is the only factor to predispose CD patients to vitamin B12 deficiency [221].
Vitamin B12 is absorbed in the distal ileum, the intestinal tract most commonly involved in CD, and this would explain the higher prevalence of vitamin B12 deficiency in CD compared to UC.
A periodical assessment of blood levels of vitamins and iron is suggested. Guideline recommendations suggest checking haemoglobin and iron status every 6–12 months for patients in remission or with mild disease, and every 3 months in case of active disease. For patients at risk of vitamin B12 or folic acid deficiency (e.g., small bowel disease or resection), serum levels should be measured at least annually, or when macrocytosis is present [193].

Main Evidence and Clinical Implications

Folate deficiency is more common in UC patients while B12 deficiency is more common in CD patients.

4.4. Vitamin D

During the last few years, there has been an increase in interest concerning the immuno-modulating role of vitamin D [25,222]. Firstly, several observational and cross-sectional studies included in three meta-analyses showed that IBD patients presented low serum vitamin D levels compared to healthy people [183,223,224]. A meta-regression analysis shows that latitude does not influence the association between IBD and vitamin D deficiency (p = 0.34) [223].

4.4.1. Clinical Disease Course and Response to Therapies

An inverse correlation between serum vitamin D levels and disease activity was observed in both UC and CD in all published meta-analyses [183,184]. IBD patients with low vitamin D levels present increased odds of disease activity, mucosal inflammation and future clinical relapse (OR: 1.53, CI 1.32–1.77; OR 1.30, CI 1.06–1.60; OR 1.23, 1.03–1.47, respectively) [184].
However, based on available research it is impossible to understand if vitamin D deficiency is a cause or an effect of disease activity.
Seven RCTs, included in a recent meta-analysis, evaluated disease activity as a dichotomous outcome in IBD patients treated with vitamin D supplementation, showing that the pooled risk ratio of clinical relapse was 0.64 (CI 0.46–0.89) among 458 IBD-treated patients. This data appeared more solid in CD patients in clinical remission (OR 0.47, CI 0.27–0.82). On the other hand, seven studies, included in the same meta-analysis, reported data concerning the impact of vitamin D supplementation on the disease activity score evaluated as a continuous outcome. The authors reported a slight, not statistically significant, effect on CD patients (SMD, −0.29; −0.71–0.14), and no differences in UC (SMD, 0.24, CI −0.61–1.10) [186].
Other four meta-analyses reported data concerning the effectiveness of vitamin D supplementation on disease course in IBD, however, none of these had the clinical relapse or the disease activity as their primary outcome. Guo et al. showed that vitamin D supplementation decreased serum CRP, but it did not decrease the disease activity index and relapse rate. However, only three RCTs were included [225]. Other two meta-analyses including both RCT and observational studies showed an improvement in Harvey Bradshaw index (HBI) of −1.47 points (CI −2.47–−0.47) and a reduction in relapse rate (OR 0.34, CI 0–20–0.58) [226,227].
Another meta-analysis on this topic, including only UC patients, showed a significant reduction in both relapse rate and disease activity, evaluated with the partial Mayo score [228].
In one of the above-mentioned meta-analyses, the authors tried to evaluate the optimal vitamin D dose supplementation with a meta-regression model showing the optimal effect with an intermediate dosage (10,000–15,000 IU/day) [186].
There are four observational studies [24,229,230,231] and two RCTs [232,233] assessing the relationship between vitamin D levels, its supplementation and the response to biologics. Bendix and colleagues showed that a seven weeks high-dose vitamin D treatment reduces the need for later infliximab dose-escalation and reduces inflammatory markers [232]. However, the other RCT did not show a significant reduction in serum TNFα levels [233]. Moreover, all the observational studies showed a significant correlation between low serum vitamin D levels and response to biological therapy. One retrospective study observed a high remission rate, higher reduction in HBI, and increased remission rate, in patients regularly treated with Infliximab and vitamin D supplementation [231]. Additionally, on the multivariate analysis low serum vitamin D levels (<25 ng/mL) were associated with primary non-response to Vedolizumab, failure after one year of follow-up (OR 26.10, CI 14.30–48.90 and OR 6.10, CI 3.06–12.17, respectively) and earlier cessation of anti TNFα therapy (HR 2.13, CI 1.03–4.39) [229,230]. Finally, serum vitamin D levels < 25 ng/mL were associated with loss of response to the biological therapy [24].

4.4.2. Risk of Surgery and Post-Operative Recurrence

Concerning the risk of surgery, a single retrospective study observed that serum vitamin D levels < 20 ng/mL were associated with an increased risk of surgery (OR 1.76, CI 1.24–2.51) and of hospitalization compared to patients with level ≥ 30 ng/mL in both CD and UC [234].
Vitamin D levels > 30 ng/mL were also associated with a reduced risk of endoscopic recurrence in patients who underwent a prior intestinal resection [235], although vitamin D supplementation does not prevent postoperative recurrence in CD patients [185].

4.4.3. Main Evidence and Clinical Implications

-
Low serum vitamin D levels are associated with an increase in disease activity and a worse clinical course in IBD patients. Furthermore, vitamin D supplementation results in a slight reduction of clinical relapse. This effect seems to be higher among CD patients.
-
Although with low-level evidence, low serum vitamin D levels are associated with an increased risk of surgery.

4.5. Other Vitamins (A, E, K, Group B, and C)

As for other nutritional deficits, vitamin deficiency is common in IBD patients, and its pathogenesis is multifactorial [192].
A meta-analysis including 19 case-control studies, showed a lower serum level of fat-soluble vitamins (A, D, E, K) in IBD patients compared to the control group.
Interestingly, in the meta-regression analysis, significant associations between vitamin A levels in CD patients, and the levels of inflammatory biomarkers (CRP: p = 0.03, and albumin p = 0.0003), were found. The data concerning vitamins E and K were not enough strong to show a correlation with disease activity, however, a clear trend was found [236]. Another study showed a lower level of vitamin K in CD. The vitamin K level (evaluated by measuring serum undercarboxylated osteocalcin) was significantly correlated with the clinical activity index among CD patients [237].
Few studies reported that vitamin C deficiency is relatively common in IBD patients, in particular in patients with reduced intake of vegetables and fruit [238,239].
Little evidence showed a plausible increase in vitamin A levels after adequate treatment for disease activity in UC patients. Plasma vitamin A is significantly lower in active UC patients compared to the control group (p = 0.0005) [240,241]. Another retrospective study including CD patients who underwent surgery showed a significantly higher basal peroxidative state and lower levels of Vitamin A and E compared to controls among the CD patients. Two months after surgery, a significant increase in serum vitamin A levels but not Vitamin E was found [242].
Concerning the other vitamin of group B only a few data are available [192,243]. However, some evidence showed that Vitamin B1 deficiency could be related to chronic fatigue in IBD patients [244].

4.5.1. Clinical Disease Course

Vitamin A: The most promising results were observed with vitamin A supplementation. In an RCT including 150 UC pa-tients with moderate-severe disease activity were randomized to receive 25.000 IU/die of Vitamin A or placebo for two months as adjunctive treatment (both groups were treated with 5-ASA for at least a month before study entry). At the end of the follow-up, a significant decrease in the Mayo Clinic score (p < 0.001) and sub-scores (p < 0.001) was observed in the intervention group. However, another small RCT, published in 1985 showed different results. In this study, there was no correlation between vitamin A levels, and activity indices, and the clinical relapse rate was similar in the two groups [187]. However, a lower relapse rate was observed in the vitamin A-treated group, despite the low sample size included in the study.
Vitamin B: A randomized, double-blinded, placebo-controlled crossover trial including IBD patients with a clinical re-mission evaluated the efficacy of 4 weeks of high-dose oral thiamine (600–1800 mg/dL of B1 vitamin) on chronic fatigue. This study showed a significant reduction in the Inflammatory Bowel Disease-Fatigue Questionnaire during the treatment period [188].
Other promising results derived from a prospective, non-randomized, study of intervention including CD patients treated with 100 mg riboflavin (vitamin B2) daily for 3 weeks. Riboflavin supplementation significantly decreased serum levels of inflammatory markers and the clinical activity evaluated with HBI [189].

4.5.2. Main Evidence and Clinical Implications

-
Despite the very low quality of available evidence, Vitamin A and B2 supplementation may have little to no effect in reducing disease activity.
-
High dose of Vitamin B1 may reduce chronic fatigue in IBD patients.

4.6. Other Trace Elements (Selenium, Zinc, Copper, Manganese)

Zinc and Selenium are involved in the regulation of the immune response, inflammatory processes, and the regulation of oxidative stress [245,246]. Considering this, their low serum concentration may exacerbate inflammation through the dysfunction of the epithelial barrier, an altered mucosal immunity, and an increased production of pro-inflammatory cytokines.

4.6.1. Selenium and Zinc

A study from Siva et al. confirmed that IBD patients with zinc deficiency are more likely to have adverse disease outcomes: a higher risk of surgery, hospitalization, and complications [247].
Evidence on the supplementation of zinc and selenium in IBD is scarce and mostly on mice models. Daily zinc supplementation in CD patients improved intestinal permeability [248], CDAI score and serum zinc level [249]. A randomized, placebo-controlled, trial demonstrated that zinc-carnosine chelate compound enemas used in patients with active UC receiving induction therapy offer a better clinical response or remission than placebo [190]. Concerning selenium, a prospective randomized interventional trial demonstrated that adding Selenium to Infliximab in IBD patients, can reduce adverse drug reactions [250] and can improve clinical symptoms, in patients with mild to moderate UC [191].

4.6.2. Copper and Manganese

Results about serum copper concentrations in IBD patients are not univocal [251,252]. In contrast to zinc, copper concentrations increase during the acute-phase response, resulting in an increased copper/zinc ratio, that should be more relevant than the concentration of copper alone [253].
Although it has an important antioxidant role, manganese is the less studied trace element. An interesting work on IBD patients with ileal pouch-anal anastomosis for a prolonged period showed a significantly increased blood manganese concentration compared to healthy controls. Authors try to interpret this as the result of an increased manganese absorption linked to iron deficiency and the use of antidiarrheal medications [254].

4.6.3. Main Evidence and Clinical Implications

Few data are available concerning the trace elements and their potential impact on the IBD disease course. More systematic screening of trace element status in IBD patients, as well as clinical trials on trace element supplementation are needed.

5. Conclusions

In a more and more ambitious approach to IBD with a treat-to-target strategy that considered tighter objectives such as mucosal healing, transmural healing, histological healing, and overall, a deep remission of the disease even the nutritional status must be considered [1,2,3,4] (Figure 1).
The evidence summarized in this review showed that many nutritional aspects could be potential targets to induce a better control of symptoms, a deeper remission, and overall improve the quality of life of IBD patients (Table 4).
Certainly, many aspects summarized in this review are still lacking strong evidence. Few data are available concerning the effect of nutritional status on the induction of remission and the impact on the risk of surgery. Obviously, some clinical outcomes need data deriving from RCTs or non-randomized studies of intervention with a long follow-up (such as the risk of surgery, POR, and clinical relapse). Often these data are still lacking. Moreover, considering that the majority of available data derive from observational studies, inclusion criteria, and the analyzed outcome are often heterogeneous.
However, the large number of studies included and analyzed in this review allow us to produce a very extensive overview concerning this issue stating practical clinical aspects and highlighting the current knowledge gap helping to drive future research. An optimal nutritional status and the good management of micronutrient deficiency, ideally with the help of dietitians, may reduce this risk of clinical relapse, risk of surgery and post-operative recurrence. Considering this, all these variables should be considered for the general assessment and monitoring of IBD patients.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/nu15173824/s1, Table S1: Systematic bibliographic research; Table S2: Articles selection process; Figure S1: PRISMA flow diagram.

Author Contributions

M.V.: Methodological assessment, write the original draft; A.C. and N.C.: Critical reviewed the manuscript, write the original draft; S.F. (Stefano Fabiani): write the original draft; G.S., S.F. (Sara Frassino), S.M. and M.M.: write the original draft, reviewed the literature; A.V. and G.L.: critical revision of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing not applicable to this article as no datasets were generated or analyzed.

