Review Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jun 28, 2021; 27(24): 3440-3465
Published online Jun 28, 2021. doi: 10.3748/wjg.v27.i24.3440
Chronic intestinal failure and short bowel syndrome in Crohn’s disease
Aysegül Aksan, Institute of Nutritional Sciences, Justus-Liebig-Universität, Giessen 35392, Germany
Aysegül Aksan, Karima Farrag, Oliver Schröder, Jürgen Stein, Department of Clinical Research, Interdisziplinäres Crohn Colitis Centrum Rhein-Main, Frankfurt am Main 60594, Germany
Karima Farrag, Oliver Schröder, Jürgen Stein, Department of Gastroenterology and Clinical Nutrition, DGD Kliniken Sachsenhausen, Teaching Hospital of the JW Goethe University, Frankfurt am Main 60594, Germany
Irina Blumenstein, Department of Gastroenterology, Hepatology and Clinical Nutrition, First Medical Clinic, JW Goethe University Hospital, Frankfurt am Main 60529, Germany
Axel U Dignass, Department of Medicine I, Agaplesion Markus Hospital, Goethe-University, Frankfurt am Main 60431, Germany
Jürgen Stein, Institute of Pharmaceutical Chemistry, JW Goethe University, 60438 Frankfurt am Main, Germany
ORCID number: Aysegül Aksan (0000-0003-2819-3484); Karima Farrag (0000-0002-5071-7072); Irina Blumenstein (0000-0002-7841-0494); Oliver Schröder (0000-0002-3182-8308); Axel U Dignass (0000-0002-9724-054X); Jürgen Stein (0000-0003-3558-3341).
Author contributions: Aksan A, Farrag K, Blumenstein I, Schröder O, Dignass A, and Stein J designed and performed the literature search, interpreted the data, and drafted the manuscript; All authors have read and approved the final manuscript.
Conflict-of-interest statement: Aksan A reports congress expenses, consultancy fees and research funding from Vifor Pharma and Immundiagnostik AG. Farrag K reports speakers’ honoraria and congress expenses from Immundiagnostik AG, Janssen Cilag GmbH; consultancy honoraria from Vifor Pharma. Blumenstein I reports consultancy honoraria from AbbVie Deutschland, Amgen GmbH, Bristol Myers Squibb, Janssen Cilag GmbH, Fresenius Kabi Deutschland, Pharmacosmos GmbH, Pfizer GmbH, Shire Deutschland GmbH, Takeda Pharma GmbH; honoraria from AbbVie Deutschland, Biogen GmbH, DGVS, Dr. Falk Pharma GmbH, Endoakademie, Ferring Arzneimittel, Fraunhofer, Fresenius Kabi Deutschland, Kompetenznetz Darmerkrankungen, MSD Sharp Dome, Mylan GmbH, Pfizer GmbH, Janssen Cilag GmbH, Shire Deutschland GmbH, Takeda Pharma GmbH. Schroeder O reports congress expenses from Abbvie, Falk, Janssen, Takeda; speakers’ honoraria from Abbvie, Falk, Janssen, Takeda, Norgine, Pfizer. Dignass A reports consultancy honoraria from Abbott, Pharmacosmos, MSD, Ferring, Janssen, Pfizer, Otsuka, Roche/Genentech, Takeda, Falk; grants from Institut für Gemeinwohl & Stiftung Leben mit Krebs; Lecturing: Falk, Ferring, MSD, Abbott, Janssen, Pfizer, Otsuka, Vifor, Stiftung Leben mit Krebs, Kompetenznetz CED, Takeda, Pharmacosmos; Manuscript preparation: Falk. Education presentations: Abbott, Pharmacosmos, Falk, Ferring. Stein J reports personal fees from Abbvie, personal fees from Dr Schär, personal fees from Falk, personal fees from Ferring, personal fees from Fresenius Kabi, personal fees from Immundiagnostik AG, personal fees from Janssen, personal fees from Medice, personal fees from MSD, personal fees from Pfizer, personal fees from Pharmacosmos, personal fees from Shire, personal fees from Shield, personal fees from Takeda, personal fees from ThermoFisher, personal fees from Vifor Pharma, from NPS, outside the submitted work.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Jürgen Stein, MD, PhD, Chief Physician, Full Professor, Department of Gastroenterology and Clinical Nutrition, DGD Kliniken Sachsenhausen, Teaching Hospital of the JW Goethe University, Schulstr. 31, Frankfurt am Main 60594, Germany. j.stein@em.uni-frankfurt.de
Received: December 25, 2020
Peer-review started: December 25, 2020
First decision: January 9, 2021
Revised: January 24, 2021
Accepted: March 7, 2021
Article in press: March 7, 2021
Published online: June 28, 2021

Abstract

Chronic intestinal failure (CIF) is a rare but feared complication of Crohn’s disease. Depending on the remaining length of the small intestine, the affected intestinal segment, and the residual bowel function, CIF can result in a wide spectrum of symptoms, from single micronutrient malabsorption to complete intestinal failure. Management of CIF has improved significantly in recent years. Advances in home-based parenteral nutrition, in particular, have translated into increased survival and improved quality of life. Nevertheless, 60% of patients are permanently reliant on parenteral nutrition. Encouraging results with new drugs such as teduglutide have added a new dimension to CIF therapy. The outcomes of patients with CIF could be greatly improved by more effective prevention, understanding, and treatment. In complex cases, the care of patients with CIF requires a multidisciplinary approach involving not only physicians but also dietitians and nurses to provide optimal intestinal rehabilitation, nutritional support, and an improved quality of life. Here, we summarize current literature on CIF and short bowel syndrome, encompassing epidemiology, pathophysiology, and advances in surgical and medical management, and elucidate advances in the understanding and therapy of CIF-related complications such as catheter-related bloodstream infections and intestinal failure-associated liver disease.

Key Words: Chronic intestinal failure, Short bowel syndrome, Crohn's disease, Inflammatory bowel disease, Parenteral nutrition, Intestinal failure-associated liver disease

Core Tip: Chronic intestinal failure (CIF) is a rare but feared severe complication of Crohn’s disease, with 60% of patients permanently dependent on parenteral nutrition. This review aims to summarize the knowledge available in the current literature describing recent advances in the management and treatment of adult patients with CIF, with emphasis on patients with Crohn’s disease. Moreover, it aims to further understanding of modern approaches to CIF complications such as catheter-related bloodstream infections and intestinal failure-associated liver disease.



INTRODUCTION

Chronic intestinal failure (CIF) is a rare but feared severe complication of Crohn’s disease (CD). Sixty percent of patients with CD and CIF are permanently dependent on parenteral nutrition (PN). According to the recommendations of the European Society for Clinical Nutrition and Metabolism (ESPEN)[1], CIF is defined as “a reduction of gut function below the minimum necessary for the absorption of macronutrients and/or water and electrolytes, such that intravenous supplementation is required to maintain health and/or growth”. ESPEN suggests classifying CIF on the basis of anatomical, functional, pathophysiological, or clinical characteristics (Figure 1) and has defined five major pathophysiological conditions causing CIF: Short bowel, intestinal fistula, intestinal dysmotility, mechanical obstruction, and extensive mucosal disease[1]. In CD, the most common cause of CIF is short bowel syndrome (SBS), in which the small bowel length, by definition, is less than 200 cm[1]. Concerning nutritional-medicinal management, patients with CIF are most frequently subdivided according to the type and extent of bowel resection and additional surgical procedures (e.g., stomata).

Figure 1
Figure 1 Four domains of chronic intestinal failure. Disease severity is defined by the content of the supplementation — fluid and electrolytes (FE) only, or admixture containing energy (parenteral nutrition (PN)). Each group is subdivided into four categories of mean daily intravenous supplementation (IVS), with volume calculated as average infusion volume per day × number of infusions per week/7.
CLASSIFICATION

According to the recommendations of ESPEN, the classification of CIF can be based on pathophysiological, anatomical, functional, or clinical criteria (Figure 1).

From a pathophysiological point of view, a classification into five types (of which SBS is one) has been proposed based on the presence of various gastrointestinal and/or systemic diseases (Table 1)[2].

Table 1 Pathophysiological classification of intestinal failure (adapted from Pironi et al[2], 2015).
Type
Primary cause
Underlying disease
Short bowel typeQuantitative/qualitative loss of resorptive surfacePost-operative, in patients with mesenteric infarction, Crohn’s disease, radiation enteritis, familial polyposis, abdominal traumata, necrotizing enterocolitis1, bariatric obesity surgery (biliopancreatic diversion with or without duodenal switch), gastroileal anastomosis, extensive tumor resection.
Congenital, in patients with gastroschisis1, intestinal atresia1, intestinal malformation, omphalocele
Fistula typeBypass of resorptive surface due to jejunocolic fistulaInflammatory bowel disease (Crohn’s disease2, diverticulitis, radiation enteritis)
Post-operative3 in patients with neoplasia (colorectal carcinoma, ovarian carcinoma, Crohn’s disease)
Iatrogenic (post-op, percutaneous drainage)
Traumata
Foreign bodies
Dysmotility typeRestricted (insufficient) nutrition intake due to postprandial exacerbation of symptoms, to the point of non-mechanic ileus in severe casesOgilvie syndrome (acute non-mechanic obstruction of the colon)
Chronic intestinal pseudo-obstruction: Primary/idiopathic (neuropathic/myopathic); Secondary (collagenous vascular disease, e.g., PSS, LE, Ehlers-Danlos syndrome; neurological disorders such as Morbus Parkinson, intestinal hypoganglionosis; endocrinopathies)
Obstruction typeReduced nutrition intake; Increased secretion of liquid and electrolytes in obstructive segments of the intestine; Loss of liquids and nutrients due to recurrent vomiting and/or "overflow sensors""Frozen abdomen" in patients with peritoneal carcinomatosis, extensive intestinal adhesion, recurrent peritonitis.
Neoplastic stenoses and/or strictures
Incarceration/strangulation of the intestine (hernia)
Volvulus
Mucosa typeExtensive loss or damage of mucosa results in insufficient resorption of nutrients and pronounced enteral lossMicrovillus inclusion disease1
Tufting enteropathy (intestinal epithelial dysplasia)
Tricho-hepato-enteric syndrome
Autoimmune enteropathy
Intestinal lymphangiectasia
Protein-losing enteropathy (Morbus Waldman)
Radiation enteritis
Chemotherapy-induced/associated enteritis

Anatomically, according to the type and extent of bowel resection and additional surgical procedures (stomata), three different categories of short bowel can be distinguished as prognostic criteria for future disease progression (Figure 2)[1,3]: Type I: Terminal jejunostomy (“very SBS” if remnant bowel length < 50 cm); Type II: Jejunoascendostomy, jejunotransversostomy, or jejunodescendostomy (rarely with colostoma); Type III: Jejunoileotransversostomy (very rarely with colostoma).

Figure 2
Figure 2  Main characteristics of different types of short bowel syndromes according to the anatomical criteria (adapted from Massironi et al[3], 2020).

The functional classification of intestinal failure, first described by Shaffer[4], is based on its time frame, metabolic course, and long-term progression: Type I: Acute and short-term. Usually a self-limiting condition, often observed in the perioperative setting and/or in association with critical illness, where patients require PN for a few days or weeks; Type II: Prolonged acute condition, often arises in patients with metabolically unstable conditions and requires complex multidisciplinary treatment and intravenous supplementation over a period of weeks to months; Type III: Chronic condition found in metabolically stable patients who need to be intravenously substituted for months or years. May be (partially) reversible or irreversible.

The clinical classification is based on the patient's energy and fluid requirements for PN in the context of clinical management. It does not aim at risk stratification and is defined according to 16 categories (Figure 1).

EPIDEMIOLOGY

While intestinal failure can occur due to a number of different underlying conditions, CD has been found to be the most common reason for intestinal failure (Figure 3)[5]. However, incidence of CIF as a complication of CD can only be indirectly evaluated based on data from home PN registries. Recent reports from national intestinal failure units in the United Kingdom show that CD is the underlying disease in 30%–32% of patients with long-term intestinal failure[6,7]. Longitudinal data from Japan indicate an incidence of CIF in CD of 8.5%-18.2% during the first 20 years after initial presentation[8,9].

Figure 3
Figure 3 Underlying diseases of chronic intestinal failure. CIF: Chronic intestinal failure; CIPO: Chronic intestinal pseudo-obstruction; CVID: Common variable immunodeficiency.

Mechanisms of CIF in CD are poorly understood. Among recognized pre-operative risk factors are systemic steroid treatment[10-13], recent weight loss[11,14], intraabdominal abscess[15-17], and smoking[14,18-20]. Whether recent administration of antitumor necrosis factor medication is associated with post-operative morbidity in CD is currently under investigation.

The peri-/post-operative events that most frequently lead to CIF in CD have been purported to be multiple resections, enterocutaneous fistulation, and malabsorption with multiple resections, leading to cumulative loss of the small bowel. However, Soop et al[21], in a longitudinal cohort study of 121 patients referred to a national intestinal failure unit from 2000 to 2018 who were diagnosed with CD and subsequently treated with PN for at least 12 mo, identified septic complications following abdominal surgery to be the most frequent event leading directly to CIF.

ANATOMICAL-PHYSIOLOGICAL ADAPTATION

Three phases of intestinal adaptation can be distinguished. Although it is not possible to define specific biological or functional demarcation points between the hypersecretion, adaptation, and stable (chronically adapted) phases, their differentiation can be a useful aid in therapy planning.