Acknowledgments

We thank Giuseppe Sparaco for the figure editing.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Veauthier, B.; Hornecker, J.R. Crohn’s Disease: Diagnosis and Management. Am. Fam. Physician 2018, 98, 661–669. [Google Scholar]
  2. Wilkins, T.; Jarvis, K.; Patel, J. Diagnosis and Management of Crohn’s Disease. Am. Fam. Physician 2011, 84, 1365–1375. [Google Scholar]
  3. Dixon, L.J.; Kabi, A.; Nickerson, K.P.; McDonald, C. Combinatorial Effects of Diet and Genetics on Inflammatory Bowel Disease Pathogenesis. Inflamm. Bowel Dis. 2015, 21, 912–922. [Google Scholar] [CrossRef]
  4. Leone, V.; Chang, E.B.; Devkota, S. Diet, Microbes, and Host Genetics: The Perfect Storm in Inflammatory Bowel Diseases. J. Gastroenterol. 2013, 48, 315–321. [Google Scholar] [CrossRef]
  5. Lane, E.R.; Zisman, T.; Suskind, D. The Microbiota in Inflammatory Bowel Disease: Current and Therapeutic Insights. J. Inflamm. Res. 2017, 10, 63–73. [Google Scholar] [CrossRef]
  6. Spooren, C.E.G.M.; Pierik, M.J.; Zeegers, M.P.; Feskens, E.J.M.; Masclee, A.A.M.; Jonkers, D.M.A.E. Review Article: The Association of Diet with Onset and Relapse in Patients with Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2013, 38, 1172–1187. [Google Scholar] [CrossRef]
  7. Wu, G.D. Diet, the Gut Microbiome and the Metabolome in IBD. In Nutrition, Gut Microbiota and Immunity: Therapeutic Targets for IBD: 79th Nestlé Nutrition Institute Workshop: New York, NY, USA, September 2013; S Karger Ag: Basel, Switzerland, 2014; pp. 73–82. [Google Scholar]
  8. Buchanan, E.; Gaunt, W.W.; Cardigan, T.; Garrik, V.; McGrogan, P.; Russel, R.K. The Use of Exclusive Enteral Nutrition for Induction of Remission in Children with Crohn’s Disease Demonstrates That Disease Phenotype Does Not Influence Clinical Remission. Aliment. Pharmacol. Ther. 2009, 30, 501–507. [Google Scholar] [CrossRef]
  9. Johnson, T. Treatment of Active Crohn’s Disease in Children Using Partial Enteral Nutrition with Liquid Formula: A Randomised Controlled Trial. Gut 2006, 55, 356–361. [Google Scholar] [CrossRef]
  10. Rubio, A.; Pigneur, B.; Garnier-Lengliné, H.; Talbotec, C.; Schmitz, J.; Canioni, D.; Goulet, O.; Ruemmele, F.M. The Efficacy of Exclusive Nutritional Therapy in Paediatric Crohn’s Disease, Comparing Fractionated Oral vs. Continuous Enteral Feeding. Aliment. Pharmacol. Ther. 2011, 33, 1332–1339. [Google Scholar] [CrossRef]
  11. Sigall-Boneh, R.; Levine, A.; Lomer, M.; Wierdsma, N.; Allan, P.; Fiorino, G.; Gatti, S.; Jonkers, D.; Kierkuś, J.; Katsanos, K.H.; et al. Research Gaps in Diet and Nutrition in Inflammatory Bowel Disease. A Topical Review by D-ECCO Working Group [Dietitians of ECCO]. J. Crohns Colitis 2017, 11, 1407–1419. [Google Scholar] [CrossRef]
  12. Gerasimidis, K.; McGrogan, P.; Edwards, C.A. The Aetiology and Impact of Malnutrition in Paediatric Inflammatory Bowel Disease. J. Hum. Nutr. Diet. 2011, 24, 313–326. [Google Scholar] [CrossRef] [PubMed]
  13. Cameron, F.L.; Gerasimidis, K.; Papangelou, A.; Missiou, D.; Garrick, V.; Cardigan, T.; Buchanan, E.; Barclay, A.R.; McGrogan, P.; Russell, R.K. Clinical Progress in the Two Years Following a Course of Exclusive Enteral Nutrition in 109 Paediatric Patients with Crohn’s Disease. Aliment. Pharmacol. Ther. 2013, 37, 622–629. [Google Scholar] [CrossRef] [PubMed]
  14. Vasseur, F.; Gower-Rousseau, C.; Vernier-Massouille, G.; Dupas, J.L.; Merle, V.; Merlin, B.; Lerebours, E.; Savoye, G.; Salomez, J.L.; Cortot, A.; et al. Nutritional Status and Growth in Pediatric Crohn’s Disease: A Population-Based Study. Am. J. Gastroenterol. 2010, 105, 1893–1900. [Google Scholar] [CrossRef] [PubMed]
  15. Singh, S.; Dulai, P.S.; Zarrinpar, A.; Ramamoorthy, S.; Sandborn, W.J. Obesity in IBD: Epidemiology, Pathogenesis, Disease Course and Treatment Outcomes. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 110–121. [Google Scholar] [CrossRef] [PubMed]
  16. Bhalme, M.; Sharma, A.; Keld, R.; Willert, R.; Campbell, S. Does Weight-Adjusted Anti-Tumour Necrosis Factor Treatment Favour Obese Patients with Crohn’s Disease? Eur. J. Gastroenterol. Hepatol. 2013, 25, 543–549. [Google Scholar] [CrossRef] [PubMed]
  17. Brown, P.; Clark, T.; Dowson, G.; Warren, L.; Hamlin, J.; Hull, M.; Subramanian, V. Relationship of Body Mass Index to Clinical Outcomes after Infliximab Therapy in Patients with Crohn’s Disease. J. Crohns Colitis 2016, 10, 1144–1150. [Google Scholar] [CrossRef] [PubMed]
  18. Guerbau, L.; Gerard, R.; Duveau, N.; Staumont-Sallé, D.; Branche, J.; Maunoury, V.; Cattan, S.; Wils, P.; Boualit, M.; Libier, L.; et al. Patients with Crohn’s Disease with High Body Mass Index Present More Frequent and Rapid Loss of Response to Infliximab. Inflamm. Bowel Dis. 2017, 23, 1853–1859. [Google Scholar] [CrossRef]
  19. Harper, J.W.; Sinanan, M.N.; Zisman, T.L. Increased Body Mass Index Is Associated with Earlier Time to Loss of Response to Infliximab in Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2013, 19, 2118–2124. [Google Scholar] [CrossRef]
  20. Adams, D.W.; Gurwara, S.; Silver, H.J.; Horst, S.N.; Beaulieu, D.B.; Schwartz, D.A.; Seidner, D.L. Sarcopenia Is Common in Overweight Patients with Inflammatory Bowel Disease and May Predict Need for Surgery. Inflamm. Bowel Dis. 2017, 23, 1182–1186. [Google Scholar] [CrossRef]
  21. Maaser, C.; Sturm, A.; Vavricka, S.R.; Kucharzik, T.; Fiorino, G.; Annese, V.; Calabrese, E.; Baumgart, D.C.; Bettenworth, D.; Borralho Nunes, P.; et al. ECCO-ESGAR Guideline for Diagnostic Assessment in IBD Part 1: Initial Diagnosis, Monitoring of Known IBD, Detection of Complications. J. Crohns Colitis 2019, 13, 144–164. [Google Scholar] [CrossRef]
  22. Gubatan, J.; Moss, A.C. Vitamin D in Inflammatory Bowel Disease: More than Just a Supplement. Curr. Opin. Gastroenterol. 2018, 34, 217–225. [Google Scholar] [CrossRef] [PubMed]
  23. Bischoff, S.C.; Barazzoni, R.; Busetto, L.; Campmans-Kuijpers, M.; Cardinale, V.; Chermesh, I.; Eshraghian, A.; Kani, H.T.; Khannoussi, W.; Lacaze, L.; et al. European Guideline on Obesity Care in Patients with Gastrointestinal and Liver Diseases—Joint European Society for Clinical Nutrition and Metabolism/United European Gastroenterology Guideline. United Eur. Gastroenterol. J. 2022, 10, 663–720. [Google Scholar] [CrossRef] [PubMed]
  24. Valvano, M.; Magistroni, M.; Mancusi, A.; D’Ascenzo, D.; Longo, S.; Stefanelli, G.; Vernia, F.; Viscido, A.; Necozione, S.; Latella, G. The Usefulness of Serum Vitamin D Levels in the Assessment of IBD Activity and Response to Biologics. Nutrients 2021, 13, 323. [Google Scholar] [CrossRef] [PubMed]
  25. Vernia, F.; Valvano, M.; Longo, S.; Cesaro, N.; Viscido, A.; Latella, G. Vitamin D in Inflammatory Bowel Diseases. Mechanisms of Action and Therapeutic Implications. Nutrients 2022, 14, 269. [Google Scholar] [CrossRef] [PubMed]
  26. Latella, G.; Caprilli, R. Metabolism of Large Bowel Mucosa in Health and Disease. Int. J. Color. Dis. 1991, 6, 127–132. [Google Scholar] [CrossRef] [PubMed]
  27. Jowett, S.L. Influence of Dietary Factors on the Clinical Course of Ulcerative Colitis: A Prospective Cohort Study. Gut 2004, 53, 1479–1484. [Google Scholar] [CrossRef] [PubMed]
  28. Sasso, A.; Latella, G. Dietary Components That Counteract the Increased Risk of Colorectal Cancer Related to Red Meat Consumption. Int. J. Food Sci. Nutr. 2018, 69, 536–548. [Google Scholar] [CrossRef] [PubMed]
  29. Morton, H.; Pedley, K.C.; Stewart, R.J.C.; Coad, J. Inflammatory Bowel Disease: Are Symptoms and Diet Linked? Nutrients 2020, 12, 2975. [Google Scholar] [CrossRef]
  30. O’Sullivan, M.; O’Morain, C. Nutrition in Inflammatory Bowel Disease. Best Pract. Res. Clin. Gastroenterol. 2006, 20, 561–573. [Google Scholar] [CrossRef]
  31. Lambert, K.; Pappas, D.; Miglioretto, C.; Javadpour, A.; Reveley, H.; Frank, L.; Grimm, M.C.; Samocha-Bonet, D.; Hold, G.L. Systematic Review with Meta-analysis: Dietary Intake in Adults with Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2021, 54, 742–754. [Google Scholar] [CrossRef]
  32. Comeche, J.M.; Comino, I.; Altavilla, C.; Tuells, J.; Gutierrez-Hervas, A.; Caballero, P. Parenteral Nutrition in Patients with Inflammatory Bowel Disease Systematic Review, Meta-Analysis and Meta-Regression. Nutrients 2019, 11, 2865. [Google Scholar] [CrossRef] [PubMed]
  33. Zachos, M.; Tondeur, M.; Griffiths, A. Enteral Nutritional Therapy for Induction of Remission in Crohn’s Disease. In The Cochrane Database of Systematic Reviews; Zachos, M., Ed.; John Wiley & Sons, Ltd: Chichester, UK, 2001. [Google Scholar]
  34. Narula, N.; Dhillon, A.; Zhang, D.; Sherlock, M.E.; Tondeur, M.; Zachos, M. Enteral Nutritional Therapy for Induction of Remission in Crohn’s Disease. Cochrane Database Syst. Rev. 2018, 4, CD000542. [Google Scholar] [CrossRef] [PubMed]
  35. Messori, A.; Trallori, G.; D’albasio, G.; Milla, M.; Vannozzi, G.; Pacini, F. Defined-Formula Diets versus Steroids in the Treatment of Active Crohn’s Disease A Meta-Analysis. Scand. J. Gastroenterol. 1996, 31, 267–272. [Google Scholar] [CrossRef] [PubMed]
  36. Fernández-Bañares, F.; Cabré, E.; Esteve-Comas, M.; Gassull, M.A. How Effective Is Enteral Nutrition in Inducing Clinical Remission in Active Crohn’s Disease? A Meta-Analysis of the Randomized Clinical Trials. J. Parenter. Enter. Nutr. 1995, 19, 356–364. [Google Scholar] [CrossRef] [PubMed]
  37. Griffiths, A.M.; Ohlsson, A.; Sherman, P.M.; Sutherland, L.R. Meta-Analysis of Enteral Nutrition as a Primary Treatment of Active Crohn’s Disease. Gastroenterology 1995, 108, 1056–1067. [Google Scholar] [CrossRef] [PubMed]
  38. Stoner, P.L.; Kamel, A.; Ayoub, F.; Tan, S.; Iqbal, A.; Glover, S.C.; Zimmermann, E.M. Perioperative Care of Patients with Inflammatory Bowel Disease: Focus on Nutritional Support. Gastroenterol. Res. Pract. 2018, 2018, 7890161. [Google Scholar] [CrossRef] [PubMed]
  39. Nguyen, D.L.; Palmer, L.B.; Nguyen, E.T.; McClave, S.A.; Martindale, R.G.; Bechtold, M.L. Specialized Enteral Nutrition Therapy in Crohn’s Disease Patients on Maintenance Infliximab Therapy: A Meta-Analysis. Ther. Adv. Gastroenterol. 2015, 8, 168–175. [Google Scholar] [CrossRef] [PubMed]
  40. Yang, H.; Feng, R.; Li, T.; Xu, S.; Hao, X.; Qiu, Y.; Chen, M. Systematic Review with Meta-Analysis of Partial Enteral Nutrition for the Maintenance of Remission in Crohn’s Disease. Nutr. Res. 2020, 81, 7–18. [Google Scholar] [CrossRef]
  41. Levenstein, S.; Prantera, C.; Luzi, C.; D’Ubaldi, A. Low Residue or Normal Diet in Crohn’s Disease: A Prospective Controlled Study in Italian Patients. Gut 1985, 26, 989–993. [Google Scholar] [CrossRef]
  42. Chiba, M.; Abe, T.; Tsuda, H.; Sugawara, T.; Tsuda, S.; Tozawa, H.; Fujiwara, K.; Imai, H. Lifestyle-Related Disease in Crohn’s Disease: Relapse Prevention by a Semi-Vegetarian Diet. World J. Gastroenterol. 2010, 16, 2484–2495. [Google Scholar] [CrossRef]
  43. Chiba, M.; Nakane, K.; Tsuji, T.; Tsuda, S.; Ishii, H.; Ohno, H.; Watanabe, K.; Obara, Y.; Komatsu, M.; Sugawara, T. Relapse Prevention by Plant-Based Diet Incorporated into Induction Therapy for Ulcerative Colitis: A Single-Group Trial. Perm. J. 2019, 23, 18–220. [Google Scholar] [CrossRef] [PubMed]
  44. Fernández-Bañares, F.; Hinojosa, J.; Sánchez-Lombraña, J.L.; Navarro, E.; Martínez-Salmerón, J.F.; García-Pugés, A.; González-Huix, F.; Riera, J.; González-Lara, V.; Domínguez-Abascal, F.; et al. Randomized Clinical Trial of Plantago Ovata Seeds (Dietary Fiber) As Compared With Mesalamine in Maintaining Remission in Ulcerative Colitis. Am. J. Gastroenterol. 1999, 94, 427–433. [Google Scholar] [CrossRef] [PubMed]
  45. Sarbagili-Shabat, C.; Albenberg, L.; Van Limbergen, J.; Pressman, N.; Otley, A.; Yaakov, M.; Wine, E.; Weiner, D.; Levine, A. A Novel UC Exclusion Diet and Antibiotics for Treatment of Mild to Moderate Pediatric Ulcerative Colitis: A Prospective Open-Label Pilot Study. Nutrients 2021, 13, 3736. [Google Scholar] [CrossRef] [PubMed]