Phase I: Acute (hypersecretory) phase

The hypersecretory phase occurs during the immediate post-operative period and may last for 2 or 3 mo, and in some cases even longer. This phase is characterized by poor absorption of almost all nutrients, including water, electrolytes, proteins, carbohydrates, fats, vitamins, and trace elements. Depending on the extent of resection, daily fluid loss of up to 5 L may occur, while some patients with jejunostomy may lose as much as 6-8 L[22].

Gastric hyperacidity (GH) — increased stomach acidity due to a transient increase in gastric acid secretion (over a period of weeks to months) — affects over half of patients with small bowel resection of 30% or more. GH has been suggested to be caused by the temporary discontinuation of an indirectly or directly acting intestinal inhibitor of gastric secretion (e.g., vasoactive intestinal peptide or gastric inhibitory polypeptide). Alternatively, it may arise due to the diminished breakdown or increased production of a stimulant. Serum levels of gastrin, for example, have been found to be increased after surgery.

Alongside diminished contact time, which leads to inadequate mixing with the chyme, GH-induced inactivation of pancreas ferments is considered vitally important. Following resection of the proximal small bowel, serum levels of cholecystokinin and secretin have been found to be substantially diminished. This generates, in effect, a disturbed positive feedback that leads to a reduction in pancreas stimulation. On the other hand, a reduction in enterokinase activity, observed mainly following duodenum resection, is of no significance.

In patients whose terminal ileum and proximal colon have been surgically removed (type I), a lack of L-cells leads to reduced synthesis of enterohormones [peptide tyrosine-tyrosine, glucagon-like peptide (GLP)-1, GLP-2], effecting an acceleration of gastrointestinal transit duration[23].

Phase II: Adaptation of the residual intestine

Following intestinal resection, structural and functional adaptation processes in the remnant bowel begin even during the early post-operative hypersecretion phase. Distal resection tends to trigger a greater adaptive response than more proximal resection; during this phase, fluid losses should fall to less than 2.5 L. Several years can pass before a stage of maximal adaptation is reached. Generally, 90%-95% of adaptation potential of the remnant bowel is realized within 2 years after resection surgery. In the stabilization phase, improvement of bowel efficiency due to enhanced adaptation leads to a reduction in diarrhea and steatorrhea. Intestinal adaptation is induced by enterotrophic hormones and growth factors, such as epidermal growth factor, growth hormone, insulin-like growth factor-1 (ILG-1) and glucagon-like peptide-2 (GLP-2), an amino acid peptide created by specific post-translational proteolytic cleavage of proglucagon. In addition, intraluminal nutrient supply and the resultant secretion of biliary and pancreatic enzymes play a vital role (for review, see[23]).

Phase III: CIF

Around 50% of patients with prolonged acute intestinal failure go on to develop CIF[24]. Patients with CIF are metabolically stable but need intravenous nutritional support for months or years (reversible CIF) or even lifelong (irreversible CIF). Home-based PN (HPN) is the mainstay of treatment. During this phase, symptoms of SBS are usually addressed with medication. Overall, the probability of weaning off HPN has been reported to be about 50% in adults and up to 73% in children. Complete commutation off HPN in patients with SBS is unlikely[24]. PN can be necessary either to substitute total caloric-nutritional needs when patients are fasting and totally dependent on HPN (total PN) or as supplemental intake in patients who partially satisfy energy requirements via oral/enteral feeding (supplemental PN).

DIAGNOSIS AND WORK-UP

Diagnostics include on the one hand the correct diagnosis of SBS/CIF, i.e., determination of the post-operative bowel anatomy (intraoperative bowel length, radiological assessment of bowel length) and the resorptive capacity of the remnant bowel, and on the other hand, the determination of specific nutrient deficiencies and their symptoms and, if applicable, any complications of enteral nutrition (EN)/PN.

As a first step, diarrhea is quantified and further characterized by stool frequency, stool weight, and 24-h steatorrhea/creatorrhea[25,26]. The resorptive surface area can be calculated using the D-xylose absorption test and/or serum citrulline[27,28]. However, data published on the validity of serum citrulline determination as a biomarker for bowel length and a prognostic parameter for successful intestinal adaptation are the subject of controversy. Assessment of early onset nutrient deficiencies requires analysis of iron (ferritin), folic acid, calcium, phosphate, and copper in serum in addition to urine concentrations of zinc and magnesium. In addition, if steatorrhea is present, serum concentrations of the fat-soluble vitamins (A, D, and E) can be utilized for dose-finding and/or correction (cave: Hypervitaminosis). Symptoms of late onset deficiencies, which often remain unnoticed for years before clinical manifestations become apparent, include megaloblastic anemia as a consequence of vitamin B12 deficiency (the liver’s stores are sufficient for 3-5 years). In this context, and also as an early marker of folic acid or vitamin B6 deficiency, homocysteine levels serve as a simple and efficient screening parameter (Table 2)[29].

Table 2 Recommended laboratory monitoring for patients receiving parenteral nutrition (adapted from Lappas et al[29], 2018).
ParameterFrequency
Initiation of therapy (acute care)
Long-term therapy
Capillary glucoseEvery 6 h until advanced to goal and as needed to maintain 140-180 mg/dLNot routine, as needed basis to coordinate with PN infusion cycle
Basic metabolic panel Phosphorus, magnesiumDaily, until advanced to goal and stable; then 1-2 times/wkWeekly, then decrease frequency as stable
CBC (with differential)Baseline; then 1-2 times/wkMonthly, then decrease frequency as stable
Liver function: ALT, AST, ALP, total bilirubinBaseline; then weeklyMonthly, then decrease frequency as stable
Serum triglyceridesBaseline if at risk; then as neededNot routine, as needed
Iron studies, 25-OH vitamin DNot routine (see Table 4)Baseline, then every 3-6 mo
Zinc, copper, selenium, manganeseNot routine (see Table 4)Baseline, then every 6 mo
MANAGEMENT OF CIF

Management of CIF focuses on optimizing nutritional status and fluid balance and achieving and/or maintaining adequate weight, while simultaneously minimizing risks associated with long-term complications[30]. The choice of therapy is contingent upon the underlying disease, concomitant disorders, and complications of therapy as well as the localization and extent of resection. Both strategies aim to compensate the diminished resorptive capacity of the remnant bowel, striving in the long-term for oral autonomy. For optimal management, ESPEN recommends that patients with CIF are supported by an expert multidisciplinary team (MDT) addressing all relevant domains of care, such as underlying disease therapy, catheter care, psychological well-being, and PN monitoring[1]. Thus, the ideal MDT should include physicians, surgeons, nurses specializing in stoma and catheter care, dieticians, pharmacists, and psychologists[31].

OPTIMIZING NUTRITIONAL STATUS

There is no specific diet for patients with CIF. Individual remnant intestinal anatomy is a crucial consideration when planning nutritional management and should be clarified prior to commencement of PN. As mentioned above, it is recommended that patients with SBS-IF are divided into three groups based on the absence or presence of the terminal ileum and the colon, since these are the main factors influencing the type and amount of nutrient supplementation required[32].

Specific caloric requirements will vary on an individual basis; however, observational studies have found that patients with SBS consume between 35 and 58 kcal/kg/d in order to meet their nutritional needs[33].

Depending on the adaptation stage, nutritional therapy measures can take the form of overlapping or combined therapy with oral and (long-term) PN/EN, with adjunctive medication as necessary. Patients with jejunostoma (type I) and a residual bowel length of less than 100 cm, as well as those with less than 50 cm continual remnant colon (Table 3), are almost certain to require PN on a permanent basis. If, however, a greater length of remnant bowel is present, it is usually possible to gradually discontinue parenteral feeding or at least progressively convert to partial PN or intravenous fluid supplementation[24,32,34].

Table 3 Nutrition therapy (macronutrients) according to presence or absence of colon (modified from Pironi et al[46], 2016).
Nutrient
Colon present (partial/complete)
Colon removed or disabled
Carbo-hydratesSlow increase of the proportion of complex carbohydrates up to approx. 40%-50% of total calorie intake, no low molecular weight sugarsSlowly increase the proportion of complex carbohydrates up to approx. 40%-50% of total calorie intake, no low molecular weight sugars, modified FODMAP diet
ProteinUp to 20%-30% of total energy intakeProtein: up to 20%-30% of total energy intake
FatUp to 20%-30% of total energy intakeFat: up to 40% of total energy intake
Ensure adequate essential fatty acids (high EFA content)Ensure adequate essential fatty acids (high EFA content)
Consider MCTsLimit MCTs
FiberDietary fiber supplements: up to 5-10 g/dDietary fiber supplements: up to 5-10 g/d
OxalateDiet low in oxalic acid and fats; Replacement of up to 50%-75% of dietary fat with MCTsNo restriction of oxalic acid necessary
FluidsEven if liquids are well tolerated, iso- and hypotonic drinks are preferableOral rehydration solutions frequently required
Lactose Low lactose diet (10-12 g)Unrestricted lactose intake

There are no controlled data describing the protein requirements of adults with CIF. Prior studies show that protein absorption depends on the length of the small bowel remnant and may increase over time as the bowel adapts, ranging from 61% to 80%[33]. In patients with IBD, supplementation must additionally compensate protein loss via the bowel[35]. Current ESPEN guidelines recommend protein substitution of 0.8-2.0 g/kg body weight (BW) per day according to individual needs, representing 20%-30% of daily calorie requirements[1]. The supply of proteins (enteral) and/or amino acids (parenteral) should be chosen to offset undermet needs and ensure the prompt correction of existing deficits (catabolism). The sparse data available on the utility of peptide-based EN are contradictory. However, a considerable increase in protein supply can be achieved through continual nasogastric application of standard polymer feeds, either alone or supplementary to oral (ad libidum) nutrition, above and beyond the immediate post-operative phase. Peptide-based enteral feeding is possibly only of additional value in SBS type I[32].

Fat restriction is not necessary in patients who lack a colon. In patients with a preserved colon, it is important to limit fat intake to 20%-25% of total calories in order to reduce the risk of nephrolithiasis from calcium oxalate stones (Table 3). In these patients, a low-fat diet has been shown to increase absorption of calcium, magnesium, and zinc[36] and to improve utilization of medium chain triglycerides for nutrient absorption without increasing stool volume and diminishing calcium and magnesium absorption[37,38]. Fat assimilation can be improved through the addition of pancreas enzyme formulations in powder or granulate form. Controlled studies are not available for CIF[32].

MANAGEMENT OF FLUID AND ELECTROLYTE BALANCE

The primary challenge in patients with jejunostomy is the management of major losses of fluid and electrolytes, particularly zinc, copper, and magnesium (Table 4)[39,40]. It is important for these patients to avoid drinking hypotonic solutions or tap water, as this can lead to worsening electrolyte disturbances by increasing stool sodium content. Conversely, hypertonic solutions such as soda and fruit juice should also be avoided as these solutions are hyperosmolar and can draw water into the gastrointestinal tract and worsen dehydration[33]. Ideally, fluids should be given in small portions between mealtimes, initially in the form of oral rehydration salts (ORS) that offer a sodium-glucose ratio optimized to enhance resorption[41]. In patients with a colon, ORS solutions based on rice starch have been shown to be particularly effective[42]. Sport electrolyte solutions, on the other hand, are not only costly but generally unsuitable, since they contain relatively large amounts of sugar and/or sweeteners in relation to their sodium content. Common sweeteners such as sorbitol or aspartame are known to cause osmotic diarrhea. In addition, fruits, fruit juice, and sweets/candies may increase the frequency of diarrhea. Glutamine-containing ORS solutions are not to be recommended, since glutamine diminishes the intake of sodium and fluids[43].In the adaptation phase (see above), filling foods such as potatoes, rice, oat flakes, or bananas can help solidify the stool. Fluid binding and swelling formulations with pectin may also be beneficial[44].