  46. Sarbagili Shabat, C.; Scaldaferri, F.; Zittan, E.; Hirsch, A.; Mentella, M.C.; Musca, T.; Cohen, N.A.; Ron, Y.; Fliss Isakov, N.; Pfeffer, J.; et al. Use of Faecal Transplantation with a Novel Diet for Mild to Moderate Active Ulcerative Colitis: The CRAFT UC Randomised Controlled Trial. J. Crohns Colitis 2022, 16, 369–378. [Google Scholar] [CrossRef] [PubMed]
  47. Yanai, H.; Levine, A.; Hirsch, A.; Boneh, R.S.; Kopylov, U.; Eran, H.B.; Cohen, N.A.; Ron, Y.; Goren, I.; Leibovitzh, H.; et al. The Crohn’s Disease Exclusion Diet for Induction and Maintenance of Remission in Adults with Mild-to-Moderate Crohn’s Disease (CDED-AD): An Open-Label, Pilot, Randomised Trial. Lancet Gastroenterol. Hepatol. 2022, 7, 49–59. [Google Scholar] [CrossRef] [PubMed]
  48. Limketkai, B.N.; Godoy-Brewer, G.; Parian, A.M.; Noorian, S.; Krishna, M.; Shah, N.D.; White, J.; Mullin, G.E. Dietary Interventions for the Treatment of Inflammatory Bowel Diseases: An Updated Systematic Review and Meta-Analysis. Clin. Gastroenterol. Hepatol. 2022, 21, 2508–2525. [Google Scholar] [CrossRef] [PubMed]
  49. Lewis, J.D.; Sandler, R.S.; Brotherton, C.; Brensinger, C.; Li, H.; Kappelman, M.D.; Daniel, S.G.; Bittinger, K.; Albenberg, L.; Valentine, J.F.; et al. A Randomized Trial Comparing the Specific Carbohydrate Diet to a Mediterranean Diet in Adults With Crohn’s Disease. Gastroenterology 2021, 161, 837–852. [Google Scholar] [CrossRef] [PubMed]
  50. Bemelman, W.A.; Warusavitarne, J.; Sampietro, G.M.; Serclova, Z.; Zmora, O.; Luglio, G.; de Buck van Overstraeten, A.; Burke, J.P.; Buskens, C.J.; Francesco, C.; et al. ECCO-ESCP Consensus on Surgery for Crohn’s Disease. J. Crohns Colitis 2018, 12, 1–16. [Google Scholar] [CrossRef]
  51. Forbes, A.; Escher, J.; Hébuterne, X.; Kłęk, S.; Krznaric, Z.; Schneider, S.; Shamir, R.; Stardelova, K.; Wierdsma, N.; Wiskin, A.E.; et al. ESPEN Guideline: Clinical Nutrition in Inflammatory Bowel Disease. Clin. Nutr. 2017, 36, 321–347. [Google Scholar] [CrossRef]
  52. Brennan, G.T.; Ha, I.; Hogan, C.; Nguyen, E.; Jamal, M.M.; Bechtold, M.L.; Nguyen, D.L. Does Preoperative Enteral or Parenteral Nutrition Reduce Postoperative Complications in Crohn’s Disease Patients: A Meta-Analysis. Eur. J. Gastroenterol. Hepatol. 2018, 30, 997–1002. [Google Scholar] [CrossRef]
  53. Grass, F.; Pache, B.; Martin, D.; Hahnloser, D.; Demartines, N.; Hübner, M. Preoperative Nutritional Conditioning of Crohn’s Patients—Systematic Review of Current Evidence and Practice. Nutrients 2017, 9, 562. [Google Scholar] [CrossRef] [PubMed]
  54. Gordon-Dixon, A.; Gore-Rodney, J.; Hampal, R.; Ross, R.; Miah, A.; Amorim Adegboye, A.R.; Grimes, C.E. The Role of Exclusive Enteral Nutrition in the Pre-Operative Optimisation of Adult Patients with Crohn’s Disease. A Systematic Review. Clin. Nutr. ESPEN 2021, 46, 99–105. [Google Scholar] [CrossRef]
  55. McLaughlin, S.D.; Culkin, A.; Cole, J.; Clark, S.K.; Tekkis, P.P.; Ciclitira, P.J.; Nicholls, R.J.; Whelan, K. Exclusive Elemental Diet Impacts on the Gastrointestinal Microbiota and Improves Symptoms in Patients with Chronic Pouchitis. J. Crohns Colitis 2013, 7, 460–466. [Google Scholar] [CrossRef]
  56. Welters, C.F.M.; Heineman, E.; Thunnissen, F.B.J.M.; van den Bogaard, A.E.J.M.; Soeters, P.B.; Baeten, C.G.M.I. Effect of Dietary Inulin Supplementation on Inflammation of Pouch Mucosa in Patients With an Ileal Pouch-Anal Anastomosis. Dis. Colon Rectum 2002, 45, 621–627. [Google Scholar] [CrossRef] [PubMed]
  57. Ianco, O. Diet of Patients after Pouch Surgery May Affect Pouch Inflammation. World J. Gastroenterol. 2013, 19, 6458. [Google Scholar] [CrossRef] [PubMed]
  58. Godny, L.; Maharshak, N.; Reshef, L.; Goren, I.; Yahav, L.; Fliss-Isakov, N.; Gophna, U.; Tulchinsky, H.; Dotan, I. Fruit Consumption Is Associated with Alterations in Microbial Composition and Lower Rates of Pouchitis. J. Crohns Colitis 2019, 13, 1265–1272. [Google Scholar] [CrossRef] [PubMed]
  59. Pironi, L.; Arends, J.; Bozzetti, F.; Cuerda, C.; Gillanders, L.; Jeppesen, P.B.; Joly, F.; Kelly, D.; Lal, S.; Staun, M.; et al. ESPEN Guidelines on Chronic Intestinal Failure in Adults. Clin. Nutr. 2016, 35, 247–307. [Google Scholar] [CrossRef] [PubMed]
  60. Nightingale, J. Guidelines for Management of Patients with a Short Bowel. Gut 2006, 55, iv1–iv12. [Google Scholar] [CrossRef]
  61. Woolf, G.M.; Miller, C.; Kurian, R.; Jeejeebhoy, K.N. Diet for Patients with a Short Bowel: High Fat or High Carbohydrate? Gastroenterology 1983, 84, 823–828. [Google Scholar] [CrossRef]
  62. Nordgaard, I.; Hansen, B.S.; Mortensen, P.B. Colon as a Digestive Organ in Patients with Short Bowel. Lancet 1994, 343, 373–376. [Google Scholar] [CrossRef]
  63. Zhou, Y.; Wu, X.T.; Yang, G.; Zhuang, W.; Wei, M. Clinical Evidence of Growth Hormone, Glutamine and a Modified Diet for Short Bowel Syndrome: Meta-Analysis of Clinical Trials. Asia Pac. J. Clin. Nutr. 2005, 14, 98–102. [Google Scholar] [PubMed]
  64. Rosenberg, I.H. Sarcopenia: Origins and Clinical Relevance. J. Nutr. 1997, 127, 990S–991S. [Google Scholar] [CrossRef] [PubMed]
  65. Cruz-Jentoft, A.J.; Baeyens, J.P.; Bauer, J.M.; Boirie, Y.; Cederholm, T.; Landi, F.; Martin, F.C.; Michel, J.P.; Rolland, Y.; Schneider, S.M.; et al. Sarcopenia: European Consensus on Definition and Diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing 2010, 39, 412–423. [Google Scholar] [CrossRef] [PubMed]
  66. Zhou, Z.; Xiong, Z.; Xie, Q.; Xiao, P.; Zhang, Q.; Gu, J.; Li, J.; Hu, D.; Hu, X.; Shen, Y.; et al. Computed Tomography-Based Multiple Body Composition Parameters Predict Outcomes in Crohn’s Disease. Insights Imaging 2021, 12, 135. [Google Scholar] [CrossRef] [PubMed]
  67. Cravo, M.L.; Velho, S.; Torres, J.; Costa Santos, M.P.; Palmela, C.; Cruz, R.; Strecht, J.; Maio, R.; Baracos, V. Lower Skeletal Muscle Attenuation and High Visceral Fat Index Are Associated with Complicated Disease in Patients with Crohn’s Disease: An Exploratory Study. Clin. Nutr. ESPEN 2017, 21, 79–85. [Google Scholar] [CrossRef] [PubMed]
  68. Labarthe, G.; Dolores, M.; Verdalle-Cazes, M.; Charpentier, C.; Roullee, P.; Dacher, J.N.; Savoye, G.; Savoye-Collet, C. Magnetic Resonance Imaging Assessment of Body Composition Parameters in Crohn’s Disease. Dig. Liver Dis. 2020, 52, 878–884. [Google Scholar] [CrossRef] [PubMed]
  69. Schneider, S.M.; Al-Jaouni, R.; Filippi, J.; Wiroth, J.B.; Zeanandin, G.; Arab, K.; Hébuterne, X. Sarcopenia Is Prevalent in Patients with Crohn’s Disease in Clinical Remission. Inflamm. Bowel Dis. 2008, 14, 1562–1568. [Google Scholar] [CrossRef]
  70. Holt, D.Q.; Moore, G.T.; Strauss, B.J.G.; Hamilton, A.L.; De Cruz, P.; Kamm, M.A. Visceral Adiposity Predicts Post-Operative Crohn’s Disease Recurrence. Aliment. Pharmacol. Ther. 2017, 45, 1255–1264. [Google Scholar] [CrossRef]
  71. Lee, C.H.; Yoon, H.; Oh, D.J.; Lee, J.M.; Choi, Y.J.; Shin, C.M.; Park, Y.S.; Kim, N.; Lee, D.H.; Kim, J.S. The Prevalence of Sarcopenia and Its Effect on Prognosis in Patients with Crohn’s Disease. Intest. Res. 2020, 18, 79–84. [Google Scholar] [CrossRef]
  72. Zhang, C.; Yu, D.; Hong, L.; Zhang, T.; Liu, H.; Fan, R.; Wang, L.; Zhong, J.; Wang, Z. Prevalence of Sarcopenia and Its Effect on Postoperative Complications in Patients with Crohn’s Disease. Gastroenterol. Res. Pract. 2021, 2021, 3267201. [Google Scholar] [CrossRef]
  73. Grova, M.; Crispino, F.; Maida, M.; Renna, S.; Casà, A.; Tesè, L.; Macaluso, F.S.; Orlando, A. Sarcopenia Is a Poor Prognostic Factor for Endoscopic Remission in Patients with Crohn’s Disease. In Proceedings of the XIII Congresso Nazionale IG-IBD, Riccione, Italy, 1–3 December 2022; pp. S84–S85. [Google Scholar]
  74. Hong, J.T.; James, S.; Tran, A.; Kutaiba, N. Sarcopenia Measurements and Clinical Outcomes in Crohn’s Disease Surgical Patients. ANZ J. Surg. 2022, 92, 3209–3213. [Google Scholar] [CrossRef] [PubMed]
  75. Boparai, G.; Kedia, S.; Kandasamy, D.; Sharma, R.; Madhusudhan, K.S.; Dash, N.R.; Sahu, P.; Pal, S.; Sahni, P.; Panwar, R.; et al. Combination of Sarcopenia and High Visceral Fat Predict Poor Outcomes in Patients with Crohn’s Disease. Eur. J. Clin. Nutr. 2021, 75, 1491–1498. [Google Scholar] [CrossRef] [PubMed]
  76. Erős, A.; Soós, A.; Hegyi, P.; Szakács, Z.; Benke, M.; Szűcs, Á.; Hartmann, P.; Erőss, B.; Sarlós, P. Sarcopenia as an Independent Predictor of the Surgical Outcomes of Patients with Inflammatory Bowel Disease: A Meta-Analysis. Surg. Today 2020, 50, 1138–1150. [Google Scholar] [CrossRef] [PubMed]
  77. Liu, S.; Ding, X.; Maggiore, G.; Pietrobattista, A.; Satapathy, S.K.; Tian, Z.; Jing, X. Sarcopenia Is Associated with Poor Clinical Outcomes in Patients with Inflammatory Bowel Disease: A Prospective Cohort Study. Ann. Transl. Med. 2022, 10, 367. [Google Scholar] [CrossRef] [PubMed]
  78. Campbell, J.P.; Teigen, L.; Manski, S.; Blumhof, B.; Guglielmo, F.F.; Shivashankar, R.; Shmidt, E. Sarcopenia Is More Prevalent Among Inflammatory Bowel Disease Patients Undergoing Surgery and Predicts Progression to Surgery Among Medically Treated Patients. Inflamm. Bowel Dis. 2022, 28, 1844–1850. [Google Scholar] [CrossRef] [PubMed]
  79. Grillot, J.; D’Engremont, C.; Parmentier, A.L.; Lakkis, Z.; Piton, G.; Cazaux, D.; Gay, C.; De Billy, M.; Koch, S.; Borot, S.; et al. Sarcopenia and Visceral Obesity Assessed by Computed Tomography Are Associated with Adverse Outcomes in Patients with Crohn’s Disease. Clin. Nutr. 2020, 39, 3024–3030. [Google Scholar] [CrossRef] [PubMed]
  80. Bamba, S.; Sasaki, M.; Takaoka, A.; Takahashi, K.; Imaeda, H.; Nishida, A.; Inatomi, O.; Sugimoto, M.; Andoh, A. Sarcopenia Is a Predictive Factor for Intestinal Resection in Admitted Patients with Crohn’s Disease. PLoS ONE 2017, 12, e0180036. [Google Scholar] [CrossRef] [PubMed]
  81. Spooren, C.E.G.M.; Lodewick, T.M.; Beelen, E.M.J.; Dijk, D.P.J.; Bours, M.J.L.; Haans, J.J.; Masclee, A.A.M.; Pierik, M.J.; Bakers, F.C.H.; Jonkers, D.M.A.E. The Reproducibility of Skeletal Muscle Signal Intensity on Routine Magnetic Resonance Imaging in Crohn’s Disease. J. Gastroenterol. Hepatol. 2020, 35, 1902–1908. [Google Scholar] [CrossRef]
  82. Nardone, O.M.; Ponsiglione, A.; de Sire, R.; Calabrese, G.; Liuzzi, R.; Testa, A.; Guarino, A.D.; Olmo, O.; Rispo, A.; Camera, L.; et al. Impact of Sarcopenia on Clinical Outcomes in a Cohort of Caucasian Active Crohn’s Disease Patients Undergoing Multidetector CT-Enterography. Nutrients 2022, 14, 3460. [Google Scholar] [CrossRef]
  83. Thiberge, C.; Charpentier, C.; Gillibert, A.; Modzelewski, R.; Dacher, J.N.; Savoye, G.; Savoye-Collet, C. Lower Subcutaneous or Visceral Adiposity Assessed by Abdominal Computed Tomography Could Predict Adverse Outcome in Patients With Crohn’s Disease. J. Crohns Colitis 2018, 12, 1429–1437. [Google Scholar] [CrossRef]
  84. Carvalho, D.; Viana, C.; Marques, I.; Costa, C.; Martins, S.F. Sarcopenia Is Associated with Postoperative Outcome in Patients with Crohn’s Disease Undergoing Bowel Resection. Gastrointest. Disord. 2019, 1, 201–209. [Google Scholar] [CrossRef]
  85. Ge, X.; Xia, J.; Wu, Y.; Ye, L.; Liu, W.; Qi, W.; Cao, Q.; Bai, R.; Zhou, W. Sarcopenia Assessed by Computed Tomography Is Associated with Colectomy in Patients with Acute Severe Ulcerative Colitis. Eur. J. Clin. Nutr. 2022, 76, 410–418. [Google Scholar] [CrossRef] [PubMed]
  86. Cushing, K.C.; Kordbacheh, H.; Gee, M.S.; Kambadakone, A.; Ananthakrishnan, A.N. Sarcopenia Is a Novel Predictor of the Need for Rescue Therapy in Hospitalized Ulcerative Colitis Patients. J. Crohns Colitis 2018, 12, 1036–1041. [Google Scholar] [CrossRef] [PubMed]
  87. Ge, X.; Jiang, L.; Yu, W.; Wu, Y.; Liu, W.; Qi, W.; Cao, Q.; Bai, R.; Zhou, W. The Importance of Sarcopenia as a Prognostic Predictor of the Clinical Course in Acute Severe Ulcerative Colitis Patients. Dig. Liver Dis. 2021, 53, 965–971. [Google Scholar] [CrossRef] [PubMed]
  88. Berger, M.; Yamada, A.; Komaki, Y.; Komaki, F.; Cohen, R.D.; Dalal, S.; Hurst, R.D.; Hyman, N.; Pekow, J.; Shogan, B.D.; et al. Low Skeletal Muscle Index Adjusted for Body Mass Index Is an Independent Risk Factor for Inflammatory Bowel Disease Surgical Complications. Crohns Colitis 360 2020, 2, otaa064. [Google Scholar] [CrossRef] [PubMed]