Table 4 Symptoms, prophylactic supplementation, and therapy of frequent (critical) nutrient deficiencies in chronic intestinal failure.
Nutrient
Symptoms of deficiency
Normal values (blood/serum)
Additional lab tests
Prophylaxis
Therapy in case of deficiency
CalciumNeuromuscular hyperarousal, cardiovascular symptoms, osteopathy2.1-2.7 mmol/L↑Alkaline phosphatase; ↑intact PTH; ↓Bone mineral density Calcium citrate, oral 1200–2000 mg/dBisphosphonate if T-Score < 2.5
MagnesiumNeuromuscular hyperarousal, osteopathy (PTH effect ↓) 0.75-1.15 mmol/L↓Magnesium in urineMagnesium citrate, oral 300 mg/d10 – 15 mmol magnesium, e.g., in 1000 mL NaCl 0.9 %
Vitamin ANight blindness, wound healing disorders1.05-2.80 µmol/L↓Plasma retinol; ↓Retinol binding protein10000-50000 U/d, if liver function normalNo corneal changes: 10000–25000 IU/d oral for 1–2 wk; Corneal changes: 50000–100000 IU i.m. followed by 50000 IU/d i.m. for 2 wk
Vitamin DOsteopathy, wound healing disorders, immune system disorders< 20 µg/L: deficiency; 20-30 g/L: insufficiency; > 30 µg/L: sufficient supplies↑Alkaline phosphatase; ↑intact PTH; ↓Bone mineral density Oral vitamin D (400–800 U/d) [ergocalciferol (vitamin D2) or cholecalciferol (vitamin D3)] or 100000 U/3–6 mo orally50000–150000 IU oral 3-5 times a week; If required: calcitriol [1,25(OH)2D] oral
Vitamin KHemorrhagic diathesisINR < 1.2PIVKA 10 mg/wkN/A
Vitamin B1Polyneuritis (“dry form“), edema, tachycardia, Cardiac insufficiency (“wet form“); Wernicke encephalopathy, ataxia (“central form“)10.64 µg/L↓Thiamine pyrophosphate; ↓Erythrocyte transketolase activityIf vomiting, aggressive oral thiamine supplementation with 100 mg/d for 7–14 dTreatment of Wernicke Encephalopathy: 500 mg i.v. 3 ×/d for 2-3 d; ≥ 250 mg/d i.v. for 5 d; 30 mg oral 2 ×/d
Vitamin B12Megaloblastic anemia, glossitis, skin and mucous membrane pallor, paresthesia, polyneuropathy, funicular myelosis156-675 pmol/L↓Holo-TC; ↓MMA; ↑HomocysteineOral: 1000 μg/wk or 250–350 μg/d, i.m./s.c.: 1000 μg/mo or 3000 μg every 6 mo1000 or 2000 μg/d oral or 1000 μg/wk i.m.
ZincWound healing disorders, hair loss, taste disturbances, predisposition to infection 11-23 mol/L↓Zinc in urineIn presence of fistula, diarrhea or stomata: 12 mg; Otherwise: 3-4 mg30-45 mg (as zinc histidine), 220-440 mg (as zinc sulphate). For each 8–15 mg of elementary zinc, 1 mg copper should be substituted
IronAnemia, hair loss, cognitive disorders, predisposition to infectionCRP < 5 µg/L: > 30 µg/L; CRP ≥ 5 µg/L: ≥ 100 g/L↓Transferrin saturation; ↑Soluble transferrin receptor; ↑Zinc protoporphyrinOral max: 100–150 mg ironParenteral, depending on iron status: Aim: normalization of Hb plus transferrin saturation 35%-50% (calculated according to Ganzoni)
CopperNeutropenia, iron deficiency anemia, central venous development disorders11-22 µmol/L↓Copper/zinc superoxide dismutaseCopper gluconate, oxide or sulphate equivalent to 2 mg elementary copper; 1 mg copper for each 8-15 mg zincCopper sulphate equivalent to 2.4 mg elementary copper in 100 ml 0.9% NaCl i.v. administered one hour/d for 5 d. Subsequently, oral substitution as required

Ostomy patients should be made aware that every intake of food can be expected to result in stool emptying via the ostomy outlet. These patients have a daily fluid requirement of approximately 3 L. Adequate fluid intake can be assessed on the basis of urine volume, with a urine volume of at least 1 L/d considered ideal (urine volume monitoring). Daily intake of 6-9 g salt (e.g., salted meat or vegetable broth) and 25-30 g dietary fiber are also recommended.

Cave: Ileo-/jejunostomy patients (type I) are extremely susceptible to thirst and increased fluid loss due to sweating, diarrhea, etc. The resulting rise in oral fluid intake can lead to increased stool volume. Thus, these patients are at increased risk of rapid dehydration, to the point of prerenal kidney failure.

MINERAL SUBSTITUTION

The electrolyte disturbance most frequently seen in SBS is sodium deficiency. As a rough guide, it can be assumed that about 100 mmol/L of sodium are lost with each liter of jejunostomy fluid. Sodium deficiency is better detected by measuring urinary sodium, which decreases before hyponatremia occurs: Urinary sodium concentration below 10 mmol/L is a diagnostic criterion for sodium depletion. Overall, the aims of treatment are to maintain normal hydration and a daily urine volume of at least 800 mL, with a urinary sodium concentration greater than 20 mmol/L[1,40,45,46].

A urine sodium to potassium ratio of ≤ 1 indicates secondary hyperaldosteronism caused by volume depletion as a result of inadequate fluid substitution[47,48]. This marker therefore requires close monitoring and therapy should seek to maintain a sodium to potassium ratio > 1[49].

While hypokalemia may occur in patients with SBS, net intestinal loss occurs only in the case of end-jejunostomy with less than 50 cm remnant jejunum. The effluent from a jejunostoma with a longer remnant jejunum or an ileostoma contains approximately 15 mmol/L potassium. Low serum potassium is most commonly due to urinary losses of potassium associated with secondary hyperaldosteronism elicited by dehydration and sodium depletion. Hypokalemia may also be caused by magnesium depletion, which disrupts many of the potassium transport mechanisms and increases renal potassium excretion; in this case, hypokalemia is resistant to potassium substitution but responds to magnesium replacement[1,50,51].

Due to potassium loss via the stool or stoma output, daily potassium requirements of patients with CIF are increased compared to healthy individuals, for whom the recommended intake is 1-1.5 mmol/kg BW per day. Potassium loss in patients with small bowel ostomy amounts to approximately 10-30 mmol/L (Table 4)[49].

Daily calcium requirements are estimated at 0.15-0.25 mmol/kg BW (300-400 mg). Calcium intake should be aimed at maintaining normal levels of parathyroid hormone, with calcium excretion in 24 h urine providing a useful additional marker[49]. Extreme depletion of magnesium stores can disrupt parathyroid hormone excretion, leading to false low measurements[52].

Magnesium requirements, estimated at 0.1-0.2 mmol/kg BW (200-300 mg) per day for healthy individuals, can be more than doubled as a consequence of magnesium losses via stool or stoma (Table 4). Magnesium supply should be sufficient to maintain normal serum magnesium levels (0.75-1.15 mmol/L). When interpreting serum magnesium values, it is important to consider that magnesium deficiency may be present even at serum levels of up to 0.85 mmol/L[53]. Magnesium secretion of < 9.7 mg/24 h in urine is regarded as a more sensitive parameter for magnesium deficiency than serum magnesium: 24 h urine levels should therefore be determined when serum magnesium is 0.75-0.85 mmol/L[54]. Since magnesium is not well absorbed, oral magnesium substitution is often inadequate. Nocturnal infusions once or twice a week are recommended, each containing 10-15 mmol magnesium, e.g., in 1000 mL NaCl 0.9%[55]. For oral substitution of calcium or magnesium, Ca2+ or Mg2+ citrate are preferable due to their superior bioavailability[56].

Daily phosphate requirements of healthy persons are estimated at 0.3-0.5 mmol/kg BW. Phosphate supply should be calculated to maintain normal serum phosphate levels while taking account of parathyroid hormone levels (there are no data from controlled trials). When initiating parenteral and/or enteral feeding in undernourished patients, the possibility of nutrition-associated hypophosphatemia must be borne in mind (refeeding syndrome)[57].

MICRONUTRIENTS

The most common micronutrient deficiencies are of vitamin D, zinc, iron, and vitamin B12. These deficiencies can be observed even in patients receiving full (“total”) PN, especially during weaning from PN to EN[58].

Vitamin B12 malabsorption occurs frequently after terminal ileum resection, even after short resection of only 50 cm. Resection of the ileocecal valve reduces intestinal transit duration and increases the risk of bacterial miscolonization of the small bowel, increasing the likelihood of additional bile acid deconjugation and depletion of the vitamin B12 intrinsic factor complex. The resulting pernicious anemia manifests as megaloblastic anemia, thrombocytopenia, and Hunter (atrophic) glossitis, along with neurological disturbances consistent with funicular myelosis (except in folic acid deficiency). To prevent vitamin B12 deficiency, 1000 IE should be given prophylactically every 2-3 mo. In case of manifest deficiency, treatment should commence with daily administration of B12 at a dose of 1000 IE intramuscular/subcutaneous for 5 d, followed by monthly applications of 1000 IE (Table 4).

Deficiencies of fat-soluble vitamins arise as a direct result of disrupted fat resorption. If untreated, these deficiencies can lead to night blindness (vitamin A), coagulation disturbances (vitamin K), and, in the long term, bone metabolism disorders and even osteoporosis (vitamin D)[59] (Table 4).

Trace element deficiencies may also occur, especially of iron, zinc, and copper[58,60,61]. Patients with intestinal failure are prone to iron deficiency as result of malabsorption, gastrointestinal blood loss, and multiple surgical procedures. Accordingly, iron deficiency is the most common micronutrient deficiency during and after transition from TPN to EN, with reported incidences of 60%-80% for iron deficiency and 30%-37% for iron deficiency anemia[62,63]. A study from the Mayo Clinic (Rochester, MI, United States), including 185 patients, showed that iron deficiency anemia developed much more rapidly in patients with fistula and bowel obstruction than in those with SBS and dysmotility[63].

Despite the high prevalence of iron deficiency, iron is not routinely added to PN formulations because of the risk of anaphylaxis and concerns about incompatibilities. Although data describing the compatibility of iron supplementation with parenteral formulations are conflicting[64], iron dextran has been found to be compatible with lipid-free solutions at an amino acid concentration > 2%. A safer approach would prescribe the intermittent infusion of therapeutic iron doses[65]. Dosage requirements for intravenous iron replacement should be calculated according to Evstatiev et al[66].

In patients with diarrhea, large quantities of zinc are lost, with losses of 12 mg zinc per liter stoma output not unusual (Table 4). This considerably exceeds not only normal total zinc requirements but also the content of standard oral mineral supplements[67,68]. In the case of manifest zinc deficiency, 30-45 mg zinc per day can be taken orally (approximately 1 h before breakfast) as zinc histidine or zinc gluconate[69]. However, response to oral supplementation is frequently inadequate, in which case parenteral substitution is indicated[68,70].

PHARMACOLOGICAL TREATMENTS

Analogous to the modified resorption of micro- and macronutrients in CIF, and depending on the underlying disease and the extent and localization of bowel resection, alterations in the bioavailability of any and every kind of drug therapy are to be anticipated. While the scale of these changes is subject to wide interindividual variation, larger drug doses may be required than are typically recommended[30,50]. Data on specific drug classes are scarce, and those that have been published are either case reports or very small cohort studies[71,72]. Therefore, sublingual, transdermal, or transnasal drug application should be chosen whenever suitable options are available[41]. Pharmacological treatments for CIF are primarily based on antisecretory and antimotility treatments intended to minimize gastrointestinal fluid losses. Based on the identification of GLP-2 as a tissue-specific intestinal growth factor in the late 1990s, hormonal treatment promoting intestinal hyperadaptation has been proposed, aimed at maximizing absorption in the remnant bowel, decreasing intestinal losses, and reducing the need for PN[73,74]. Table 5 shows current drug therapy options for the treatment of CIF.

Table 5 Symptomatic treatment options in short bowel syndrome.
Symptom
Drug/therapy
Dose (per day)
Gastral hypersecretionProton pump inhibitors20-40 mg i.v. (p.o.)
Clonidine2 × 75-150 µg s.c./p.o.
Octreotide (sandostatin)13-4 × 50-100 µg s.c.
HypermotilityLoperamide4-6 mg p.o. (max daily dose 16 mg)
Diphenoxylate4 × 2.5-7.5 mg (max daily dose 20-25 mg)
Codeine30 mg p.o.
Opium tincture4 × 0.3-1 mL (10-60 mg) p.o.
Secretory diarrheaOctreotide (sandostatin)12-3 × 50-100 µg s.c.
Budesonide (e.g., entocort)3 × 3 mg p.o.
Clonidine2 × 75-150 µg s.c.
Fat malabsorptionPancrelipase (e.g., Creon)40000 IU with main meals (15000 IU with snacks)
Lactose malabsorptionLactase formulations (L-products)Depending on severity
Reduced fluid resorptionLocust/carob bean gum flour added to drinks (yoghurt)Approx. ½-1 tablespoon per glass/pot
Kaopectate (kaolin/pectin)4 × 1 tablespoon
Antisecretory treatment

For the majority of these medications, only scant scientific evidence is available, or none at all. For example, despite the complete absence of data from controlled studies to endorse the application of proton pump inhibitors in the hypersecretion phase, they are widely recommended as a means of reducing fluid volume and improving the effectiveness of pancreas ferments. Their use in the case of high output stomata is at least supported by two small monocentric studies, whereby intravenous is evidentially superior to oral application[75,76]. Longer-term or permanent intake of proton pump inhibitors is, however, associated with increased risks for osteoporosis and vitamin B12 deficiency[41].

Typical second-line agents used to combat gastric hypersecretion include histamine type 2 receptor (H2) antagonists (e.g., famotidine, ranitidine, cimetidine) and α2-adrenergic receptor agonists (e.g., clonidine)[75,77]. As with antimotility agents, acid suppressors should be initiated at a low dose and titrated upward to yield maximal efficacy with minimal adverse events. The optimal duration of post-operative antacid therapy is unknown, but it should be discontinued in the event of worsening diarrhea[30].

Data from two smaller trials in patients with CD and ileostoma demonstrated that topically acting steroids may improve the absorptive capacity of the intestinal mucosa for water, independently of their anti-inflammatory effects[78,79].

A number of randomized controlled trials (RCTs) have been published examining the efficacy of crofelemer, a dual inhibitor of cyclic adenosine monophosphate- and calcium-mediated chloride secretion, in chronic acquired immunodeficiency syndrome-associated diarrhea (for reviews, see[80,81]).

Antimotility treatment

The use of antimotility drugs to control diarrhea in CIF has been validated mainly by small studies in patients with ileostomy[59,77]. Recommendations for their use are additionally based on extensive practical experience of their application in diarrhea of infectious and non-infectious etiology[49].

Accelerated intestinal motility is typically treated with opioids or the opioid receptor agonists loperamide and diphenoxylate-atropine as the first-line choices. Unlike diphenoxylate, which crosses the blood-brain barrier, loperamide, a peripherally restricted µ-opioid receptor agonist, does not engender undesirable central nervous system effects such as sedation, euphoria, or addiction[30,82,83]. Because loperamide enters the enterohepatic circulation, higher doses (up to 16 tablets/d) may be needed in patients whose ileum has been shortened or removed. Other antimotility agents include codeine[82,84], morphine, and opium tincture[30]. However, these opioids are not restricted to the peripheral nervous system and can thus generate central nervous system effects[30]. When used together, loperamide and codeine may have a synergistic effect[82].

Besides alleviating gastric hypersecretion, clonidine, a centrally acting α2 adrenergic and imidazoline receptor agonist (which can also be administered transdermally), has been shown to diminish stomach and colon motility and inhibit intestinal chloride secretion. As with H2-antagonists, evidence supporting clonidine use in patients with CIF is based only on small case series[85-88]. Dosage of the selected antimotility agent-administered 30 min before meals and at bedtime-should be escalated in a stepwise manner at intervals of 3-5 d until benefit is observed, adverse events occur, or the recommended maximum dosage is reached[30]. Whereas the development of tolerance to analgesic properties of opioids or opioid receptor agonists is well recognized, tolerance to the antidiarrheal effect is rare and the effective dose may remain constant for months to years[30].

Hormonal treatment

In patients with SBS-type CIF, intestinal adaptation can be additionally stimulated by the application of enterotrophic hormones and growth factors, such as growth hormone with or without glutamine or the glucagon-like peptides GLP-1 and GLP-2[89-93]. Treatment with somatostatin analogues has also been studied.

Since the pilot trial of Jeppesen et al[94] in 2003, a number of trials of teduglutide, a long-acting GLP-2 analogue, have shown an overall 20%-40% reduction in PN dependence, with some patients able to discontinue PN entirely. On the evidence of two 24-wk RCTs, teduglutide is recommended to minimize the number of infusion days in patients with stable infusion-dependent CIF. Improvements in stool consistency and general condition of the patients were demonstrated in both of the aforementioned trials[95,96].

Most recently, first results from a European interdisciplinary center-based retrospective data analysis on teduglutide treatment for SBS in clinical practice have been published. The results show that, even when applied in routine medical care to patients with anatomically and clinically heterogeneous SBS-CIF, teduglutide induced functional and structural changes, allowing a gradual reduction of parenteral support. The data suggest that treatment with teduglutide results in an improved intestinal function and a compensatory effect on nutritional status[97].

It must, however, be kept in mind that all existing GLP-2 trials were conducted in patients at a late phase of intestinal adaptation, months or years after surgery. If therapy were initiated immediately after resection, adaptation would probably be induced more quickly (accelerated) and potentially to a greater degree (supraphysiologic). Conversely, and importantly, because GLP-2 is trophic to the intestinal mucosa, it may carry a risk of promoting or inducing the growth of localized polyps, or more ominously, malignancies. A recent systematic review indicated that treatment with teduglutide for up to 30 mo in individuals without known pre-existing cancer did not confer an increased risk of intestinal neoplasia[98]. However, based on animal data showing that GLP-2 may promote the growth of existing neoplasia, endoscopic examination of the remnant colon is mandatory before beginning teduglutide therapy[74].

In 2013, an open-label, placebo-controlled study showed that GLP-1 decreased diarrhea and fecal excretion in SBS patients. More recently, liraglutide, a GLP-1 analogue, was administered daily subcutaneously in patients with end-jejunostomy over 8 wk in an open-label pilot trial[72]. Liraglutide reduced ostomy wet weight output (by 474 ± 563 g/d), increased intestinal wet weight output (by 464 ± 557 g/d), and improved intestinal energy absorption (by 902 ± 882 kJ/d), with statistical significance in all three instances. Combination therapy with GLP-1 and GLP-2 has been shown to be more effective than GLP-2 alone[99].

Over the past two decades, several clinical studies evaluating the effects of growth hormone, alone or in combination with a high-carbohydrate, low-fat diet and/or glutamine, on intestinal adaptation and absorption in pediatric and adult populations, have demonstrated conflicting findings[89,100-107]. The positive effects of high-dose growth hormone treatment, mainly on intestinal water absorption, have been described in patients with SBS with a colon in continuity[74]. Although high-dose growth hormone therapy is already in use, no recommendation for routine application of growth hormone is given in the latest guidelines of either the German Nutrition Society[49] or ESPEN[108].

Octreotide, a long-acting analogue of the peptide hormone somatostatin, has been demonstrated to improve diarrhea in patients with CIF by inhibiting gastrin and other gastrointestinal hormones, by inactivating adenylate cyclase, thereby inhibiting intestinal ion secretion and prolonging intestinal transit time[109-111]. Longer-term application of octreotide is associated with an increased risk of gallstones, a recognized complication of SBS, and impairment of intestinal adaptation[30,112].

COMPLICATIONS AND SPECIAL SITUATIONS
Nephrolithiasis and enteric hyperoxaluria

Enteric hyperoxaluria (EH) was first described as a complication in the early 1970s, upon recognition of the association of small bowel resection and subsequent hyperoxaluria and nephrolithiasis: When significant lengths of the ileum are resected (> 60 cm), bypassed, or dysfunctional due to inflammation (e.g., in CD), both fatty acids and bile acids are delivered to the colon at an increased rate, subsequently complexing calcium and thereby reducing free calcium and increasing free oxalate in the colon.

The management of EH focuses on lowering diet oxalate intake and reducing colonic oxalate absorption. However, many patients find restricting oxalate intake to a therapeutic target level of approximately 50 mg/d (< 10 mg oxalate per mealtime) through avoidance of oxalate-rich foods (e.g., rhubarb, spinach, sorrel, cocoa, chocolate, and cola)[58] difficult on an ongoing basis. Since steatorrhea leads to the complexation of calcium by free fatty acids in the colon, and thus to an increase in free oxalate concentration, reducing fat intake is a useful additional dietary intervention. As an alternative, medium chain fatty acids can be used as a substitute for long chain fatty acids[22,113]. As the mainstay of EH therapy, calcium (1000-1200 mg/d) should be orally substituted. If gastric suppression is required, calcium citrate supplements should be favored over the more commonly-used calcium carbonate salts, since the latter are insoluble at a neutral pH[113].

Choleretic diarrhea/bile acid loss syndrome

Colestyramine has been recommended on the basis of clinical consensus and practical experience to treat choleretic diarrhea associated with compensated bile acid malabsorption (i.e., without significant steatorrhea)[49]. While few controlled studies of colestyramine have been conducted, a recent trial comparing the drug to hydroxypropyl cellulose (which is not a placebo) found colestyramine to have a significantly greater effect. A systematic review demonstrated a dose-response relationship between severity of bile acid diarrhea and treatment response, ranging from 96% in patients with severe to 70% in patients with mild bile acid diarrhea[114].

Newer bile acid sequestrants such as colestipol and colesevelam, which are much more selective bile acid binders than colestyramine[115], are reportedly associated with better compliance and fewer side effects[116,117]. The efficacy of colesevelam has now been confirmed in a first RCT[118]. Neither colesevelam nor colestyramine is suitable for the treatment of uncompensated bile acid loss syndrome, since the associated steatorrhea is exacerbated by both compounds (Table 6).

Table 6 Major complications of short bowel syndrome: risk factors, prevention and treatment (adapted from Pironi et al[46], 2016).

Risk factors
Prevention and/or treatment
Bacterial overgrowth/ miscolonizationIleocecal valve resection; Reduced intestinal motility (Ogilvie syndrome; chronic intestinal pseudo-obstruction)Metronidazole (500 mg, 2 times per day), vancomycin (125 mg, 4 times per day), neomycin (500 mg, 3 times per day), clindamycin (300 mg, 3 times per day) tetracycline (500 mg, 3 times per day), rifaximin (550 mg, 2 times per day)
Renal failureDehydration; CRBSI; Nephrocalcinosis; Kidney stonesOptimize fluid and sodium balance; Optimize CVC care; Prevent urinary calcium oxalate formation
Calcium oxalate, kidney stonesSBS with colon in continuity and fat malabsorption (enteric hyperoxaluria); Pyridoxine or thiamine deficiency; Excess of ascorbic acid; Dehydration; Low urinary citrate; Low urinary magnesiumReduce or avoid excess lipid in the diet; Reduce food with high oxalate content; Oral calcium at mealtime (1 g); Oral cholestyramine; Optimize fluid balance; Optimize acid-base balance; Optimize magnesium status; Limit ascorbic acid supplementation
BAMS
—CompensatedExtent of resection < 100 cm; Fecal bile acid excretion increased; Adequate hepatic compensation of bile acid loss; ≥ reduction of bile acid pool; no or minimal steatorrheaColestyramine/Colesevelam
—DecompensatedExtent of resection > 100 cm, fecal bile acid excretion increased; Inadequate hepatic compensation of bile acid loss; ≥ reduction of bile acid pool ≥ steatorrheaFat-modified/-reduced diet; Cholylsarcosine/ox gall1
GallstonesProlonged oral fasting; Interrupted bile acid entero-hepatic circulation; Prolonged treatment with anticholinergic and narcotic drugsLimit periods of oral fasting; Limit narcotic or anticholinergic treatment; Use oral and/or enteral feeding as much as possible
IFALD-cholestasisSBS with < 50 cm of residual small bowel; SBS without colon; CRBSI episodes; Chronic intraabdominal inflammation and/or small bowel bacterial overgrowth; Interrupted enterohepatic circulation of bile acid; Oral fasting; PN-overfeeding; i.v. soya-based lipid emulsion ≥ 1 g/kg/dAvoid oral fasting; Optimize CVC care; Treat intraabdominal inflammation foci; Rehabilitative surgical procedures; Optimize i.v. feeding; i.v. soya-based lipid emulsion < 1 g/kg/d and/or i.v. fish oil lipid emulsion
D-lactic acid acidosisSBS with a colon in continuity; Carbohydrate and soluble fiber-based diet; Ingestion of rapidly fermentable simple sugars; Feeding D-lactate containing food; High blood and urinary oxalate; Thiamine deficiency; Antibiotic and/or probiotic courses; Dehydration; Decreased renal function; Decreased liver functionLow carbohydrate and simple sugar diet; Antibiotics active against D-lactate-producing bacteria orally, such as metronidazole (500 mg, 2 times per day), vancomycin (125 mg, 4 times per day), neomycin (500 mg, 3 times per day), clindamycin (300 mg, 3 times per day), tetracycline (500 mg, 3 times per day), rifaximin (550 mg, 2 times a day); Thiamine supplementation; Reduction of oxalate absorption; Optimize fluid balance

A promising therapeutic approach in the treatment of (decompensated) bile acid loss syndrome is the administration of cholylsarcosine, a conjugated bile acid that has no secretagogue effects. Smaller studies showed a significant increase in calcium and fat absorption in patients treated daily with 4-6 g cholylsarcosine[119-121].

A recent proof-of-concept study assessed the usefulness of obeticholic acid, a potent farnesoid x receptor agonist, in patients with bile acid diarrhea, and found statistically significant increases in fasting serum FGF19, both in patients with idiopathic bile acid diarrhea and in those with secondary disease and ileal resection of less than 45 cm[122].

Gallstones

Gallstones have been reported in 31%-45% of patients with SBS (with or without a colon). Reported risk factors are prolonged periods of starvation, large fluctuations in BW, medications such as opiates, and lipid emulsions (in TPN). In addition, diseases affecting the ileum (e.g., CD) and absence of ileum and/or ileocecal valve following surgical resection alter enterohepatic circulation and cause a loss of bile salts (Table 7), thus leading to cholesterol supersaturation and sludge formation[123-125].

Table 7 Pathophysiological characteristics of short bowel syndrome with and without colon in continuity (adapted from Pironi et al[46], 2016).
Characteristic
End-jejunostomy
Jejunocolic or jejunoileal anastomosis
Structural and functional adaptation, to increase nutrient absorptionNo evidence thereof at any time after surgeryPossible up to 2 yr after surgery
Gastric hypersecretion (up to 6 mo after resection)PresentPresent
Gastric emptying and small bowel transitAccelerated gastric emptying for liquidsSlowed
Accelerated small bowel transit
GI hormone secretion (PYY, GLP-1, GLP-2)Decreased/absentIncreased
Energy absorption from microbiota SCFA, production in the colonAbsentIncreased up to 1000 kcal (4.2 MJ) per day
Water and sodium absorption in the remnant small bowelPossible “net secretion” when jejunum length < 100 cm (more fluid and sodium lost than ingested)Colon adaptation can increase the absorption of water up to 6 liters and sodium up to 800 mmol per day
Vitamin B12 and bile salt absorptionAbsentPartially conserved or absent
Magnesium absorptionDecreasedDecreased
Remnant small bowel cut-off length for HPN weaning> 115 cmJejunocolic anastomosis > 60 cm
Jejunoileal anastomosis with ICV and entire colon
> 35 cm
Small intestinal bacterial overgrowth

Especially after ileocecal valve resection, patients with CIF are at risk of small intestinal bacterial overgrowth (SIBO), i.e., the miscolonization of the upper small bowel by colon-derived bacteria. By promoting bile acid deconjugation, SIBO not only exacerbates steatorrhea, thereby disrupting the absorption of fat and fat-soluble vitamins[126,127], but also hinders intestinal adaptation[127,128]. SIBO is also perceived as a predisposing factor for intestinal failure-associated liver disease (IFALD), since hepatotoxins (e.g., lithocholic acid) produced by anaerobic bacteria in the small bowel may lead to hepatic injury. While diagnosis can be made using H2 or C13 breath tests, their sensitivity is not very high[127]. Some authors therefore suggest a “therapeutic trial” approach on the basis of clinical suspicion, even in non-responders[62,129,130]. Adjunctive intermitting-alternating antibiotic therapy should be administered for a period of 7-10 d (Table 6)[63].