  89. Pedersen, M.; Cromwell, J.; Nau, P. Sarcopenia Is a Predictor of Surgical Morbidity in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2017, 23, 1867–1872. [Google Scholar] [CrossRef] [PubMed]
  90. Trinder, M.W.; Clifford, M.; Jones, A.L.; Shepherd, T.; Jacob, A.O. The Impact of Sarcopenia on Outcomes in Patients with Inflammatory Bowel Disease Undergoing Colorectal Surgery. ANZ J. Surg. 2022, 92, 397–402. [Google Scholar] [CrossRef] [PubMed]
  91. Alipour, O.; Lee, V.; Tejura, T.K.; Wilson, M.L.; Memel, Z.; Cho, J.; Cologne, K.; Hwang, C.; Shao, L. The Assessment of Sarcopenia Using Psoas Muscle Thickness per Height Is Not Predictive of Post-Operative Complications in IBD. Scand. J. Gastroenterol. 2021, 56, 1175–1181. [Google Scholar] [CrossRef]
  92. Zhang, T.; Cao, L.; Cao, T.; Yang, J.; Gong, J.; Zhu, W.; Li, N.; Li, J. Prevalence of Sarcopenia and Its Impact on Postoperative Outcome in Patients with Crohn’s Disease Undergoing Bowel Resection. J. Parenter. Enter. Nutr. 2017, 41, 592–600. [Google Scholar] [CrossRef]
  93. Zager, Y.; Khalilieh, S.; Ganaiem, O.; Gorgov, E.; Horesh, N.; Anteby, R.; Kopylov, U.; Jacoby, H.; Dreznik, Y.; Dori, A.; et al. Low Psoas Muscle Area Is Associated with Postoperative Complications in Crohn’s Disease. Int. J. Color. Dis. 2021, 36, 543–550. [Google Scholar] [CrossRef]
  94. Galata, C.; Hodapp, J.; Weiß, C.; Karampinis, I.; Vassilev, G.; Reißfelder, C.; Otto, M. Skeletal Muscle Mass Index Predicts Postoperative Complications in Intestinal Surgery for Crohn’s Disease. J. Parenter. Enter. Nutr. 2020, 44, 714–721. [Google Scholar] [CrossRef] [PubMed]
  95. Celentano, V.; Kamil-Mustafa, L.; Beable, R.; Ball, C.; Flashman, K.G.; Jennings, Z.; O’ Leary, D.P.; Higginson, A.; Luxton, S. Preoperative Assessment of Skeletal Muscle Mass during Magnetic Resonance Enterography in Patients with Crohn’s Disease. Updat. Surg. 2021, 73, 1419–1427. [Google Scholar] [CrossRef] [PubMed]
  96. Fujikawa, H.; Araki, T.; Okita, Y.; Kondo, S.; Kawamura, M.; Hiro, J.; Toiyama, Y.; Kobayashi, M.; Tanaka, K.; Inoue, Y.; et al. Impact of Sarcopenia on Surgical Site Infection after Restorative Proctocolectomy for Ulcerative Colitis. Surg. Today 2017, 47, 92–98. [Google Scholar] [CrossRef] [PubMed]
  97. Harper, J.W.; Zisman, T.L. Interaction of Obesity and Inflammatory Bowel Disease. World J. Gastroenterol. 2016, 22, 7868. [Google Scholar] [CrossRef] [PubMed]
  98. Moran, G.W.; Dubeau, M.F.; Kaplan, G.G.; Panaccione, R.; Ghosh, S. The Increasing Weight of Crohn’s Disease Subjects in Clinical Trials. Inflamm. Bowel Dis. 2013, 19, 2949–2956. [Google Scholar] [CrossRef] [PubMed]
  99. Boutros, M.; Maron, D. Inflammatory Bowel Disease in the Obese Patient. Clin. Colon Rectal Surg. 2011, 24, 244–252. [Google Scholar] [CrossRef] [PubMed]
  100. Johnson, A.; Loftus, E. Obesity in Inflammatory Bowel Disease: A Review of Its Role in the Pathogenesis, Natural History, and Treatment of IBD. Saudi J. Gastroenterol. 2021, 27, 183. [Google Scholar] [CrossRef] [PubMed]
  101. Connelly, T.M.; Juza, R.M.; Sangster, W.; Sehgal, R.; Tappouni, R.F.; Messaris, E. Volumetric Fat Ratio and Not Body Mass Index Is Predictive of Ileocolectomy Outcomes in Crohn’s Disease Patients. Dig. Surg. 2014, 31, 219–224. [Google Scholar] [CrossRef]
  102. Gonçalves, P.; Magro, F.; Martel, F. Metabolic Inflammation in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2015, 21, 453–467. [Google Scholar] [CrossRef]
  103. Swanson, S.M.; Harper, J.; Zisman, T.L. Obesity and Inflammatory Bowel Disease. Curr. Opin. Gastroenterol. 2018, 34, 112–119. [Google Scholar] [CrossRef]
  104. Seminerio, J.L.; Koutroubakis, I.E.; Ramos-Rivers, C.; Hashash, J.G.; Dudekula, A.; Regueiro, M.; Baidoo, L.; Barrie, A.; Swoger, J.; Schwartz, M.; et al. Impact of Obesity on the Management and Clinical Course of Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2015, 21, 2857–2863. [Google Scholar] [CrossRef] [PubMed]
  105. Juillerat, P.; Sokol, H.; Froehlich, F.; Yajnik, V.; Beaugerie, L.; Lucci, M.; Burnand, B.; Macpherson, A.J.; Cosnes, J.; Korzenik, J.R. Factors Associated with Durable Response to Infliximab in Crohn’s Disease 5 Years and Beyond. Inflamm. Bowel Dis. 2015, 21, 60–70. [Google Scholar] [CrossRef] [PubMed]
  106. Singh, S.; Proudfoot, J.; Xu, R.; Sandborn, W.J. Obesity and Response to Infliximab in Patients with Inflammatory Bowel Diseases: Pooled Analysis of Individual Participant Data from Clinical Trials. Am. J. Gastroenterol. 2018, 113, 883–889. [Google Scholar] [CrossRef] [PubMed]
  107. Rowan, C.R.; McManus, J.; Boland, K.; O’Toole, A. Visceral Adiposity and Inflammatory Bowel Disease. Int. J. Color. Dis. 2021, 36, 2305–2319. [Google Scholar] [CrossRef] [PubMed]
  108. Hu, Q.; Ren, J.; Li, G.; Wu, X.; Li, J. The Impact of Obesity on the Clinical Course of Inflammatory Bowel Disease: A Meta-Analysis. Med. Sci. Monit. 2017, 23, 2599–2606. [Google Scholar] [CrossRef] [PubMed]
  109. Pringle, P.L.; Stewart, K.O.; Peloquin, J.M.; Sturgeon, H.C.; Nguyen, D.; Sauk, J.; Garber, J.J.; Yajnik, V.; Ananthakrishnan, A.N.; Chan, A.T.; et al. Body Mass Index, Genetic Susceptibility, and Risk of Complications Among Individuals with Crohn’s Disease. Inflamm. Bowel Dis. 2015, 21, 2304–2310. [Google Scholar] [CrossRef] [PubMed]
  110. Hass, D.; Brensinger, C.; LEWIS, J.; Lichtenstein, G. The Impact of Increased Body Mass Index on the Clinical Course of Crohn’s Disease. Clin. Gastroenterol. Hepatol. 2006, 4, 482–488. [Google Scholar] [CrossRef] [PubMed]
  111. Van Der Sloot, K.W.J.; Joshi, A.D.; Bellavance, D.R.; Gilpin, K.K.; Stewart, K.O.; Lochhead, P.; Garber, J.J.; Giallourakis, C.; Yajnik, V.; Ananthakrishnan, A.N.; et al. Visceral Adiposity, Genetic Susceptibility, and Risk of Complications Among Individuals with Crohn’s Disease. Inflamm. Bowel Dis. 2017, 23, 82–88. [Google Scholar] [CrossRef]
  112. Jiang, K.; Chen, B.; Lou, D.; Zhang, M.; Shi, Y.; Dai, W.; Shen, J.; Zhou, B.; Hu, J. Systematic Review and Meta-Analysis: Association between Obesity/Overweight and Surgical Complications in IBD. Int. J. Color. Dis. 2022, 37, 1485–1496. [Google Scholar] [CrossRef]
  113. Hicks, G.; Abdulaal, A.; Slesser, A.A.P.; Mohsen, Y. Outcomes of Inflammatory Bowel Disease Surgery in Obese versus Non-Obese Patients: A Meta-Analysis. Tech. Coloproctology 2019, 23, 947–955. [Google Scholar] [CrossRef]
  114. Emile, S.H.; Khan, S.M.; Wexner, S.D. A Systematic Review and Meta-Analysis of the Outcome of Ileal Pouch Anal Anastomosis in Patients with Obesity. Surgery 2021, 170, 1629–1636. [Google Scholar] [CrossRef]
  115. Causey, M.W.; Johnson, E.K.; Miller, S.; Martin, M.; Maykel, J.; Steele, S.R. The Impact of Obesity on Outcomes Following Major Surgery for Crohn’s Disease: An American College of Surgeons National Surgical Quality Improvement Program Assessment. Dis. Colon Rectum 2011, 54, 1488–1495. [Google Scholar] [CrossRef]
  116. Gao, X.H.; Yu, G.Y.; Khan, F.; Li, J.Q.; Stocchi, L.; Hull, T.L.; Shen, B. Greater Peripouch Fat Area on CT Image Is Associated with Chronic Pouchitis and Pouch Failure in Inflammatory Bowel Diseases Patients. Dig. Dis. Sci. 2020, 65, 3660–3671. [Google Scholar] [CrossRef]
  117. Wei, Y.; Zhu, F.; Gong, J.; Yang, J.; Zhang, T.; Gu, L.; Zhu, W.; Guo, Z.; Li, Y.; Li, N.; et al. High Visceral to Subcutaneous Fat Ratio Is Associated with Increased Postoperative Inflammatory Response after Colorectal Resection in Inflammatory Bowel Disease. Gastroenterol. Res. Pract. 2018, 2018, 6270514. [Google Scholar] [CrossRef]
  118. Schoenefuss, F.; Hoffmann, P. Serum γ-Globulin and Albumin Concentrations Predict Secondary Loss of Response to Anti-TNFα in Inflammatory Bowel Disease Patients. Eur. J. Gastroenterol. Hepatol. 2019, 31, 1563–1568. [Google Scholar] [CrossRef]
  119. Tighe, D.; Hall, B.; Jeyarajah, S.K.; Smith, S.; Breslin, N.; Ryan, B.; McNamara, D. One-Year Clinical Outcomes in an IBD Cohort Who Have Previously Had Anti-TNFa Trough and Antibody Levels Assessed. Inflamm. Bowel Dis. 2017, 23, 1154–1159. [Google Scholar] [CrossRef]
  120. Dotan, I.; Ron, Y.; Yanai, H.; Becker, S.; Fishman, S.; Yahav, L.; Ben Yehoyada, M.; Mould, D.R. Patient Factors That Increase Infliximab Clearance and Shorten Half-Life in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2014, 20, 2247–2259. [Google Scholar] [CrossRef]
  121. Berends, S.E.; Strik, A.S.; Jansen, J.M.; de Boer, N.K.; van Egmond, P.S.; Brandse, J.F.; Mathôt, R.A.; D’Haens, G.R.; Löwenberg, M. Pharmacokinetics of Golimumab in Moderate to Severe Ulcerative Colitis: The GO-KINETIC Study. Scand. J. Gastroenterol. 2019, 54, 700–706. [Google Scholar] [CrossRef]
  122. Verma, A.; Varma, S.; Freedberg, D.E.; Axelrad, J.E. A Simple Emergency Department-Based Score Predicts Complex Hospitalization in Patients with Inflammatory Bowel Disease. Dig. Dis. Sci. 2022, 67, 629–638. [Google Scholar] [CrossRef]
  123. Suzuki, Y.; Matsui, T.; Ito, H.; Ashida, T.; Nakamura, S.; Motoya, S.; Matsumoto, T.; Sato, N.; Ozaki, K.; Watanabe, M.; et al. Circulating Interleukin 6 and Albumin, and Infliximab Levels Are Good Predictors of Recovering Efficacy After Dose Escalation Infliximab Therapy in Patients with Loss of Response to Treatment for Crohn’s Disease. Inflamm. Bowel Dis. 2015, 21, 2114–2122. [Google Scholar] [CrossRef]
  124. Steinhardt, H.J.; Loeschke, K.; Kasper, H.; Holtermüller, K.H.; Schäfer, H. European Cooperative Crohn’s Disease Study (ECCDS): Clinical Features and Natural History. Digestion 1985, 31, 97–108. [Google Scholar] [CrossRef]
  125. Ghaly, S.; Murray, K.; Baird, A.; Martin, K.; Prosser, R.; Mill, J.; Simms, L.A.; Hart, P.H.; Radford-Smith, G.; Bampton, P.A.; et al. High Vitamin D-Binding Protein Concentration, Low Albumin, and Mode of Remission Predict Relapse in Crohn’s Disease. Inflamm. Bowel Dis. 2016, 22, 2456–2464. [Google Scholar] [CrossRef]
  126. Moroi, R.; Endo, K.; Yamamoto, K.; Naito, T.; Onodera, M.; Kuroha, M.; Kanazawa, Y.; Kimura, T.; Kakuta, Y.; Masamune, A.; et al. Long-Term Prognosis of Japanese Patients with Biologic-Naïve Crohn’s Disease Treated with Anti-Tumor Necrosis Factor-α Antibodies. Intest. Res. 2019, 17, 94–106. [Google Scholar] [CrossRef]
  127. Imaeda, H.; Takahashi, K.; Fujimoto, T.; Bamba, S.; Tsujikawa, T.; Sasaki, M.; Fujiyama, Y.; Andoh, A. Clinical Utility of Newly Developed Immunoassays for Serum Concentrations of Adalimumab and Anti-Adalimumab Antibodies in Patients with Crohn’s Disease. J. Gastroenterol. 2014, 49, 100–109. [Google Scholar] [CrossRef]
  128. Ravindran, P.; Ansari, N.; Young, C.J.; Solomon, M.J. Definitive Surgical Closure of Enterocutaneous Fistula: Outcome and Factors Predictive of Increased Postoperative Morbidity. Color. Dis. 2014, 16, 209–218. [Google Scholar] [CrossRef]
  129. Yoneno, K.; Hisamatsu, T.; Matsuoka, K.; Okamoto, S.; Takayama, T.; Ichikawa, R.; Sujino, T.; Miyoshi, J.; Takabayashi, K.; Mikami, Y.; et al. Risk and Management of Intra-Abdominal Abscess in Crohn’s Disease Treated with Infliximab. Digestion 2014, 89, 201–208. [Google Scholar] [CrossRef]
  130. Lok, K.H.; Ng, C.H.; Hung, H.G.; Li, K.F.; Li, K.K.; Szeto, M.L. Correlation of Serum Biomarkers with Clinical Severity and Mucosal Inflammation in Chinese Ulcerative Colitis Patients. J. Dig. Dis. 2008, 9, 219–224. [Google Scholar] [CrossRef]
  131. Khan, N.; Patel, D.; Shah, Y.; Trivedi, C.; Yang, Y.X. Albumin as a Prognostic Marker for Ulcerative Colitis. World J. Gastroenterol. 2017, 23, 8008–8016. [Google Scholar] [CrossRef]
  132. Lee, S.H.; Walshe, M.; Oh, E.H.; Hwang, S.W.; Park, S.H.; Yang, D.H.; Byeon, J.S.; Myung, S.J.; Yang, S.K.; Greener, T.; et al. Early Changes in Serum Albumin Predict Clinical and Endoscopic Outcomes in Patients With Ulcerative Colitis Starting Anti-TNF Treatment. Inflamm. Bowel Dis. 2021, 27, 1452–1461. [Google Scholar] [CrossRef]
  133. Langholz, E.; Munkholm, P.; Davidsen, M.; Nielsen, O.H.; Binder, V. Changes in Extent of Ulcerative Colitis A Study on the Course and Prognostic Factors. Scand. J. Gastroenterol. 1996, 31, 260–266. [Google Scholar] [CrossRef]