D-lactic acidosis

D-lactic acidosis is a rare, but often overlooked, condition observed only in patients with a preserved colon. It is caused by an increased intake of refined carbohydrates that, having been broken down by bacteria into short-chain fatty acids, lactate when they pass into the colon. The associated decrease in colonic pH promotes the growth of gram-positive, acid-resistant, D-lactate producing anaerobes (e.g., Bifidobacterium, Lactobacillus, Eubacteriaceae). This results in insufficient metabolism of human D-lactate, and D-lactic acidosis occurs, with associated neurological symptoms such as vision disturbances, confusion, and gait insecurity. Such symptoms are often mistakenly assumed to be signs of alcohol abuse. Diagnosis is made by determining D-lactate levels in the blood[131,132]. The pathogenesis of neurological symptoms associated with D-lactate acidosis remains unclear. One hypothesis is that D-lactate itself is toxic to the brain or alters neuro-transmitter production. Other potentially neurotoxic substances or false neurotransmitters produced in variable quantities during periods of D-lactate elevation may also be involved[46] (Table 6). Due to the similarity of symptoms of D-lactic acidosis to those of Wernicke encephalopathy, prophylactic administration of thiamine has been recommended[131,132].

Diseases of the bone metabolism

Diseases of the bone metabolism, such as osteoporosis and osteomalacia, may occur as long-term complications of TPN and have been reported to occur in 40%-100% of patients with CIF[133-135]. The main causal factors include not only malabsorption of calcium, magnesium[136], and vitamin D but also the presence of underlying inflammatory disease (e.g., CD[137]), combined, as a rule, with many years of steroid intake[135]. Patients on long-term HPN are recommended to undergo bone density testing every 2 years, with frequent assessment of calcium and phosphorus balance, as well as vitamin D levels. Vitamin D substitution should be orientated at 25-hydroxy levels > 30 ng/mL[138].

MANAGEMENT OF THERAPY-RELATED COMPLICATIONS

Therapy-related complications most commonly arise from long-term HPN. While catheter-associated complications are frequent, a variety of organic and metabolic disorders may also occur (for detailed review, see[139]).

Catheter-associated complications

Direct catheter-associated complications in patients receiving long-term HPN can be characterized as either catheter occlusion or thrombotic or infectious complications.

Catheter occlusion can be partial or complete. Factors that influence its occurrence include duration of catheter placement, catheter size and material, type and meticulousness of care of the central venous access route, and the composition of applied infusion solutions. Signs and symptoms of occlusion include resistance when flushing, sluggish flow, inability to infuse fluids, frequent occlusion alarm on the infusion pump, infiltration, extravasation, swelling or leaking at the insertion site upon infusion or during flushing, inability to withdraw blood, and sluggish blood return upon aspiration of blood[140-143].

Possible causes of catheter occlusion include: (1) mechanical problems; (2) non-thrombotic obstruction (precipitate of formulations such as medications or PN constituents within the catheter lumen); and (3) thrombotic obstruction (clot, thrombus, or fibrin deposition).

Any obvious mechanical obstruction should be ruled out before checking for non-thrombotic and thrombotic occlusion[144]. Mechanical occlusions arise from internal or external problems with the catheter, such as kinking in the catheter or tubing, catheter migration or malpositioning, clogging of the cap/needleless connector or filter, or excessive tightness of the retaining suture[145]. Depending on its underlying cause(s), mechanical occlusion can be remedied by removing the venous catheters, avoiding twisting/kinking when dressing, or, if occlusion or malpositioning of the needle is suspected, replacing the non-coring needle.

Catheter-associated infections (CAI) — in particular, catheter-related bloodstream infections (CRBSIs) — remain the "Achilles’ heel" of HPN treatment. Infections are classified as follows: Local infections: Catheter exit site infections, port recess infections, and tunnel infections in long-term subcutaneous tunnel catheters (Hickmann catheters); CRBSI: Defined as an infection in which the same organism can be isolated in cultures from the catheter and from peripheral blood, and clinical symptoms of sepsis are present, in the absence of an alternative focus of infection. CRBSI occurs in association with 5%-10% of all central venous catheters, equivalent to 0.3-30 cases/1000 catheter d, and accounts for approximately 70% of all home PN-related hospital admissions[146-148].

More than 50% of all CAIs are caused by gram-positive pathogens, 33% by coagulase-negative staphylococci, and approximately 20%-22% by pathogenic fungi. About a quarter are mixed infections[149]. The chances of saving an infected (tunneled) catheter vary from 50% to 85%, depending on the pathogen responsible[49].

Different catheter-locking solutions have been studied for their effectiveness in preventing CRBSIs, including antiseptic agents, antibiotics, and anticoagulants. Due to the supposed need for an anticoagulant, heparin has most commonly been used as a catheter-locking solution. Its use, however, is no longer recommended, as it has been shown to potentially increase the risk of CRBSI by promoting the formation of an intraluminal biofilm[1,150,151].

Based on its ability to prevent microbial adhesion to the inner catheter surface, and to destroy microbial cell membranes and toxins, use of the broad-spectrum antiseptic agent taurolidine as a catheter-locking solution was described in 1998. Subsequent studies and a meta-analysis confirmed that, compared with heparin, taurolidine decreases CRBSI incidence in patients with catheters. Based on these data and the most recently published RCT by Wouters et al[152], and in view of its favorable safety and cost profile, taurolidine solution (e.g., 0.2%) can be applied as a secondary prophylactic locking solution as soon as infection occurs and may also be used for primary CRBSI prophylaxis. However, effective training of the patient (and/or their family) and nursing staff is recognized as the single most effective primary prophylaxis[1,153].

IFALD

Based on the multifactorial nature of IFALD, a recent international position paper suggested defining it as “liver injury as a result of one or more factors relating to intestinal failure including, but not limited to, PN and occurring in the absence of another primary parenchymal liver pathology”[154,155]. Based on retrospective data in adult patients with CIF, Sasdelli et al[156] recently reported an IFALD prevalence of 13%-40% for cholestasis, 27%-90% for steatosis, 2%-5% for fibrosis, and 8%-75% for unclassified IFALD, depending on the criteria adopted.

The pathogenesis of IFALD is not yet fully understood and likely multifactorial, with both PN and patient-dependent associated factors playing a role (Table 8)[123,157]. Recommended measures for the prevention and therapy of IFALD are[158,159]: (1) maintenance of suitable proportions of nutrients, avoiding in particular high-calorie and high-glucose feeds, and insulin application, with 20% up to maximum 50% of total energy intake as lipids (approximately 1g fat/kg BW); (2) cyclical infusion feeding periods of 12-16 h, maintaining an overnight fasting period of 8-12 h; (3) enteral feeding, even if the quantities are insufficient to cover requirements, e.g., as a supplement; (4) in patients with cholestasis and/or “sludge” production, administration of ursodesoxycholic acid (10-15 mg/kg BW, e.g., as juice) or cholecystokinin (cave: possibility of anaphylactoid reaction), with additional dose reduction of fat-soluble vitamins, copper, and manganese; and (5) administration of “essential” substrates such as carnitine, taurine, or choline in infants and children. On the basis of affirmative results from a number of randomized studies, fish oil-based lipid solutions are recommended for IFALD therapy in children. Furthermore, an initial study showed a positive effect of fish oil on hepatopathy in adults with CIF[158] (for review, see[160]). Medium-chain triglyceride- and/or olive oil-based lipid solutions are not recommended either for primary or secondary prophylaxis of IFALD[158]. The same applies to probiotics.

Table 8 Intestinal failure-associated liver disease risk factors in adult patients with home-based parenteral nutrition (adapted from Van Gossum et al[157], 2019).
PN-related
Patient-related
Energy overfeedingLack of oral feeding
Glucose overload > 7 mg/kg/minShort bowel syndrome (small bowel remnant < 50 cm)
Lipid emulsion overloadInflammation/infection
Soya-based lipid emulsion > 1 g/kg/dSepsis (e.g., central venous catheter related)
Continuous infusion (24/24 h)Small intestinal bacterial overgrowth
Contaminants (phytosterols)Gut inflammation
Antioxidant deficiencyViral infection (e.g., hepatitis B, C)
Nutrient deficiency (choline, carnitine, methionine, taurine, essential fatty acid deficiency, etc.)Autoimmune
Hepatotoxic medications
SURGICAL PROCEDURES
Reconstructive surgery

A range of operative techniques have been developed over the past years to allow patients with CIF at least the possibility of enteral feeding. These techniques have two common goals: To increase gastrointestinal transit duration and to increase the resorptive surface of the bowel. In principle, three surgical procedures may be applied (for review, see[161-163]): (1) in cases where bowel length is critical, longitudinal lengthening and tailoring, first described by Bianchi in 1980, accomplishes intestinal tapering without loss of surface; (2) in the serial transverse enteroplasty procedure described by Kim et al[164] in 2003, the intestinal lumen is narrowed by firing a series of staples perpendicularly to the long axis of the bowel in a zig-zag pattern; (3) if the remaining small bowel is too short (i.e., < 70-80 cm) to achieve weaning from TPN, a reversed antiperistaltic intestinal segment (10-15 cm in adults) can be placed to slow intestinal transit and thereby enhance nutrient and fluid absorption.

Intestinal transplantation

Intestinal transplantation, alone or in combination with other organs (visceral transplantation), is reserved for patients with life-threatening complications of PN defined by significant liver injury with elevated hepatic enzymes, multiple line infections, a single episode of life-threatening catheter-related sepsis, thrombosis of two of the central veins, or frequent episodes of dehydration (for review, see[165]).

CONCLUSION

While considerable advances have been made in recent years with regard to the understanding and management of CIF and its various related complications, it remains a challenging disorder. Home-based PN has brought distinct benefits, particularly in terms of higher survival rates and improved patient quality of life. Nevertheless, more than half of patients with CIF require PN on a permanent basis. While the traditional mainstays of pharmacological management are antimotility and antisecretory drugs, new hormonally acting drugs that stimulate intestinal adaptation, such as teduglutide, have had encouraging results, adding a new dimension to CIF therapy. More work is needed to improve understanding of CIF mechanisms, and to advance preventive and therapeutic approaches. Optimization of patient outcomes in complex CIF requires a dedicated multidisciplinary team of physicians, dietitians, and nurses, whose foci of care include maximizing intestinal rehabilitation, providing nutritional support, and improving quality of life.

ACKNOWLEDGEMENTS

The authors would like to thank Janet Collins (Interdisziplinäres Crohn-Colitis Centrum Rhein-Main, Frankfurt, Germany) for editing, correcting, and proof-reading the manuscript.

Footnotes

Manuscript source: Unsolicited manuscript

Corresponding Author's Membership in Professional Societies: American Gastroenterological Association; and Fellow of the European Board of Gastroenterology.

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Germany

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Picarelli A S-Editor: Zhang H L-Editor: Filipodia P-Editor: Ma YJ

References
1.  Pironi L, Arends J, Bozzetti F, Cuerda C, Gillanders L, Jeppesen PB, Joly F, Kelly D, Lal S, Staun M, Szczepanek K, Van Gossum A, Wanten G, Schneider SM;  Home Artificial Nutrition & Chronic Intestinal Failure Special Interest Group of ESPEN. ESPEN guidelines on chronic intestinal failure in adults. Clin Nutr. 2016;35:247-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 401]  [Cited by in F6Publishing: 445]  [Article Influence: 55.6]  [Reference Citation Analysis (0)]
2.  Pironi L, Arends J, Baxter J, Bozzetti F, Peláez RB, Cuerda C, Forbes A, Gabe S, Gillanders L, Holst M, Jeppesen PB, Joly F, Kelly D, Klek S, Irtun Ø, Olde Damink SW, Panisic M, Rasmussen HH, Staun M, Szczepanek K, Van Gossum A, Wanten G, Schneider SM, Shaffer J;  Home Artificial Nutrition & Chronic Intestinal Failure;  Acute Intestinal Failure Special Interest Groups of ESPEN. ESPEN endorsed recommendations. Definition and classification of intestinal failure in adults. Clin Nutr. 2015;34:171-180.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 346]  [Cited by in F6Publishing: 357]  [Article Influence: 35.7]  [Reference Citation Analysis (0)]
3.  Massironi S, Cavalcoli F, Rausa E, Invernizzi P, Braga M, Vecchi M. Understanding short bowel syndrome: Current status and future perspectives. Dig Liver Dis. 2020;52:253-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 71]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
4.  Shaffer J. Intestinal failure: definition and service development. Clin Nutr. 2002;21:144e145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
5.  Pironi L, Konrad D, Brandt C, Joly F, Wanten G, Agostini F, Chambrier C, Aimasso U, Zeraschi S, Kelly D, Szczepanek K, Jukes A, Di Caro S, Theilla M, Kunecki M, Daniels J, Serlie M, Poullenot F, Wu J, Cooper SC, Rasmussen HH, Compher C, Seguy D, Crivelli A, Pagano MC, Hughes SJ, Guglielmi FW, Kozjek NR, Schneider SM, Gillanders L, Ellegard L, Thibault R, Matras P, Zmarzly A, Matysiak K, Van Gossum A, Forbes A, Wyer N, Taus M, Virgili NM, O'Callaghan M, Chapman B, Osland E, Cuerda C, Sahin P, Jones L, Lee ADW, Bertasi V, Orlandoni P, Izbéki F, Spaggiari C, Díez MB, Doitchinova-Simeonova M, Garde C, Serralde-Zúñiga AE, Olveira G, Krznaric Z, Czako L, Kekstas G, Sanz-Paris A, Jáuregui EP, Murillo AZ, Schafer E, Arends J, Suárez-Llanos JP, Shaffer J, Lal S. Clinical classification of adult patients with chronic intestinal failure due to benign disease: An international multicenter cross-sectional survey. Clin Nutr. 2018;37:728-738.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 94]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
6.  Dibb M, Soop M, Teubner A, Shaffer J, Abraham A, Carlson G, Lal S. Survival and nutritional dependence on home parenteral nutrition: Three decades of experience from a single referral centre. Clin Nutr. 2017;36:570-576.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 66]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
7.  Lloyd DA, Vega R, Bassett P, Forbes A, Gabe SM. Survival and dependence on home parenteral nutrition: experience over a 25-year period in a UK referral centre. Aliment Pharmacol Ther. 2006;24:1231-1240.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 107]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
8.  Watanabe Y, Miyoshi N, Fujino S, Takahashi H, Haraguchi N, Hata T, Matsuda C, Yamamoto H, Doki Y, Mori M, Mizushima T. Cumulative Inflammation Could Be a Risk Factor for Intestinal Failure in Crohn's Disease. Dig Dis Sci. 2019;64:2280-2285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
9.  Watanabe K, Sasaki I, Fukushima K, Futami K, Ikeuchi H, Sugita A, Nezu R, Mizushima T, Kameoka S, Kusunoki M, Yoshioka K, Funayama Y, Watanabe T, Fujii H, Watanabe M. Long-term incidence and characteristics of intestinal failure in Crohn's disease: a multicenter study. J Gastroenterol. 2014;49:231-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
10.  Fumery M, Seksik P, Auzolle C, Munoz-Bongrand N, Gornet JM, Boschetti G, Cotte E, Buisson A, Dubois A, Pariente B, Zerbib P, Chafai N, Stefanescu C, Panis Y, Marteau P, Pautrat K, Sabbagh C, Filippi J, Chevrier M, Houze P, Jouven X, Treton X, Allez M;  REMIND study group investigators. Postoperative Complications after Ileocecal Resection in Crohn's Disease: A Prospective Study From the REMIND Group. Am J Gastroenterol. 2017;112:337-345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 121]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
11.  Alves A, Panis Y, Bouhnik Y, Pocard M, Vicaut E, Valleur P. Risk factors for intra-abdominal septic complications after a first ileocecal resection for Crohn's disease: a multivariate analysis in 161 consecutive patients. Dis Colon Rectum. 2007;50:331-336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 234]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
12.  Yamamoto T, Allan RN, Keighley MR. Risk factors for intra-abdominal sepsis after surgery in Crohn's disease. Dis Colon Rectum. 2000;43:1141-1145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 260]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
13.  Tzivanakis A, Singh JC, Guy RJ, Travis SP, Mortensen NJ, George BD. Influence of risk factors on the safety of ileocolic anastomosis in Crohn's disease surgery. Dis Colon Rectum. 2012;55:558-562.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 76]  [Article Influence: 6.3]  [Reference Citation Analysis (1)]
14.  Neary PM, Aiello AC, Stocchi L, Shawki S, Hull T, Steele SR, Delaney CP, Holubar SD. High-Risk Ileocolic Anastomoses for Crohn's Disease: When Is Diversion Indicated? J Crohns Colitis. 2019;13:856-863.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
15.  Lau C, Dubinsky M, Melmed G, Vasiliauskas E, Berel D, McGovern D, Ippoliti A, Shih D, Targan S, Fleshner P. The impact of preoperative serum anti-TNFα therapy levels on early postoperative outcomes in inflammatory bowel disease surgery. Ann Surg. 2015;261:487-496.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 132]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
16.  Huang W, Tang Y, Nong L, Sun Y. Risk factors for postoperative intra-abdominal septic complications after surgery in Crohn's disease: A meta-analysis of observational studies. J Crohns Colitis. 2015;9:293-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 115]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
17.  Brouquet A, Maggiori L, Zerbib P, Lefevre JH, Denost Q, Germain A, Cotte E, Beyer-Berjot L, Munoz-Bongrand N, Desfourneaux V, Rahili A, Duffas JP, Pautrat K, Denet C, Bridoux V, Meurette G, Faucheron JL, Loriau J, Guillon F, Vicaut E, Benoist S, Panis Y;  GETAID chirurgie group. Anti-TNF Therapy Is Associated With an Increased Risk of Postoperative Morbidity After Surgery for Ileocolonic Crohn Disease: Results of a Prospective Nationwide Cohort. Ann Surg. 2018;267:221-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 91]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
18.  Sørensen LT, Jørgensen T, Kirkeby LT, Skovdal J, Vennits B, Wille-Jørgensen P. Smoking and alcohol abuse are major risk factors for anastomotic leakage in colorectal surgery. Br J Surg. 1999;86:927-931.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 247]  [Article Influence: 9.9]  [Reference Citation Analysis (0)]
19.  Morar PS, Hodgkinson JD, Thalayasingam S, Koysombat K, Purcell M, Hart AL, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 76]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
20.  Lin YS, Cheng SW, Wang YH, Chen KH, Fang CJ, Chen C. Systematic review with meta-analysis: risk of post-operative complications associated with pre-operative exposure to anti-tumour necrosis factor agents for Crohn's disease. Aliment Pharmacol Ther. 2019;49:966-977.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
21.  Soop M, Khan H, Nixon E, Teubner A, Abraham A, Carlson G, Lal S. Causes and Prognosis of Intestinal Failure in Crohn's Disease: An 18-year Experience From a National Centre. J Crohns Colitis. 2020;14:1558-1564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
22.  Baker ML, Williams RN, Nightingale JM. Causes and management of a high-output stoma. Colorectal Dis. 2011;13:191-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 125]  [Cited by in F6Publishing: 109]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
23.  Tappenden KA. Intestinal adaptation following resection. JPEN J Parenter Enteral Nutr. 2014;38:23S-31S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 164]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
24.  Ukleja A. Weaning from Parenteral Nutrition. Gastroenterol Clin North Am. 2019;48:525-550.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
25.  Stein J, Purschian B, Bieniek U, Caspary WF, Lembcke B. Near-infrared reflectance analysis: a new dimension in the investigation of malabsorption syndromes. Eur J Gastroen Hepat. 1994;6:889-894.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
26.  Picarelli A, Greco M, Di Giovambattista F, Ramazzotti A, Cedrone C, Corazziari E, Torsoli A. Quantitative determination of faecal fat, nitrogen and water by means of a spectrophotometric technique: near infrared reflectance analysis (NIRA). Assessment of its accuracy and reproducibility compared with chemical methods. Clin Chim Acta. 1995;234:147-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
27.  Jeppesen PB, Gabe SM, Seidner DL, Lee HM, Olivier C. Citrulline correlations in short bowel syndrome-intestinal failure by patient stratification: Analysis of 24 weeks of teduglutide treatment from a randomized controlled study. Clin Nutr. 2020;39:2479-2486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
28.  Crenn P, Messing B, Cynober L. Citrulline as a biomarker of intestinal failure due to enterocyte mass reduction. Clin Nutr. 2008;27:328-339.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 280]  [Cited by in F6Publishing: 281]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
29.  Lappas BM, Patel D, Kumpf V, Adams DW, Seidner DL. Parenteral Nutrition: Indications, Access, and Complications. Gastroenterol Clin North Am. 2018;47:39-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
30.  Kumpf VJ. Pharmacologic management of diarrhea in patients with short bowel syndrome. JPEN J Parenter Enteral Nutr. 2014;38:38S-44S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
31.  Dibb M, Lal S. Monitoring long-term parenteral nutrition. Curr Opin Gastroenterol. 2019;35:119-125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 6]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
32.  Jeppesen PB, Fuglsang KA. Nutritional Therapy in Adult Short Bowel Syndrome Patients with Chronic Intestinal Failure. Gastroenterol Clin North Am. 2018;47:61-75.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
33.  Austin K, Bonnes S, Daniel H. Controversy in Nutrition Recommendations for Short Bowel Syndrome: How Type of SBS Impacts Response. Curr Gastroenterol Rep. 2019;21:64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
34.  Ukleja A, Scolapio JS, Buchman AL. Nutritional management of short bowel syndrome. Semin Gastrointest Dis. 2002;13:161-168.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Levitt DG, Levitt MD. Protein losing enteropathy: comprehensive review of the mechanistic association with clinical and subclinical disease states. Clin Exp Gastroenterol. 2017;10:147-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 79]  [Article Influence: 11.3]  [Reference Citation Analysis (1)]
36.  Hessov I, Andersson H, Isaksson B. Effects of a low-fat diet on mineral absorption in small-bowel disease. Scand J Gastroenterol. 1983;18:551-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 25]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
37.  Nightingale JM, Lennard-Jones JE, Gertner DJ, Wood SR, Bartram CI. Colonic preservation reduces need for parenteral therapy, increases incidence of renal stones, but does not change high prevalence of gall stones in patients with a short bowel. Gut. 1992;33:1493-1497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 194]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
38.  Jeppesen PB, Mortensen PB. Significance of a preserved colon for parenteral energy requirements in patients receiving home parenteral nutrition. Scand J Gastroenterol. 1998;33:1175-1179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 45]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
39.  Miranda SC, Ribeiro ML, Ferriolli E, Marchini JS. Hypomagnesemia in short bowel syndrome patients. Sao Paulo Med J. 2000;118:169-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
40.  Tappenden KA. Pathophysiology of short bowel syndrome: considerations of resected and residual anatomy. JPEN J Parenter Enteral Nutr. 2014;38:14S-22S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 110]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
41.  Nightingale JM, Lennard-Jones JE, Walker ER, Farthing MJ. Oral salt supplements to compensate for jejunostomy losses: comparison of sodium chloride capsules, glucose electrolyte solution, and glucose polymer electrolyte solution. Gut. 1992;33:759-761.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 47]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
42.  Atia AN, Buchman AL. Oral rehydration solutions in non-cholera diarrhea: a review. Am J Gastroenterol. 2009;104:2596-604; quiz 2605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 56]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
43.  Buchman AL, Scolapio J, Fryer J. AGA technical review on short bowel syndrome and intestinal transplantation. Gastroenterology. 2003;124:1111-1134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 376]  [Cited by in F6Publishing: 301]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
44.  Leiss O. [Clinical management of patients with short bowel syndrome]. Z Gastroenterol. 2005;43:661-675.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 2]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
45.  Jeppesen PB. Spectrum of short bowel syndrome in adults: intestinal insufficiency to intestinal failure. JPEN J Parenter Enteral Nutr. 2014;38:8S-13S.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 110]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
46.  Pironi L. Definitions of intestinal failure and the short bowel syndrome. Best Pract Res Clin Gastroenterol. 2016;30:173-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 140]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
47.  Ladefoged K, Olgaard K. Sodium homeostasis after small-bowel resection. Scand J Gastroenterol. 1985;20:361-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 38]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
48.  Lauverjat M, Hadj Aissa A, Vanhems P, Boulétreau P, Fouque D, Chambrier C. Chronic dehydration may impair renal function in patients with chronic intestinal failure on long-term parenteral nutrition. Clin Nutr. 2006;25: 75-81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 47]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
49.  Lamprecht G, Pape UF, Witte M, Pascher A;  Committee udDS. S3-Leitlinie der Deutschen Gesellschaft für Ernährungsmedizin e.V. in Zusammenarbeit mit der AKE, der GESKES und der DGVS Klinische Ernährung in der Gastroenterologie (Teil 3) – Chronisches Darmversagen. Aktuel Ernahrungsmed. 2014;39:e57-e71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
50.  Nightingale J, Woodward JM;  Small Bowel and Nutrition Committee of the British Society of Gastroenterology. Guidelines for management of patients with a short bowel. Gut. 2006;55 Suppl 4:iv1-i12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 160]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
51.  Jeppesen PB, Mortensen PB. Intestinal failure defined by measurements of intestinal energy and wet weight absorption. Gut. 2000;46:701-706.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 119]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