  134. Kumar, S.; Ghoshal, U.C.; Aggarwal, R.; Saraswat, V.A.; Choudhuri, G. Severe Ulcerative Colitis: Prospective Study of Parameters Determining Outcome. J. Gastroenterol. Hepatol. Aust. 2004, 19, 1247–1252. [Google Scholar] [CrossRef] [PubMed]
  135. Arias, M.T.; Vande Casteele, N.; Vermeire, S.; de Buck van Overstraeten, A.; Billiet, T.; Baert, F.; Wolthuis, A.; Van Assche, G.; Noman, M.; Hoffman, I.; et al. A Panel to Predict Long-Term Outcome of Infliximab Therapy for Patients with Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 2015, 13, 531–538. [Google Scholar] [CrossRef] [PubMed]
  136. Le Baut, G.; Kirchgesner, J.; Amiot, A.; Lefevre, J.H.; Chafai, N.; Landman, C.; Nion, I.; Bourrier, A.; Delattre, C.; Martineau, C.; et al. A Scoring System to Determine Patients’ Risk of Colectomy Within 1 Year After Hospital Admission for Acute Severe Ulcerative Colitis. Clin. Gastroenterol. Hepatol. 2021, 19, 1602–1610. [Google Scholar] [CrossRef] [PubMed]
  137. Tanaka, M.; Takagi, T.; Naito, Y.; Uchiyama, K.; Hotta, Y.; Toyokawa, Y.; Kashiwagi, S.; Kamada, K.; Ishikawa, T.; Yasuda, H.; et al. Low Serum Albumin at Admission Is a Predictor of Early Colectomy in Patients with Moderate to Severe Ulcerative Colitis. JGH Open 2021, 5, 377–381. [Google Scholar] [CrossRef] [PubMed]
  138. Con, D.; Andrew, B.; Nicolaides, S.; van Langenberg, D.R.; Vasudevan, A. Biomarker Dynamics during Infliximab Salvage for Acute Severe Ulcerative Colitis: C-Reactive Protein (CRP)-Lymphocyte Ratio and CRP-Albumin Ratio Are Useful in Predicting Colectomy. Intest. Res. 2022, 20, 101–113. [Google Scholar] [CrossRef] [PubMed]
  139. Gibson, D.J.; Hartery, K.; Doherty, J.; Nolan, J.; Keegan, D.; Byrne, K.; Martin, S.T.; Buckley, M.; Sheridan, J.; Horgan, G.; et al. CRP/Albumin Ratio: An Early Predictor of Steroid Responsiveness in Acute Severe Ulcerative Colitis. J. Clin. Gastroenterol. 2018, 52, e48–e52. [Google Scholar] [CrossRef] [PubMed]
  140. Choy, M.C.; Seah, D.; Gorelik, A.; An, Y.K.; Chen, C.Y.; Macrae, F.A.; Sparrow, M.P.; Connell, W.R.; Moore, G.T.; Radford-Smith, G.; et al. Predicting Response after Infliximab Salvage in Acute Severe Ulcerative Colitis. J. Gastroenterol. Hepatol. 2018, 33, 1347–1352. [Google Scholar] [CrossRef] [PubMed]
  141. Stidham, R.W.; Guentner, A.S.; Ruma, J.L.; Govani, S.M.; Waljee, A.K.; Higgins, P.D.R. Intestinal Dilation and Platelet:Albumin Ratio Are Predictors of Surgery in Stricturing Small Bowel Crohn’s Disease. Clin. Gastroenterol. Hepatol. 2016, 14, 1112–1119. [Google Scholar] [CrossRef]
  142. Syal, G.; Robbins, L.; Kashani, A.; Bonthala, N.; Feldman, E.; Fleshner, P.; Vasiliauskas, E.; McGovern, D.; Ha, C.; Targan, S.; et al. Hypoalbuminemia and Bandemia Predict Failure of Infliximab Rescue Therapy in Acute Severe Ulcerative Colitis. Dig. Dis. Sci. 2021, 66, 199–205. [Google Scholar] [CrossRef]
  143. Ishida, N.; Miyazu, T.; Tamura, S.; Tani, S.; Yamade, M.; Iwaizumi, M.; Hamaya, Y.; Osawa, S.; Furuta, T.; Sugimoto, K. Early Serum Albumin Changes in Patients with Ulcerative Colitis Treated with Tacrolimus Will Predict Clinical Outcome. World J. Gastroenterol. 2021, 27, 3109–3120. [Google Scholar] [CrossRef]
  144. Ishida, N.; Tani, S.; Asai, Y.; Miyazu, T.; Tamura, S.; Yamade, M.; Iwaizumi, M.; Hamaya, Y.; Osawa, S.; Furuta, T.; et al. Lymphocyte to Monocyte Ratio and Serum Albumin Changes Predict Tacrolimus Therapy Outcomes in Patients with Ulcerative Colitis. Sci. Rep. 2022, 12, 13572. [Google Scholar] [CrossRef]
  145. Yamamoto, T.; Shimoyama, T.; Umegae, S.; Kotze, P.G. Impact of Preoperative Nutritional Status on the Incidence Rate of Surgical Complications in Patients with Inflammatory Bowel Disease With Vs Without Preoperative Biologic Therapy: A Case-Control Study. Clin. Transl. Gastroenterol. 2019, 10, e00050. [Google Scholar] [CrossRef]
  146. Saha, A.K.; Tapping, C.R.; Foley, G.T.; Baker, R.P.; Sagar, P.M.; Burke, D.A.; Sue-Ling, H.M.; Finan, P.J. Morbidity and Mortality after Closure of Loop Ileostomy. Color. Dis. 2009, 11, 866–871. [Google Scholar] [CrossRef]
  147. Telem, D.A. Risk Factors for Anastomotic Leak Following Colorectal Surgery. Arch. Surg. 2010, 145, 371. [Google Scholar] [CrossRef]
  148. Dai, X.; Ge, X.; Yang, J.; Zhang, T.; Xie, T.; Gao, W.; Gong, J.; Zhu, W. Increased Incidence of Prolonged Ileus after Colectomy for Inflammatory Bowel Diseases under ERAS Protocol: A Cohort Analysis. J. Surg. Res. 2017, 212, 86–93. [Google Scholar] [CrossRef]
  149. Heimann, T.M.; Swaminathan, S.; Greenstein, A.J.; Steinhagen, R.M. Incidence and Factors Correlating With Incisional Hernia Following Open Bowel Resection in Patients With Inflammatory Bowel Disease. Ann. Surg. 2018, 267, 532–536. [Google Scholar] [CrossRef]
  150. Almogy, G.; Bodian, C.A.; Greenstein, A.J. Surgery for Late-Onset Ulcerative Colitis: Predictors of Short-Term Outcome. Scand. J. Gastroenterol. 2002, 37, 1025–1028. [Google Scholar] [CrossRef]
  151. Gu, J.; Stocchi, L.; Remzi, F.; Kiran, R.P. Factors Associated with Postoperative Morbidity, Reoperation and Readmission Rates after Laparoscopic Total Abdominal Colectomy for Ulcerative Colitis. Color. Dis. 2013, 15, 1123–1129. [Google Scholar] [CrossRef]
  152. Gu, J.; Stocchi, L.; Gorgun, E.; Remzi, F.H. Risk Factors Associated with Portomesenteric Venous Thrombosis in Patients Undergoing Restorative Proctocolectomy for Medically Refractory Ulcerative Colitis. Color. Dis. 2016, 18, 393–399. [Google Scholar] [CrossRef]
  153. Nisar, P.J.; Appau, K.A.; Remzi, F.H.; Kiran, R.P. Preoperative Hypoalbuminemia Is Associated with Adverse Outcomes after Ileoanal Pouch Surgery. Inflamm. Bowel Dis. 2012, 18, 1034–1041. [Google Scholar] [CrossRef]
  154. Li, J.; Lyu, H.; Yang, H.; Li, Y.; Tan, B.; Wei, M.M.; Sun, X.Y.; Li, J.N.; Wu, B.; Qian, J.M. Preoperative Corticosteroid Usage and Hypoalbuminemia Increase Occurrence of Short-Term Postoperative Complications in Chinese Patients with Ulcerative Colitis. Chin. Med. J. 2016, 129, 435–441. [Google Scholar] [CrossRef]
  155. Sofo, L.; Caprino, P.; Schena, C.A.; Sacchetti, F.; Potenza, A.E.; Ciociola, A. New Perspectives in the Prediction of Postoperative Complications for High-Risk Ulcerative Colitis Patients: Machine Learning Preliminary Approach. Eur. Rev. Med. Pharmacol. Sci. 2020, 24, 12781–12787. [Google Scholar] [CrossRef]
  156. El-Hussuna, A.; Iesalnieks, I.; Horesh, N.; Hadi, S.; Dreznik, Y.; Zmora, O. The Effect of Pre-Operative Optimization on Post-Operative Outcome in Crohn’s Disease Resections. Int. J. Color. Dis. 2017, 32, 49–56. [Google Scholar] [CrossRef]
  157. Galata, C.; Kienle, P.; Weiss, C.; Seyfried, S.; Reißfelder, C.; Hardt, J. Risk Factors for Early Postoperative Complications in Patients with Crohn’s Disease after Colorectal Surgery Other than Ileocecal Resection or Right Hemicolectomy. Int. J. Color. Dis. 2019, 34, 293–300. [Google Scholar] [CrossRef]
  158. Riss, S.; Bittermann, C.; Zandl, S.; Kristo, I.; Stift, A.; Papay, P.; Vogelsang, H.; Mittlböck, M.; Herbst, F. Short-Term Complications of Wide-Lumen Stapled Anastomosis after Ileocolic Resection for Crohn’s Disease: Who Is at Risk? Color. Dis. 2010, 12, e298–e303. [Google Scholar] [CrossRef]
  159. Sacchetti, F.; Caprino, P.; Potenza, A.E.; Pastena, D.; Presacco, S.; Sofo, L. Early and Late Outcomes of a Series of 255 Patients with Crohn’s Disease Who Underwent Resection: 10 Years of Experience at a Single Referral Center. Updat. Surg. 2022, 74, 1657–1664. [Google Scholar] [CrossRef]
  160. Ghoneima, A.S.; Flashman, K.; Dawe, V.; Baldwin, E.; Celentano, V. High Risk of Septic Complications Following Surgery for Crohn’s Disease in Patients with Preoperative Anaemia, Hypoalbuminemia and High CRP. Int. J. Color. Dis. 2019, 34, 2185–2188. [Google Scholar] [CrossRef]
  161. Liu, X.; Wu, X.; Zhou, C.; Hu, T.; Ke, J.; Chen, Y.; He, X.; Zheng, X.; He, X.; Hu, J.; et al. Preoperative Hypoalbuminemia Is Associated with an Increased Risk for Intra-Abdominal Septic Complications after Primary Anastomosis for Crohn’s Disease. Gastroenterol. Rep. 2017, 5, 298–304. [Google Scholar] [CrossRef]
  162. Morar, P.S.; Hodgkinson, J.D.; Thalayasingam, S.; Koysombat, K.; Purcell, M.; Hart, A.L.; Warusavitarne, J.; Faiz, O. Determining Predictors for Intra-Abdominal Septic Complications Following Ileocolonic Resection for Crohn’s Disease--Considerations in Pre-Operative and Peri-Operative Optimisation Techniques to Improve Outcome. J. Crohns Colitis 2015, 9, 483–491. [Google Scholar] [CrossRef]
  163. Zhang, T.; Yang, J.; Ding, C.; Li, Y.; Gu, L.; Wei, Y.; Cao, L.; Gong, J.; Zhu, W.; Li, N.; et al. Preoperative Intra-Abdominal Sepsis, Not Penetrating Behavior Itself, Is Associated With Worse Postoperative Outcome After Bowel Resection for Crohn Disease. Medicine 2015, 94, e1987. [Google Scholar] [CrossRef]
  164. Zhu, F.; Li, Y.; Guo, Z.; Cao, L.; Feng, D.; Zhang, T.; Zhu, W.; Gong, J. Nomogram to Predict Postoperative Intra-Abdominal Septic Complications After Bowel Resection and Primary Anastomosis for Crohn’s Disease. Dis. Colon Rectum 2020, 63, 629–638. [Google Scholar] [CrossRef] [PubMed]
  165. Müller, C.; Stift, A.; Argeny, S.; Bergmann, M.; Gnant, M.; Marolt, S.; Unger, L.; Riss, S. Delta Albumin Is a Better Prognostic Marker for Complications Following Laparoscopic Intestinal Resection for Crohn’s Disease than Albumin Alone—A Retrospective Cohort Study. PLoS ONE 2018, 13, e0206911. [Google Scholar] [CrossRef] [PubMed]
  166. El-Hussuna, A.; Karer, M.L.M.; Uldall Nielsen, N.N.; Mujukian, A.; Fleshner, P.R.; Iesalnieks, I.; Horesh, N.; Kopylov, U.; Jacoby, H.; Al-Qaisi, H.M.; et al. Postoperative Complications and Waiting Time for Surgical Intervention after Radiologically Guided Drainage of Intra-Abdominal Abscess in Patients with Crohn’s Disease. BJS Open 2021, 5, zrab075. [Google Scholar] [CrossRef] [PubMed]
  167. Hiraoka, S.; Takashima, S.; Kondo, Y.; Inokuchi, T.; Sugihara, Y.; Takahara, M.; Kawano, S.; Harada, K.; Kato, J.; Okada, H. Efficacy of Restarting Anti-Tumor Necrosis Factor α Agents after Surgery in Patients with Crohn’s Disease. Intest. Res. 2018, 16, 75. [Google Scholar] [CrossRef] [PubMed]
  168. Ananthakrishnan, A.N.; McGinley, E.L.; Binion, D.G.; Saeian, K. A Novel Risk Score to Stratify Severity of Crohn’s Disease Hospitalizations. Am. J. Gastroenterol. 2010, 105, 1799–1807. [Google Scholar] [CrossRef] [PubMed]
  169. Vohra, I.; Attar, B.; Haghbin, H.; Mutneja, H.; Katiyar, V.; Sharma, S.; Abegunde, A.T.; Demetria, M.; Gandhi, S. Incidence and Risk Factors for 30-Day Readmission in Ulcerative Colitis: Nationwide Analysis in Biologic Era. Eur. J. Gastroenterol. Hepatol. 2021, 33, 1174–1184. [Google Scholar] [CrossRef] [PubMed]
  170. Lee, H.J.; Jung, E.S.; Lee, J.H.; Hong, S.P.; Kim, T.I.; Kim, W.H.; Cheon, J.H. Long-Term Clinical Outcomes and Factors Predictive of Relapse after 5-Aminosalicylate or Sulfasalazine Therapy in Patients with Mild-to-Moderate Ulcerative Colitis. Hepato-Gastroenterology 2012, 59, 1415–1420. [Google Scholar]
  171. Abou Khalil, M.; Boutros, M.; Nedjar, H.; Morin, N.; Ghitulescu, G.; Vasilevsky, C.A.; Gordon, P.; Rahme, E. Incidence Rates and Predictors of Colectomy for Ulcerative Colitis in the Era of Biologics: Results from a Provincial Database. J. Gastrointest. Surg. 2018, 22, 124–132. [Google Scholar] [CrossRef]
  172. Aydinli, H.H.; Aytac, E.; Remzi, F.H.; Bernstein, M.; Grucela, A.L. Factors Associated with Short-Term Morbidity in Patients Undergoing Colon Resection for Crohn’s Disease. J. Gastrointest. Surg. 2018, 22, 1434–1441. [Google Scholar] [CrossRef]
  173. Prakash, K.; Varma, D.; Mahadevan, P.; Narayanan, R.G.; Philip, M. Surgical Treatment for Small Bowel Crohn’s Disease: An Experience of 28 Cases. Indian J. Gastroenterol. 2008, 27, 12–15. [Google Scholar]
  174. Iesalnieks, I.; Spinelli, A.; Frasson, M.; Di Candido, F.; Scheef, B.; Horesh, N.; Iborra, M.; Schlitt, H.J.; El-Hussuna, A. Risk of Postoperative Morbidity in Patients Having Bowel Resection for Colonic Crohn’s Disease. Tech. Coloproctol. 2018, 22, 947–953. [Google Scholar] [CrossRef]
  175. Hu, T.; Wu, X.; Hu, J.; Chen, Y.; Liu, H.; Zhou, C.; He, X.; Zhi, M.; Wu, X.; Lan, P. Incidence and Risk Factors for Incisional Surgical Site Infection in Patients with Crohn’s Disease Undergoing Bowel Resection. Gastroenterol. Rep. 2018, 6, 189–194. [Google Scholar] [CrossRef]
  176. Larson, D.W.; Abd El Aziz, M.A.; Perry, W.; Behm, K.T.; Shawki, S.; Mandrekar, J.; Mathis, K.L.; Grass, F. Surgical Resection for Crohn’s and Cancer: A Comparison of Disease-Specific Risk Factors and Outcomes. Dig. Surg. 2021, 38, 120–127. [Google Scholar] [CrossRef]
  177. Pozios, I.; Seeliger, H.; Lauscher, J.C.; Stroux, A.; Weixler, B.; Kamphues, C.; Beyer, K.; Kreis, M.E.; Lehmann, K.S.; Seifarth, C. Risk Factors for Upper and Lower Type Prolonged Postoperative Ileus Following Surgery for Crohn’s Disease. Int. J. Color. Dis. 2021, 36, 2165–2175. [Google Scholar] [CrossRef]
  178. Herrera-deGuise, C.; Casellas, F.; Robles, V.; Navarro, E.; Borruel, N. Iron Deficiency in the Absence of Anemia Impairs the Perception of Health-Related Quality of Life of Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2016, 22, 1450–1455. [Google Scholar] [CrossRef]
  179. Peyrin-Biroulet, L.; Lopez, A.; Cummings, J.R.F.; Dignass, A.; Detlie, T.E.; Danese, S. Review Article: Treating-to-Target for Inflammatory Bowel Disease-Associated Anaemia. Aliment. Pharmacol. Ther. 2018, 48, 610–617. [Google Scholar] [CrossRef]
  180. Auerbach, M.; Deloughery, T. Single-Dose Intravenous Iron for Iron Deficiency: A New Paradigm. Hematology 2016, 2016, 57–66. [Google Scholar] [CrossRef]
  181. Çekiç, C.; İpek, S.; Aslan, F.; Akpınar, Z.; Arabul, M.; Topal, F.; Sarıtaş Yüksel, E.; Alper, E.; Ünsal, B. The Effect of Intravenous Iron Treatment on Quality of Life in Inflammatory Bowel Disease Patients with Nonanemic Iron Deficiency. Gastroenterol. Res. Pract. 2015, 2015, 582163. [Google Scholar] [CrossRef]
  182. Huguet, J.M.; Cortés, X.; Boscá-Watts, M.M.; Muñoz, M.; Maroto, N.; Iborra, M.; Hinojosa, E.; Capilla, M.; Asencio, C.; Amoros, C.; et al. Ferric Carboxymaltose Improves the Quality of Life of Patients with Inflammatory Bowel Disease and Iron Deficiency without Anaemia. J. Clin. Med. 2022, 11, 2786. [Google Scholar] [CrossRef]
  183. Sadeghian, M.; Saneei, P.; Siassi, F.; Esmaillzadeh, A. Vitamin D Status in Relation to Crohn’s Disease: Meta-Analysis of Observational Studies. Nutrition 2016, 32, 505–514. [Google Scholar] [CrossRef]
  184. Gubatan, J.; Chou, N.D.; Nielsen, O.H.; Moss, A.C. Systematic Review with Meta-Analysis: Association of Vitamin D Status with Clinical Outcomes in Adult Patients with Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2019, 50, 1146–1158. [Google Scholar] [CrossRef]
  185. De Bruyn, J.R.; Bossuyt, P.; Ferrante, M.; West, R.L.; Dijkstra, G.; Witteman, B.J.; Wildenberg, M.; Hoentjen, F.; Franchimont, D.; Clasquin, E.; et al. High-Dose Vitamin D Does Not Prevent Postoperative Recurrence of Crohn’s Disease in a Randomized Placebo-Controlled Trial. Clin. Gastroenterol. Hepatol. 2021, 19, 1573–1582. [Google Scholar] [CrossRef]
  186. Valvano, M.; Magistroni, M.; Cesaro, N.; Carlino, G.; Monaco, S.; Fabiani, S.; Vinci, A.; Vernia, F.; Viscido, A.; Latella, G. Effectiveness of Vitamin D Supplementation on Disease Course in Inflammatory Bowel Disease Patients: Systematic Review With Meta-Analysis. Inflamm. Bowel Dis. 2022, izac253. [Google Scholar] [CrossRef]
  187. Wright, J.P.; Mee, A.S.; Parfitt, A.; Marks, I.N.; Burns, D.G.; Sherman, M.; Tigler-Wybrandi, N.; Isaacs, S. Vitamin A Therapy in Patients with Crohn’s Disease. Gastroenterology 1985, 88, 512–514. [Google Scholar] [CrossRef]
  188. Bager, P.; Hvas, C.L.; Rud, C.L.; Dahlerup, J.F. Randomised Clinical Trial: High-Dose Oral Thiamine versus Placebo for Chronic Fatigue in Patients with Quiescent Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2021, 53, 79–86. [Google Scholar]
  189. Von Martels, J.Z.H.; Bourgonje, A.R.; Klaassen, M.A.Y.; Alkhalifah, H.A.A.; Sadaghian Sadabad, M.; Vich Vila, A.; Gacesa, R.; Gabriëls, R.Y.; Steinert, R.E.; Jansen, B.H.; et al. Riboflavin Supplementation in Patients with Crohn’s Disease [the RISE-UP Study]. J. Crohns Colitis 2020, 14, 595–607. [Google Scholar] [CrossRef]
  190. Itagaki, M.; Saruta, M.; Saijo, H.; Mitobe, J.; Arihiro, S.; Matsuoka, M.; Kato, T.; Ikegami, M.; Tajiri, H. Efficacy of Zinc–Carnosine Chelate Compound, Polaprezinc, Enemas in Patients with Ulcerative Colitis. Scand. J. Gastroenterol. 2014, 49, 164–172. [Google Scholar] [CrossRef]
  191. Stedman, J.D.; Spyrou, N.M.; Millar, A.D.; Altaf, W.J.; Akanle, O.A.; Rampton, D.S. Selenium Supplementation in the Diets of Patients Suffering from Ulcerative Colitis. J. Radioanal. Nucl. Chem. 1997, 217, 189–191. [Google Scholar] [CrossRef]
  192. Wu, Y.; Liu, C.; Dong, W. Adjunctive Therapeutic Effects of Micronutrient Supplementation in Inflammatory Bowel Disease. Front. Immunol. 2023, 14, 1143123. [Google Scholar] [CrossRef]
  193. Dignass, A.U.; Gasche, C.; Bettenworth, D.; Birgegård, G.; Danese, S.; Gisbert, J.P.; Gomollon, F.; Iqbal, T.; Katsanos, K.; Koutroubakis, I.; et al. European Consensus on the Diagnosis and Management of Iron Deficiency and Anaemia in Inflammatory Bowel Diseases. J. Crohns Colitis 2015, 9, 211–222. [Google Scholar] [CrossRef]
  194. Bager, P.; Befrits, R.; Wikman, O.; Lindgren, S.; Moum, B.; Hjortswang, H.; Dahlerup, J.F. High Burden of Iron Deficiency and Different Types of Anemia in Inflammatory Bowel Disease Outpatients in Scandinavia: A Longitudinal 2-Year Follow-up Study. Scand. J. Gastroenterol. 2013, 48, 1286–1293. [Google Scholar] [CrossRef] [PubMed]
  195. Bager, P.; Befrits, R.; Wikman, O.; Lindgren, S.; Moum, B.; Hjortswang, H.; Dahlerup, J.F. The Prevalence of Anemia and Iron Deficiency in IBD Outpatients in Scandinavia. Scand. J. Gastroenterol. 2011, 46, 304–309. [Google Scholar] [CrossRef] [PubMed]
  196. Burisch, J.; Vegh, Z.; Katsanos, K.H.; Christodoulou, D.K.; Lazar, D.; Goldis, A.; O’Morain, C.; Fernandez, A.; Pereira, S.; Myers, S.; et al. Occurrence of Anaemia in the First Year of Inflammatory Bowel Disease in a European Population-Based Inception Cohort—An ECCO-EpiCom Study. J. Crohns Colitis 2017, 11, 1213–1222. [Google Scholar] [CrossRef] [PubMed]
  197. Al Khoury, A.; Singh, K.; Kurti, Z.; Gonczi, L.; Golovics, P.; Kohen, R.; Afif, W.; Wild, G.; Bitton, A.; Bessissow, T.; et al. The Burden of Anemia Remains Significant over Time in Patients with Inflammatory Bowel Diseases at a Tertiary Referral Center. J. Gastrointestin. Liver Dis. 2020, 29, 555–559. [Google Scholar] [CrossRef] [PubMed]
  198. Santos, B.R.M.D.; Santos, C.H.M.D.; Santos, V.R.M.D.; Torrez, C.Y.G.; Palomares-junior, D. Predictive factors for loss of response to anti-tnf in crohn’s disease. ABCD Arq. Bras. Cir. Dig. 2020, 33, e1522. [Google Scholar] [CrossRef] [PubMed]
  199. Wang, C.; He, L.; Zhang, J.; Ouyang, C.; Wu, X.; Lu, F.; Liu, X. Clinical, Laboratory, Endoscopical and Histological Characteristics Predict Severe Ulcerative Colitis. Hepato-Gastroenterology 2013, 60, 318–323. [Google Scholar] [PubMed]
  200. Alves, R.A.; Miszputen, S.J.; Figueiredo, M.S. Anemia in Inflammatory Bowel Disease: Prevalence, Differential Diagnosis and Association with Clinical and Laboratory Variables. Sao Paulo Med. J. 2014, 132, 140–146. [Google Scholar] [CrossRef]
  201. Vegh, Z.; Kurti, Z.; Gonczi, L.; Golovics, P.A.; Lovasz, B.D.; Szita, I.; Balogh, M.; Pandur, T.; Vavricka, S.R.; Rogler, G.; et al. Association of Extraintestinal Manifestations and Anaemia with Disease Outcomes in Patients with Inflammatory Bowel Disease. Scand. J. Gastroenterol. 2016, 51, 848–854. [Google Scholar] [CrossRef]
  202. Koutroubakis, I.E.; Ramos–Rivers, C.; Regueiro, M.; Koutroumpakis, E.; Click, B.; Schoen, R.E.; Hashash, J.G.; Schwartz, M.; Swoger, J.; Baidoo, L.; et al. Persistent or Recurrent Anemia Is Associated With Severe and Disabling Inflammatory Bowel Disease. Clin. Gastroenterol. Hepatol. 2015, 13, 1760–1766. [Google Scholar] [CrossRef]
  203. Stein, J.; Hartmann, F.; Dignass, A.U. Diagnosis and Management of Iron Deficiency Anemia in Patients with IBD. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 599–610. [Google Scholar] [CrossRef]
  204. Parra, R.S.; Feitosa, M.R.; Ferreira, S.C.; Rocha, J.J.R.D.; Troncon, L.E.; Troncon, L.E.D.A.; Féres, O. Anemia and iron deficiency in inflammatory bowel disease patients in a referral center in Brazil: Prevalence and risk factors. Arq. Gastroenterol. 2020, 57, 272–277. [Google Scholar] [CrossRef] [PubMed]
  205. Gasche, C. Iron, Anaemia, and Inflammatory Bowel Diseases. Gut 2004, 53, 1190–1197. [Google Scholar] [CrossRef] [PubMed]
  206. González Alayón, C.; Pedrajas Crespo, C.; Marín Pedrosa, S.; Benítez, J.M.; Iglesias Flores, E.; Salgueiro Rodríguez, I.; Medina Medina, R.; García-Sánchez, V. Prevalencia de Déficit de Hierro Sin Anemia En La Enfermedad Inflamatoria Intestinal y Su Impacto En La Calidad de Vida. Gastroenterol. Hepatol. 2018, 41, 22–29. [Google Scholar] [CrossRef] [PubMed]
  207. Bergamaschi, G.; Castiglione, F.; D’Incà, R.; Astegiano, M.; Fries, W.; Milla, M.; Ciacci, C.; Rizzello, F.; Saibeni, S.; Ciccocioppo, R.; et al. Prevalence, Pathogenesis and Management of Anemia in Inflammatory Bowel Disease: An IG-IBD Multicenter, Prospective, and Observational Study. Inflamm. Bowel Dis. 2023, 29, 76–84. [Google Scholar] [CrossRef] [PubMed]
  208. Peyrin-Biroulet, L.; Bouguen, G.; Laharie, D.; Pellet, G.; Savoye, G.; Gilletta, C.; Michiels, C.; Buisson, A.; Fumery, M.; Trochu, J.N.; et al. Iron Deficiency in Patients with Inflammatory Bowel Diseases: A Prospective Multicenter Cross-Sectional Study. Dig. Dis. Sci. 2022, 67, 5637–5646. [Google Scholar] [CrossRef] [PubMed]
  209. Eliadou, E.; Kini, G.; Huang, J.; Champion, A.; Inns, S.J. Intravenous Iron Replacement Improves Quality of Life in Hypoferritinemic Inflammatory Bowel Disease Patients with and without Anemia. Dig. Dis. 2017, 35, 444–448. [Google Scholar] [CrossRef] [PubMed]
  210. De Silva, A.D.; Tsironi, E.; Feakins, R.M.; Rampton, D.S. Efficacy and Tolerability of Oral Iron Therapy in Inflammatory Bowel Disease: A Prospective, Comparative Trial. Aliment. Pharmacol. Ther. 2005, 22, 1097–1105. [Google Scholar] [CrossRef]
  211. Oldenburg, B.; Koningsberger, J.C.; Van Berge Henegouwen, G.P.; Van Asbeck, B.S.; Marx, J.J.M. Iron and Inflammatory Bowel Disease. Aliment. Pharmacol. Ther. 2001, 15, 429–438. [Google Scholar] [CrossRef]
  212. Erichsen, K.; Ulvik, R.J.; Nysaeter, G.; Johansen, J.; Ostborg, J.; Berstad, A.; Berge, R.K.; Hausken, T. Oral Ferrous Fumarate or Intravenous Iron Sucrose for Patients with Inflammatory Bowel Disease. Scand. J. Gastroenterol. 2005, 40, 1058–1065. [Google Scholar] [CrossRef]
  213. Zhu, A.; Kaneshiro, M.; Kaunitz, J.D. Evaluation and Treatment of Iron Deficiency Anemia: A Gastroenterological Perspective. Dig. Dis. Sci. 2010, 55, 548–559. [Google Scholar] [CrossRef]
  214. Bonovas, S.; Fiorino, G.; Allocca, M.; Lytras, T.; Tsantes, A.; Peyrin-Biroulet, L.; Danese, S. Intravenous Versus Oral Iron for the Treatment of Anemia in Inflammatory Bowel Disease. Medicine 2016, 95, e2308. [Google Scholar] [CrossRef] [PubMed]
  215. Moore, R.A.; Gaskell, H.; Rose, P.; Allan, J. Meta-Analysis of Efficacy and Safety of Intravenous Ferric Carboxymaltose (Ferinject) from Clinical Trial Reports and Published Trial Data. BMC Blood Disord. 2011, 11, 4. [Google Scholar] [CrossRef] [PubMed]
  216. Hoffbrand, A.V.; Stewart, J.S.; Booth, C.C.; Mollin, D.L. Folate Deficiency in Crohn’s Disease: Incidence, Pathogenesis, and Treatment. BMJ 1968, 2, 71–75. [Google Scholar] [CrossRef] [PubMed]
  217. Hwang, C.; Ross, V.; Mahadevan, U. Micronutrient Deficiencies in Inflammatory Bowel Disease: From A to Zinc. Inflamm. Bowel Dis. 2012, 18, 1961–1981. [Google Scholar] [CrossRef] [PubMed]
  218. Pan, Y.; Liu, Y.; Guo, H.; Jabir, M.S.; Liu, X.; Cui, W.; Li, D. Associations between Folate and Vitamin B12 Levels and Inflammatory Bowel Disease: A Meta-Analysis. Nutrients 2017, 9, 382. [Google Scholar] [CrossRef] [PubMed]