52.  Martin KJ, González EA, Slatopolsky E. Clinical consequences and management of hypomagnesemia. J Am Soc Nephrol. 2009;20:2291-2295.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 97]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
53.  Musso CG. Magnesium metabolism in health and disease. Int Urol Nephrol. 2009;41: 357-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 133]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
54.  Fleming CR, George L, Stoner GL, Tarrosa VB, Moyer TP. The importance of urinary magnesium values in patients with gut failure. Mayo Clin Proc. 1996;71:21-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
55.  Pape UF, Weylandt KH, Knappe-Drzikova B, Gerlach U, Pascher A. Kurzdarmsyndrom und Darmversagen: Diagnostik und Therapie. Aktuel Ernahrungsmed. 2013;38:132-146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
56.  Walker AF, Marakis G, Christie S, Byng M. Mg citrate found more bioavailable than other Mg preparations in a randomised, double-blind study. Magnes Res. 2003;16:183-191.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Blumenstein I, Shastri YM, Stein J. Gastroenteric tube feeding: techniques, problems and solutions. World J Gastroenterol. 2014;20:8505-8524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 241]  [Cited by in F6Publishing: 216]  [Article Influence: 21.6]  [Reference Citation Analysis (9)]
58.  Yang CF, Duro D, Zurakowski D, Lee M, Jaksic T, Duggan C. High prevalence of multiple micronutrient deficiencies in children with intestinal failure: a longitudinal study. J Pediatr 2011; 159: 39-44. e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 85]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
59.  Ubesie AC, Heubi JE, Kocoshis SA, Henderson CJ, Mezoff AG, Rao MB, Cole CR. Vitamin D deficiency and low bone mineral density in pediatric and young adult intestinal failure. J Pediatr Gastroenterol Nutr. 2013;57:372-376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 29]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
60.  Mziray-Andrew CH, Sentongo TA. Nutritional deficiencies in intestinal failure. Pediatr Clin North Am. 2009;56:1185-1200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
61.  Ubesie AC, Kocoshis SA, Mezoff AG, Henderson CJ, Helmrath MA, Cole CR. Multiple micronutrient deficiencies among patients with intestinal failure during and after transition to enteral nutrition. J Pediatr. 2013;163:1692-1696.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 52]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
62.  Khaodhiar L, Keane-Ellison M, Tawa NE, Thibault A, Burke PA, Bistrian BR. Iron deficiency anemia in patients receiving home total parenteral nutrition. JPEN J Parenter Enteral Nutr. 2002;26:114-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 30]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
63.  Hwa YL, Rashtak S, Kelly DG, Murray JA. Iron Deficiency in Long-Term Parenteral Nutrition Therapy. JPEN J Parenter Enteral Nutr. 2016;40:869-876.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
64.  Allwood MC, Martin H, Greenwood M, Maunder M. Precipitation of trace elements in parenteral nutrition mixtures. Clin Nutr. 1998;17:223-226.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 18]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
65.  Robinson CA, Sawyer JE. Y-site compatibility of medications with parenteral nutrition. J Pediatr Pharmacol Ther. 2009;14:48-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
66.  Evstatiev R, Marteau P, Iqbal T, Khalif IL, Stein J, Bokemeyer B, Chopey IV, Gutzwiller FS, Riopel L, Gasche C;  FERGI Study Group. FERGIcor, a randomized controlled trial on ferric carboxymaltose for iron deficiency anemia in inflammatory bowel disease. Gastroenterology 2011; 141: 846-853. e1-2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 228]  [Cited by in F6Publishing: 226]  [Article Influence: 17.4]  [Reference Citation Analysis (0)]
67.  Wolman SL, Anderson GH, Marliss EB, Jeejeebhoy KN. Zinc in total parenteral nutrition: requirements and metabolic effects. Gastroenterology. 1979;76:458-467.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Jeejeebhoy K. Zinc: an essential trace element for parenteral nutrition. Gastroenterology. 2009;137:S7-12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 85]  [Cited by in F6Publishing: 80]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
69.  Okawada M, Holst JJ, Teitelbaum DH. Administration of a dipeptidyl peptidase IV inhibitor enhances the intestinal adaptation in a mouse model of short bowel syndrome. Surgery. 2011;150:217-223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
70.  Coughlin S, Roth L, Lurati G, Faulhaber M. Somatostatin analogues for the treatment of enterocutaneous fistulas: a systematic review and meta-analysis. World J Surg. 2012;36:1016-1029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 28]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
71.  Stein J, Stier C, Raab H, Weiner R. Review article: The nutritional and pharmacological consequences of obesity surgery. Aliment Pharmacol Ther. 2014;40:582-609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 170]  [Cited by in F6Publishing: 140]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
72.  Hvistendahl M, Brandt CF, Tribler S, Naimi RM, Hartmann B, Holst JJ, Rehfeld JF, Hornum M, Andersen JR, Henriksen BM, Brøbech Mortensen P, Jeppesen PB. Effect of Liraglutide Treatment on Jejunostomy Output in Patients With Short Bowel Syndrome: An Open-Label Pilot Study. JPEN J Parenter Enteral Nutr. 2018;42:112-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 20]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
73.  Pape UF, Maasberg S, Pascher A. Pharmacological strategies to enhance adaptation in intestinal failure. Curr Opin Organ Transplant. 2016;21:147-152.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
74.  Billiauws L, Joly F. Emerging treatments for short bowel syndrome in adult patients. Expert Rev Gastroenterol Hepatol. 2019;13:241-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (1)]
75.  Jeppesen PB, Staun M, Tjellesen L, Mortensen PB. Effect of intravenous ranitidine and omeprazole on intestinal absorption of water, sodium, and macronutrients in patients with intestinal resection. Gut. 1998;43:763-769.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 114]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
76.  Kunkel D, Basseri B, Low K, Lezcano S, Soffer EE, Conklin JL, Mathur R, Pimentel M. Efficacy of the glucagon-like peptide-1 agonist exenatide in the treatment of short bowel syndrome. Neurogastroenterol Motil. 2011;23:739-e328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 52]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
77.  de Vries FEE, Reeskamp LF, van Ruler O, van Arum I, Kuin W, Dijksta G, Haveman JW, Boermeester MA, Serlie MJ. Systematic review: pharmacotherapy for high-output enterostomies or enteral fistulas. Aliment Pharmacol Ther. 2017;46:266-273.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
78.  Ecker KW, Stallmach A, Löffler J, Greinwald R, Achenbach U. Long-term treatment of high intestinal output syndrome with budesonide in patients with Crohn's disease and ileostomy. Dis Colon Rectum. 2005;48:237-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
79.  Ecker KW, Stallmach A, Seitz G, Gierend M, Greinwald R, Achenbach U. Oral budesonide significantly improves water absorption in patients with ileostomy for Crohn disease. Scand J Gastroenterol. 2003;38:288-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
80.  Chordia P, MacArthur RD. Crofelemer, a novel agent for treatment of non-infectious diarrhea in HIV-infected persons. Expert Rev Gastroenterol Hepatol. 2013;7:591-600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
81.  Crutchley RD, Miller J, Garey KW. Crofelemer, a novel agent for treatment of secretory diarrhea. Ann Pharmacother. 2010;44: 878-884.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 22]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
82.  King RF, Norton T, Hill GL. A double-blind crossover study of the effect of loperamide hydrochloride and codeine phosphate on ileostomy output. Aust N Z J Surg. 1982;52:121-124.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 59]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
83.  Stevens PJ, Dunbar F, Briscoe P. Potential of Loperamide Oxide in the Reduction of Ileostomy and Colostomy Output. Clin Drug Investig. 1995;10:158-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
84.  Newton CR. Effect of codeine phosphate, Lomotil, and Isogel on iileostomy function. Gut. 1978;19:377-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 54]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
85.  Buchman AL, Fryer J, Wallin A, Ahn CW, Polensky S, Zaremba K. Clonidine reduces diarrhea and sodium loss in patients with proximal jejunostomy: a controlled study. JPEN J Parenter Enteral Nutr. 2006;30:487-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 51]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
86.  McDoniel K, Taylor B, Huey W, Eiden K, Everett S, Fleshman J, Buchman TG, Alpers D, Klein S. Use of clonidine to decrease intestinal fluid losses in patients with high-output short-bowel syndrome. JPEN J Parenter Enteral Nutr. 2004;28:265-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
87.  Rubinoff MJ, Piccione PR, Holt PR. Clonidine prolongs human small intestine transit time: use of the lactulose-breath hydrogen test. Am J Gastroenterol. 1989;84:372-374.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Baumer P, Danays T, Lion L, Cosnes J, Gendre JP, Le Quintrec Y. [Effect of clonidine on oro-cecal transit time in normal man]. Ann Gastroenterol Hepatol (Paris). 1989;25:5-9.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Byrne TA, Wilmore DW, Iyer K, Dibaise J, Clancy K, Robinson MK, Chang P, Gertner JM, Lautz D. Growth hormone, glutamine, and an optimal diet reduces parenteral nutrition in patients with short bowel syndrome: a prospective, randomized, placebo-controlled, double-blind clinical trial. Ann Surg. 2005;242:655-661.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 125]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
90.  Drucker DJ. Biologic actions and therapeutic potential of the proglucagon-derived peptides. Nat Clin Pract Endocrinol Metab. 2005;1:22-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 176]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
91.  Drucker DJ. Gut adaptation and the glucagon-like peptides. Gut. 2002;50:428-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 90]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
92.  Jeppesen PB. The use of hormonal growth factors in the treatment of patients with short-bowel syndrome. Drugs. 2006;66:581-589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
93.  Jeppesen PB. New approaches to the treatments of short bowel syndrome-associated intestinal failure. Curr Opin Gastroenterol. 2014;30:182-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 25]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
94.  Jeppesen PB, Sanguinetti EL, Buchman A, Howard L, Scolapio JS, Ziegler TR, Gregory J, Tappenden KA, Holst J, Mortensen PB. Teduglutide (ALX-0600), a dipeptidyl peptidase IV resistant glucagon-like peptide 2 analogue, improves intestinal function in short bowel syndrome patients. Gut. 2005;54:1224-1231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 344]  [Cited by in F6Publishing: 326]  [Article Influence: 17.2]  [Reference Citation Analysis (0)]
95.  Jeppesen PB, Gilroy R, Pertkiewicz M, Allard JP, Messing B, O'Keefe SJ. Randomised placebo-controlled trial of teduglutide in reducing parenteral nutrition and/or intravenous fluid requirements in patients with short bowel syndrome. Gut. 2011;60:902-914.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 283]  [Cited by in F6Publishing: 273]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
96.  Jeppesen PB, Pertkiewicz M, Messing B, Iyer K, Seidner DL, O'keefe SJ, Forbes A, Heinze H, Joelsson B. Teduglutide reduces need for parenteral support among patients with short bowel syndrome with intestinal failure. Gastroenterology 2012; 143: 1473-1481. e3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 297]  [Article Influence: 24.8]  [Reference Citation Analysis (0)]
97.  Pevny S, Maasberg S, Rieger A, Karber M, Blüthner E, Knappe-Drzikova B, Thurmann D, Büttner J, Weylandt KH, Wiedenmann B, Müller VA, Bläker H, Pascher A, Pape UF. Experience with teduglutide treatment for short bowel syndrome in clinical practice. Clin Nutr. 2019;38:1745-1755.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
98.  Ring LL, Nerup N, Jeppesen PB, Svendsen LB, Achiam MP. Glucagon like peptide-2 and neoplasia; a systematic review. Expert Rev Gastroenterol Hepatol. 2018;12:257-264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
99.  Pironi L, Guidetti C, Incasa E, Poggioli G, Paganelli F, Merli C, Fumi L, Miglioli M. Oral rehydration solution containing rice maltodextrins in patients with total colectomy and high intestinal output. Int J Clin Pharmacol Res. 2000;20:55-60.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Szkudlarek J, Jeppesen PB, Mortensen PB. Effect of high dose growth hormone with glutamine and no change in diet on intestinal absorption in short bowel patients: a randomised, double blind, crossover, placebo controlled study. Gut. 2000;47:199-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 158]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
101.  Jeppesen PB, Szkudlarek J, Høy CE, Mortensen PB. Effect of high-dose growth hormone and glutamine on body composition, urine creatinine excretion, fatty acid absorption, and essential fatty acids status in short bowel patients: a randomized, double-blind, crossover, placebo-controlled study. Scand J Gastroenterol. 2001;36:48-54.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 48]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
102.  Li-Ling, Irving M. The effectiveness of growth hormone, glutamine and a low-fat diet containing high-carbohydrate on the enhancement of the function of remnant intestine among patients with short bowel syndrome: a review of published trials. Clin Nutr. 2001;20:199-204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 25]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
103.  Seguy D, Vahedi K, Kapel N, Souberbielle JC, Messing B. Low-dose growth hormone in adult home parenteral nutrition-dependent short bowel syndrome patients: a positive study. Gastroenterology. 2003;124:293-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 183]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
104.  Goulet O, Dabbas-Tyan M, Talbotec C, Kapel N, Rosilio M, Souberbielle JC, Corriol O, Ricour C, Colomb V. Effect of recombinant human growth hormone on intestinal absorption and body composition in children with short bowel syndrome. JPEN J Parenter Enteral Nutr. 2010;34:513-520.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
105.  Wales PW, Nasr A, de Silva N, Yamada J. Human growth hormone and glutamine for patients with short bowel syndrome. Cochrane Database Syst Rev. 2010: CD006321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 37]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
106.  Peretti N, Loras-Duclaux I, Kassai B, Restier-Miron L, Guimber D, Gottrand F, Coopman S, Michaud L, Marinier E, Yantren H, Michalski MC, Aubert F, Mercier C, Pelosse M, Lopez M, Chatelain P, Lachaux A. Growth hormone to improve short bowel syndrome intestinal autonomy: a pediatric randomized open-label clinical trial. JPEN J Parenter Enteral Nutr. 2011;35:723-731.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