  219. Ward, M.G.; Kariyawasam, V.C.; Mogan, S.B.; Patel, K.V.; Pantelidou, M.; Sobczyńska-Malefora, A.; Porté, F.; Griffin, N.; Anderson, S.H.C.; Sanderson, J.D.; et al. Prevalence and Risk Factors for Functional Vitamin B12 Deficiency in Patients with Crohn’s Disease. Inflamm. Bowel Dis. 2015, 21, 2839–2847. [Google Scholar] [CrossRef] [PubMed]
  220. Yakut, M.; Ustün, Y.; Kabaçam, G.; Soykan, I. Serum Vitamin B12 and Folate Status in Patients with Inflammatory Bowel Diseases. Eur. J. Intern. Med. 2010, 21, 320–323. [Google Scholar] [CrossRef]
  221. Battat, R.; Kopylov, U.; Szilagyi, A.; Saxena, A.; Rosenblatt, D.S.; Warner, M.; Bessissow, T.; Seidman, E.; Bitton, A. Vitamin B12 Deficiency in Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2014, 20, 1120–1128. [Google Scholar] [CrossRef]
  222. Giustina, A.; Di Filippo, L.; Allora, A.; Bikle, D.D.; Cavestro, G.M.; Feldman, D.; Latella, G.; Minisola, S.; Napoli, N.; Trasciatti, S.; et al. Vitamin D and Malabsorptive Gastrointestinal Conditions: A Bidirectional Relationship? Rev. Endocr. Metab. Disord. 2023, 24, 121–138. [Google Scholar] [CrossRef]
  223. Del Pinto, R.; Pietropaoli, D.; Chandar, A.K.; Ferri, C.; Cominelli, F. Association Between Inflammatory Bowel Disease and Vitamin D Deficiency. Inflamm. Bowel Dis. 2015, 21, 2708–2717. [Google Scholar] [CrossRef]
  224. Li, X.X.; Liu, Y.; Luo, J.; Huang, Z.D.; Zhang, C.; Fu, Y. Vitamin D Deficiency Associated with Crohn’s Disease and Ulcerative Colitis: A Meta-Analysis of 55 Observational Studies. J. Transl. Med. 2019, 17, 323. [Google Scholar] [CrossRef] [PubMed]
  225. Guo, Y.; Zhang, T.; Wang, Y.; Liu, R.; Chang, M.; Wang, X. Effects of Oral Vitamin D Supplementation on Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Food Funct. 2021, 12, 7588–7606. [Google Scholar] [CrossRef] [PubMed]
  226. Guzman-Prado, Y.; Samson, O.; Segal, J.P.; Limdi, J.K.; Hayee, B. Vitamin D Therapy in Adults With Inflammatory Bowel Disease: A Systematic Review and Meta-Analysis. Inflamm. Bowel Dis. 2020, 26, 1819–1830. [Google Scholar] [CrossRef] [PubMed]
  227. Li, J.; Chen, N.; Wang, D.; Zhang, J.; Gong, X. Efficacy of Vitamin D in Treatment of in Fl Ammatory Bowel Disease. Medicine 2018, 97(46), e12662. [Google Scholar] [CrossRef] [PubMed]
  228. Guo, X.; Liu, C.; Huang, Y. Efficacy and Safety of Vitamin D Adjuvant Therapy for Ulcerative Colitis: A Meta-Analysis. Comput. Math. Methods Med. 2022, 2022, 6836942. [Google Scholar] [CrossRef] [PubMed]
  229. Gubatan, J.; Rubin, S.J.S.; Bai, L.; Haileselassie, Y.; Levitte, S.; Balabanis, T.; Patel, A.; Sharma, A.; Sinha, S.R.; Habtezion, A. Vitamin D Is Associated with A4β7+ Immunophenotypes and Predicts Vedolizumab Therapy Failure in Patients with Inflammatory Bowel Disease. J. Crohns Colitis 2021, 15, 1980–1990. [Google Scholar] [CrossRef] [PubMed]
  230. Zator, Z.A.; Cantu, S.M.; Konijeti, G.G.; Nguyen, D.D.; Sauk, J.; Yajnik, V.; Ananthakrishnan, A.N. Pretreatment 25-Hydroxyvitamin D Levels and Durability of Anti–Tumor Necrosis Factor–α Therapy in Inflammatory Bowel Diseases. J. Parenter. Enter. Nutr. 2014, 38, 385–391. [Google Scholar] [CrossRef] [PubMed]
  231. Xia, S.; Min, Q.; Shao, X.; Lin, D.; Ma, G.; Wu, H.; Cao, S.; Jiang, Y. Influence of Vitamin D3 Supplementation on Infliximab Effectiveness in Chinese Patients With Crohn’s Disease: A Retrospective Cohort Study. Front. Nutr. 2021, 8, 739285. [Google Scholar] [CrossRef]
  232. Bendix, M.; Dige, A.; Jørgensen, S.P.; Dahlerup, J.F.; Bibby, B.M.; Deleuran, B.; Agnholt, J. Seven Weeks of High-Dose Vitamin D Treatment Reduces the Need for Infliximab Dose-Escalation and Decreases Inflammatory Markers in Crohn’s Disease during One-Year Follow-Up. Nutrients 2021, 13, 1083. [Google Scholar] [CrossRef]
  233. Dadaei, T.; Safapoor, M.H.; Aghdaei, H.A.; Balaii, H.; Pourhoseingholi, M.A.; Naderi, N.; Zojaji, H.; Azimzadeh, P.; Mohammadi, P.; Zali, M.R. Effect of Vitamin D3 Supplementation on TNF-α Serum Level and Disease Activity Index in Iranian IBD Patients. Gastroenterol. Hepatol. Bed Bench 2015, 8, 49–55. [Google Scholar]
  234. Ananthakrishnan, A.N.; Cagan, A.; Gainer, V.S.; Cai, T.; Cheng, S.C.; Savova, G.; Chen, P.; Szolovits, P.; Xia, Z.; De Jager, P.L.; et al. Normalization of Plasma 25-Hydroxy Vitamin D Is Associated with Reduced Risk of Surgery in Crohn’s Disease. Inflamm. Bowel Dis. 2013, 19, 1921–1927. [Google Scholar] [CrossRef] [PubMed]
  235. Yamada, A.; Komaki, Y.; Komaki, F.; Haider, H.; Micic, D.; Pekow, J.; Dalal, S.; Cohen, R.D.; Cannon, L.; Umanskiy, K.; et al. The Correlation between Vitamin D Levels and the Risk of Postoperative Recurrence in Crohn’s Disease. Digestion 2021, 102, 767–775. [Google Scholar] [CrossRef]
  236. Fabisiak, N.; Fabisiak, A.; Watala, C.; Fichna, J. Fat-Soluble Vitamin Deficiencies and Inflammatory Bowel Disease: Systematic Review and Meta-Analysis. J. Clin. Gastroenterol. 2017, 51, 878–889. [Google Scholar] [CrossRef] [PubMed]
  237. Nakajima, S.; Iijima, H.; Egawa, S.; Shinzaki, S.; Kondo, J.; Inoue, T.; Hayashi, Y.; Ying, J.; Mukai, A.; Akasaka, T.; et al. Association of Vitamin K Deficiency with Bone Metabolism and Clinical Disease Activity in Inflammatory Bowel Disease. Nutrition 2011, 27, 1023–1028. [Google Scholar] [CrossRef]
  238. Imes, S.; Dinwoodie, A.; Walker, K.; Pinchbeck, B.; Thomson, A.B.R. Vitamin C Status in 137 Outpatients with Crohn’s Disease: Effect of Diet Counseling. J. Clin. Gastroenterol. 1986, 8, 443–446. [Google Scholar] [CrossRef] [PubMed]
  239. Dunleavy, K.A.; Ungaro, R.C.; Manning, L.; Gold, S.; Novak, J.; Colombel, J.F. Vitamin C Deficiency in Inflammatory Bowel Disease: The Forgotten Micronutrient. Crohns Colitis 360 2021, 3, otab009. [Google Scholar] [CrossRef] [PubMed]
  240. Ramakrishna, B.; Varghese, R.; Jayakumar, S.; Mathan, M.; Balasubramanian, K. Circulating Antioxidants in Ulcerative Colitis and Their Relationship to Disease Severity and Activity. J. Gastroenterol. Hepatol. 1997, 12, 490–494. [Google Scholar] [CrossRef]
  241. Verma, P.; Subodh, S.; Tiwari, V.; Rampal, R.; Tuteja, A.; Toteja, G.S.; Gupta, S.D.; Ahuja, V. Correlation of Serum Vitamin A Levels with Disease Activity Indices and Colonic IL-23R and FOXP3 MRNA Expression in Ulcerative Colitis Patients. Scand. J. Immunol. 2016, 84, 110–117. [Google Scholar] [CrossRef]
  242. Sampietro, G.M.; Cristaldi, M.; Cervato, G.; Maconi, G.; Danelli, P.; Cervellione, R.; Rovati, M.; Bianchi Porro, G.; Cestaro, B.; Taschieri, A.M. Oxidative Stress, Vitamin A and Vitamin E Behaviour in Patients Submitted to Conservative Surgery for Complicated Crohn’s Disease. Dig. Liver Dis. 2002, 34, 696–701. [Google Scholar] [CrossRef]
  243. Vagianos, K.; Bernstein, C.N. Homocysteinemia and B Vitamin Status among Adult Patients with Inflammatory Bowel Disease: A One-Year Prospective Follow-up Study. Inflamm. Bowel Dis. 2012, 18, 718–724. [Google Scholar] [CrossRef]
  244. Costantini, A.; Pala, M.I. Thiamine and Fatigue in Inflammatory Bowel Diseases: An Open-Label Pilot Study. J. Altern. Complement. Med. 2013, 19, 704–708. [Google Scholar] [CrossRef] [PubMed]
  245. Ye, R.; Huang, J.; Wang, Z.; Chen, Y.; Dong, Y. Trace Element Selenium Effectively Alleviates Intestinal Diseases. Int. J. Mol. Sci. 2021, 22, 11708. [Google Scholar] [CrossRef] [PubMed]
  246. Vaghari-Tabari, M.; Jafari-Gharabaghlou, D.; Sadeghsoltani, F.; Hassanpour, P.; Qujeq, D.; Rashtchizadeh, N.; Ghorbanihaghjo, A. Zinc and Selenium in Inflammatory Bowel Disease: Trace Elements with Key Roles? Biol. Trace Elem. Res. 2021, 199, 3190–3204. [Google Scholar] [CrossRef] [PubMed]
  247. Siva, S.; Rubin, D.T.; Gulotta, G.; Wroblewski, K.; Pekow, J. Zinc Deficiency Is Associated with Poor Clinical Outcomes in Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2017, 23, 152–157. [Google Scholar] [CrossRef] [PubMed]
  248. Sturniolo, G.C.; Di Leo, V.; Ferronato, A.; D’Odorico, A.; D’Incà, R. Zinc Supplementation Tightens “Leaky Gut” in Crohn’s Disease. Inflamm. Bowel Dis. 2001, 7, 94–98. [Google Scholar] [CrossRef]
  249. Sakurai, K.; Furukawa, S.; Katsurada, T.; Otagiri, S.; Yamanashi, K.; Nagashima, K.; Onishi, R.; Yagisawa, K.; Nishimura, H.; Ito, T.; et al. Effectiveness of Administering Zinc Acetate Hydrate to Patients with Inflammatory Bowel Disease and Zinc Deficiency: A Retrospective Observational Two-Center Study. Intest. Res. 2022, 20, 78–89. [Google Scholar] [CrossRef] [PubMed]
  250. Younus, M.; Taher, M.A.; ALMaliki, J.; Alkhalidi, N.; Hussein, R. Selenium Supplementation May Decrease the Rate of Infliximab ADRs in IBD Patients. World J. Pharm. Res. 2015, 4, 193–203. [Google Scholar]
  251. Dalekos, G.N.; Ringstad, J.; Savaidis, L.; Seferiadis, K.I.; Tsianos, E.V. Zinc, Copper and Immunological Markers in the Circulation of Well Nourished Patients with Ulcerative Colitis. Eur. J. Gastroenterol. Hepatol. 1998, 10, 331–338. [Google Scholar] [CrossRef]
  252. Poursadegh, F.; Ahadi, M.; Vosoughinia, H.; Salehi, M.; Beheshti Namdar, A.; Farzanehfar, M.R.; Memar, B.; Ziaolhagh, R. A STROBE Compliant Observational Study on Trace Elements in Patients with Ulcerative Colitis and Their Relationship with Disease Activity. Medicine 2018, 97, e13523. [Google Scholar] [CrossRef]
  253. Fernández-Bañares, F.; Mingorance, M.D.; Esteve, M.; Cabré, E.; Lachica, M.; Abad-Lacruz, A.; Gil, A.; Humbert, P.; Boix, J.; Gassull, M.A. Serum Zinc, Copper, and Selenium Levels in Inflammatory Bowel Disease: Effect of Total Enteral Nutrition on Trace Element Status. Am. J. Gastroenterol. 1990, 85, 1584–1589. [Google Scholar]
  254. El Muhtaseb, M.S.H.; Duncan, A.; Talwar, D.K.; O’Reilly, D.S.J.; McKee, R.F.; Anderson, J.H.; Finlay, I.G. Assessment of Dietary Intake and Trace Element Status in Patients with Ileal Pouch-Anal Anastomosis. Dis. Colon Rectum 2007, 50, 1553–1557. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Nutrition, nutritional status, micronutrients deficiency and disease course of Inflammatory Bowel Disease [11].
Figure 1. Nutrition, nutritional status, micronutrients deficiency and disease course of Inflammatory Bowel Disease [11].
Nutrients 15 03824 g001
Table 1. Nutritional support for induction and maintenance of remission in UC and CD.
Table 1. Nutritional support for induction and maintenance of remission in UC and CD.
Nutritional SupportInduction of RemissionMaintenance of Remission
UCCDUCCD
EN
[32,33,34,35,36,37,38,39,40]
N.A.Less effective than steroidsN.A.Higher clinical remission maintenance and lower rate of clinical relapse
Fibres
[41,42,43,44]
Better outcome in remission and colectomy rateNo advantage in reducing fibresProtective effect against relapseProtective effect against relapse
UCED
[45,46]
Higher remission and mucosal healing ratesN.A.N.A.N.A.
CDED
[47]
N.A.Effective for induction and maintenance of remissionN.A.N.A.
Oral diets
[27,48]
Not helpfulHigher induction of remission rateRed meat diet may increase relapse rate; no advantage for other types of dietNo advantage in low red meat and low refined carbohydrates
UC: Ulcerative colitis; CD: Crohn Disease; N.A: Not available; EN: Enteral Nutrition; UCED: Ulcerative Colitis Exclusion Diet; CDED: Crohn’s disease exclusion diet.
Table 2. Nutritional support and surgical outcome in UC and CD.
Table 2. Nutritional support and surgical outcome in UC and CD.
Nutritional SupportPrior to SurgeryAfeter Surgery
UCCDUC: PouchitisCD: Short Bowel Syndrome
EN
[52,55]
N.A.Reduce postoperative complicationClinical improvement; no improvement in endoscopy and histology N.A.
Fibres
[40,42,43,44,50,51,52,53,54,55,56,57,58]
N.A.N.A.Endoscopic and histologic improvementN.A.
High-carbohydrate-low-fat diet
[59]
N.A.N.A.N.A.Carbohydrates in patients without a colon have no effects; fat reduction is not recommended
UC: Ulcerative colitis; CD: Crohn Disease; N.A.: Not available; EN: Enteral Nutrition.
Table 3. Anemia, micronutrients deficiency and disease course.
Table 3. Anemia, micronutrients deficiency and disease course.

Anemia and Micronutrients Deficiency
Effect on Disease CourseEffect of
Supplementation
CDUC
Anemia [168,169,170,171,172,173,174,175,176,177]Higher risk of hospitalization
Higher risk of surgery
Higher risk of surgery related complication
Higher risk of hospitalization
Higher risk of clinical Relapse
Higher risk of surgery
N.A.
Iron [178,179,180]Low QoL
Worst disease control
Low QoLImprove QoL [181,182]
Vitamin B12 and Folic acidN.A.N.A.N.A.
Vitamin D [183,184,185]Higher risk of active disease
Higher risk of clinical relapse
Higher risk of surgery
Higher risk of active disease
Higher risk of clinical relapse
Higher risk of surgery
Lower risk of clinical relapse [186]
Vitamin AN.A.Lower risk of clinical relapse [187]
Vitamin B1N.A.Reduction of IBD chronic fatigue [188]
Vitamin B2N.A.Reduction of clinical activity [189]
Vitamin E, K, CN.A.N.A.
ZincN.A.Higher clinical
response [190]
SeleniumN.A.Improve clinical symptoms [191]
Copper and ManganeseN.A.N.A.
UC: Ulcerative colitis; CD: Crohn Disease; N.A.: Not available; QoL: Quality of life.
Table 4. Summary of main findings of nutrition, nutrition status, and micronutrients deficiency.
Table 4. Summary of main findings of nutrition, nutrition status, and micronutrients deficiency.
Nutrition and Nutritional Status
Disease Clinical CourseInduction of RemissionMaintenance of RemissionRisk of SurgerySurgery-Related Complications; POR
Exclusion DietsInsufficient dataInsufficient data, but most don’t impactInsufficient data, but most don’t impactInsufficient dataInsufficient data
EN/PNMaybe usefulInferior to steroidsMaybe usefulInsufficient dataReduce the risk
SarcopeniaSlight increase in worse clinical and endoscopic outcomes in CD patientsInsufficient dataSlight increase of recurrence after surgery in CD patients.Slight increase in IBD patients.Slight increase in POR in CD patients. Higher rate of complications (both in UC and CD patients).
ObesityLacking and contrasting data.May worsen the effectiveness of therapiesInsufficient datalikely increases the risk for surgery (in CD)increases postoperative complications; risk factor for endoscopic recurrence after surgery (in CD)
HypoalbuminemiaProbably result in a worst clinical courseInsufficient dataProbably increase the numbers of flaresResult in a large reduction of surgery free survivalProbably increase post-surgical complications
Micronutrients deficiency
Disease clinical courseInduction of remissionMaintenance of remissionRisk of surgerySurgery-related complications; POR
AnaemiaProbably result in a worst clinical courseInsufficient dataInsufficient dataInsufficient dataProbably result in a higher risk of perioperative complication
Iron deficiencyProbably result in a worst clinical courseInsufficient dataInsufficient dataInsufficient dataInsufficient data
Vitamin B12 and Folic acid deficiencyInsufficient dataInsufficient dataInsufficient dataInsufficient dataInsufficient data
Vitamin D deficiencyProbably get worse clinical courseInsufficient dataIncrease of clinical relapseInsufficient dataInsufficient data
Other vitamins deficiencyVitamin A, B1 and B2 deficiency Probably get worse disease clinical courseInsufficient dataInsufficient dataInsufficient dataInsufficient data
Other micronutrients deficiencyZinc and selenium probably get worse clinical courseInsufficient dataInsufficient dataInsufficient dataInsufficient data
CD: Crohn Disease; IBD: Inflammatory Bowel Disease; EN/PN: Enteral Nutrition/Parenteral Nutrition; POR: Post-operative recurrence.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Valvano, M.; Capannolo, A.; Cesaro, N.; Stefanelli, G.; Fabiani, S.; Frassino, S.; Monaco, S.; Magistroni, M.; Viscido, A.; Latella, G. Nutrition, Nutritional Status, Micronutrients Deficiency, and Disease Course of Inflammatory Bowel Disease. Nutrients 2023, 15, 3824. https://doi.org/10.3390/nu15173824

AMA Style

Valvano M, Capannolo A, Cesaro N, Stefanelli G, Fabiani S, Frassino S, Monaco S, Magistroni M, Viscido A, Latella G. Nutrition, Nutritional Status, Micronutrients Deficiency, and Disease Course of Inflammatory Bowel Disease. Nutrients. 2023; 15(17):3824. https://doi.org/10.3390/nu15173824

Chicago/Turabian Style

Valvano, Marco, Annalisa Capannolo, Nicola Cesaro, Gianpiero Stefanelli, Stefano Fabiani, Sara Frassino, Sabrina Monaco, Marco Magistroni, Angelo Viscido, and Giovanni Latella. 2023. "Nutrition, Nutritional Status, Micronutrients Deficiency, and Disease Course of Inflammatory Bowel Disease" Nutrients 15, no. 17: 3824. https://doi.org/10.3390/nu15173824

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

Article Metrics

Back to TopTop