107.  Seguy D, Darmaun D, Duhamel A, Thuillier F, Cynober L, Cortot A, Gottrand F, Messing B. Growth hormone enhances fat-free mass and glutamine availability in patients with short-bowel syndrome: an ancillary double-blind, randomized crossover study. Am J Clin Nutr. 2014;100:850-858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
108.  Van Gossum A, Cabre E, Hébuterne X, Jeppesen P, Krznaric Z, Messing B, Powell-Tuck J, Staun M, Nightingale J;  ESPEN. ESPEN Guidelines on Parenteral Nutrition: gastroenterology. Clin Nutr. 2009;28:415-427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 65]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
109.  Nehra V, Camilleri M, Burton D, Oenning L, Kelly DG. An open trial of octreotide long-acting release in the management of short bowel syndrome. Am J Gastroenterol. 2001;96:1494-1498.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 48]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
110.  Szilagyi A, Shrier I. Systematic review: the use of somatostatin or octreotide in refractory diarrhoea. Aliment Pharmacol Ther. 2001;15:1889-1897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 44]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
111.  O'Keefe SJ, Peterson ME, Fleming CR. Octreotide as an adjunct to home parenteral nutrition in the management of permanent end-jejunostomy syndrome. JPEN J Parenter Enteral Nutr. 1994;18:26-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 35]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
112.  Crenn P, Morin MC, Joly F, Penven S, Thuillier F, Messing B. Net digestive absorption and adaptive hyperphagia in adult short bowel patients. Gut. 2004;53:1279-1286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 110]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
113.  Asplin JR. The management of patients with enteric hyperoxaluria. Urolithiasis. 2016;44:33-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 56]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
114.  Wedlake L, A'Hern R, Russell D, Thomas K, Walters JR, Andreyev HJ. Systematic review: the prevalence of idiopathic bile acid malabsorption as diagnosed by SeHCAT scanning in patients with diarrhoea-predominant irritable bowel syndrome. Aliment Pharmacol Ther. 2009;30:707-717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 283]  [Article Influence: 18.9]  [Reference Citation Analysis (0)]
115.  Mottacki N, Simrén M, Bajor A. Review article: bile acid diarrhoea - pathogenesis, diagnosis and management. Aliment Pharmacol Ther. 2016;43:884-898.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 62]  [Article Influence: 7.8]  [Reference Citation Analysis (0)]
116.  Orekoya O, McLaughlin J, Leitao E, Johns W, Lal S, Paine P. Quantifying bile acid malabsorption helps predict response and tailor sequestrant therapy. Clin Med (Lond). 2015;15:371.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 4]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
117.  Wedlake L, Thomas K, Lalji A, Anagnostopoulos C, Andreyev HJ. Effectiveness and tolerability of colesevelam hydrochloride for bile-acid malabsorption in patients with cancer: a retrospective chart review and patient questionnaire. Clin Ther. 2009;31:2549-2558.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 57]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
118.  Sueyoshi R, Woods Ignatoski KM, Okawada M, Hartmann B, Holst J, Teitelbaum DH. Stimulation of intestinal growth and function with DPP4 inhibition in a mouse short bowel syndrome model. Am J Physiol Gastrointest Liver Physiol. 2014;307:G410-G419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
119.  Heydorn S, Jeppesen PB, Mortensen PB. Bile acid replacement therapy with cholylsarcosine for short-bowel syndrome. Scand J Gastroenterol. 1999;34:818-823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 52]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
120.  Gruy-Kapral C, Little KH, Fordtran JS, Meziere TL, Hagey LR, Hofmann AF. Conjugated bile acid replacement therapy for short-bowel syndrome. Gastroenterology. 1999;116:15-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 41]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
121.  Fürst T, Bott C, Stein J, Dressman JB. Enteric-coated cholylsarcosine microgranules for the treatment of short bowel syndrome. J Pharm Pharmacol. 2005;57:53-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
122.  Walters JR, Johnston IM, Nolan JD, Vassie C, Pruzanski ME, Shapiro DA. The response of patients with bile acid diarrhoea to the farnesoid X receptor agonist obeticholic acid. Aliment Pharmacol Ther. 2015;41:54-64.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 129]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
123.  Bond A, Huijbers A, Pironi L, Schneider SM, Wanten G, Lal S. Review article: diagnosis and management of intestinal failure-associated liver disease in adults. Aliment Pharmacol Ther. 2019;50:640-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 26]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
124.  Bond A, Taylor M, Abraham A, Teubner A, Soop M, Carlson G, Lal S. Examining the pathophysiology of short bowel syndrome and glucagon-like peptide 2 analogue suitability in chronic intestinal failure: experience from a national intestinal failure unit. Eur J Clin Nutr. 2019;73:751-756.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
125.  Appleton ND, Lal S, Carlson GL, Shaw S, Stevens P, Peristerakis I, Soop M. Cholelithiasis and Related Morbidity in Chronic Intestinal Failure: a Longitudinal Cohort Study from a National Specialized Centre. J Gastrointest Surg. 2019;23:2002-2006.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
126.  Grace E, Shaw C, Whelan K, Andreyev HJ. Review article: small intestinal bacterial overgrowth--prevalence, clinical features, current and developing diagnostic tests, and treatment. Aliment Pharmacol Ther. 2013;38:674-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 123]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
127.  Stein JM, Schneider AR. [Bacterial overgrowth syndrome]. Z Gastroenterol. 2007;45:620-628.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 6]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
128.  O'Keefe SJ. Bacterial overgrowth and liver complications in short bowel intestinal failure patients. Gastroenterology. 2006;130:S67-S69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
129.  Bohm M, Siwiec RM, Wo JM. Diagnosis and management of small intestinal bacterial overgrowth. Nutr Clin Pract. 2013;28:289-299.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 78]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
130.  Capron JP, Gineston JL, Herve MA, Braillon A. Metronidazole in prevention of cholestasis associated with total parenteral nutrition. Lancet. 1983;1:446-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 118]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
131.  Petersen C. D-lactic acidosis. Nutr Clin Pract. 2005;20:634-645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 102]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
132.  Fabian E, Kramer L, Siebert F, Högenauer C, Raggam RB, Wenzl H, Krejs GJ. D-lactic acidosis - case report and review of the literature. Z Gastroenterol. 2017;55:75-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 8]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
133.  Pironi L, Tjellesen L, De Francesco A, Pertkiewicz M, Morselli Labate AM, Staun M, Przedlacki J, Lezo A, Orlandoni P, Pasanisi F;  ESPEN-home artificial nutrition working group. Bone mineral density in patients on home parenteral nutrition: a follow-up study. Clin Nutr. 2004;23:1288-1302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
134.  Pironi L, Labate AM, Pertkiewicz M, Przedlacki J, Tjellesen L, Staun M, De Francesco A, Gallenca P, Guglielmi FW, Van Gossum A, Orlandoni P, Contaldo F, Villares JM;  Espen-Home Artificial Nutrition Working Group. Prevalence of bone disease in patients on home parenteral nutrition. Clin Nutr. 2002;21:289-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 74]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
135.  Nightingale JM. Hepatobiliary, renal and bone complications of intestinal failure. Best Pract Res Clin Gastroenterol. 2003;17:907-929.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 67]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
136.  Castiglioni S, Cazzaniga A, Albisetti W, Maier JA. Magnesium and osteoporosis: current state of knowledge and future research directions. Nutrients. 2013;5:3022-3033.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 257]  [Cited by in F6Publishing: 237]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
137.  Gupta S, Wu X, Moore T, Shen B. Frequency, risk factors, and adverse sequelae of bone loss in patients with ostomy for inflammatory bowel diseases. Inflamm Bowel Dis. 2014;20:259-264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 11]  [Article Influence: 1.1]  [Reference Citation Analysis (1)]
138.  Bischoff-Ferrari HA, Willett WC, Orav EJ, Lips P, Meunier PJ, Lyons RA, Flicker L, Wark J, Jackson RD, Cauley JA, Meyer HE, Pfeifer M, Sanders KM, Stähelin HB, Theiler R, Dawson-Hughes B. A pooled analysis of vitamin D dose requirements for fracture prevention. N Engl J Med. 2012;367:40-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 575]  [Cited by in F6Publishing: 498]  [Article Influence: 41.5]  [Reference Citation Analysis (0)]
139.  Stein J, Böhles H.   Lipid and Lipoprotein Metabolism. In: Stein J, Jauch KW, editors. Clinical Nutrition and Infusion Therapy [Klinische Ernährung und Infusionstherapie]. 1st ed. Berlin, Heidelberg, New York: Springer-Verlag, 2003: 171-190.  [PubMed]  [DOI]  [Cited in This Article: ]
140.  Cook N. Central venous catheters: preventing infection and occlusion. Br J Nurs. 1999;8:980-982, 984, 986.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
141.  Haire WD, Herbst SLJNiCP.   Invited Review: Use of Alteplase (t-PA) for the Management of Thrombotic Catheter Dysfunction: Guidelines From a Consensus Conference of the National Association of Vascular Access Networks (NAVAN). 2000; 15: 265-275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
142.  Bhutta ST, Culp WC. Evaluation and management of central venous access complications. Tech Vasc Interv Radiol. 2011;14:217-224.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 47]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
143.  Reed T, Phillips S. Management of central venous catheter occlusions and repairs. J Intraven Nurs. 1996;19:289-294.  [PubMed]  [DOI]  [Cited in This Article: ]
144.  Baskin JL, Pui CH, Reiss U, Wilimas JA, Metzger ML, Ribeiro RC, Howard SC. Management of occlusion and thrombosis associated with long-term indwelling central venous catheters. Lancet. 2009;374:159-169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 256]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
145.  Kerner JA Jr, Garcia-Careaga MG, Fisher AA, Poole RL. Treatment of catheter occlusion in pediatric patients. JPEN J Parenter Enteral Nutr. 2006;30:S73-S81.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 49]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
146.  Tribler S, Brandt CF, Fuglsang KA, Staun M, Broebech P, Moser CE, Scheike T, Jeppesen PB. Catheter-related bloodstream infections in patients with intestinal failure receiving home parenteral support: risks related to a catheter-salvage strategy. Am J Clin Nutr. 2018;107:743-753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
147.  Wanten G, Calder PC, Forbes A. Managing adult patients who need home parenteral nutrition. BMJ. 2011;342:d1447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 45]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
148.  Dibb MJ, Abraham A, Chadwick PR, Shaffer JL, Teubner A, Carlson GL, Lal S. Central Venous Catheter Salvage in Home Parenteral Nutrition Catheter-Related Bloodstream Infections: Long-Term Safety and Efficacy Data. JPEN J Parenter Enteral Nutr. 2016;40:699-704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 55]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
149.  Thompson JS, Rochling FA, Weseman RA, Mercer DF. Current management of short bowel syndrome. Curr Probl Surg. 2012;49:52-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 71]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
150.  Shanks RM, Donegan NP, Graber ML, Buckingham SE, Zegans ME, Cheung AL, O'Toole GA. Heparin stimulates Staphylococcus aureus biofilm formation. Infect Immun. 2005;73:4596-4606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 197]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
151.  Allon M. Prophylaxis against dialysis catheter-related bacteremia: a glimmer of hope. Am J Kidney Dis. 2008;51:165-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 29]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
152.  Wouters Y, Theilla M, Singer P, Tribler S, Jeppesen PB, Pironi L, Vinter-Jensen L, Rasmussen HH, Rahman F, Wanten GJA. Randomised clinical trial: 2% taurolidine versus 0.9% saline locking in patients on home parenteral nutrition. Aliment Pharmacol Ther. 2018;48:410-422.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 37]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
153.  Barco S, Serlie MJ, Coppens M. Primary thromboprophylaxis for adult patients on home parenteral nutrition: A comment on the 2016 ESPEN guideline. Clin Nutr. 2016;35:1579-1580.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
154.  Lal S, Pironi L, Wanten G, Arends J, Bozzetti F, Cuerda C, Joly F, Kelly D, Staun M, Szczepanek K, Van Gossum A, Schneider SM;  Home Artificial Nutrition & Chronic Intestinal Failure Special Interest Group of the European Society for Clinical Nutrition and Metabolism (ESPEN). Clinical approach to the management of Intestinal Failure Associated Liver Disease (IFALD) in adults: A position paper from the Home Artificial Nutrition and Chronic Intestinal Failure Special Interest Group of ESPEN. Clin Nutr. 2018;37:1794-1797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 70]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
155.  Morgan J, Dibb M, Lal S. Intestinal failure-associated liver disease in adult patients. Curr Opin Clin Nutr Metab Care. 2019;22:383-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
156.  Sasdelli AS, Agostini F, Pazzeschi C, Guidetti M, Lal S, Pironi L. Assessment of Intestinal Failure Associated Liver Disease according to different diagnostic criteria. Clin Nutr. 2019;38:1198-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
157.  Van Gossum A, Demetter P. Hepatobiliary Complications of Chronic Intestinal Failure. Gastroenterol Clin North Am. 2019;48:551-564.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
158.  Barco S, Heuschen CB, Salman B, Brekelmans MP, Serlie MJ, Middeldorp S, Coppens M. Home parenteral nutrition-associated thromboembolic and bleeding events: results of a cohort study of 236 individuals. J Thromb Haemost. 2016;14:1364-1373.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 18]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
159.  Lacaille F, Gupte G, Colomb V, D'Antiga L, Hartman C, Hojsak I, Kolacek S, Puntis J, Shamir R;  ESPGHAN Working Group of Intestinal Failure and Intestinal Transplantation. Intestinal failure-associated liver disease: a position paper of the ESPGHAN Working Group of Intestinal Failure and Intestinal Transplantation. J Pediatr Gastroenterol Nutr. 2015;60:272-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 129]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
160.  Cuerda C, Pironi L;  Home Artificial Nutrition and Chronic Intestinal Failure Special Interest Group of ESPEN. Reply, Letter to the Editor - Primary thromboprophylaxis for adult patients on home parenteral nutrition: A comment on the 2016 ESPEN guideline. Clin Nutr. 2016;35:1581-1582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (1)]
161.  Rege AS, Sudan DL. Autologous gastrointestinal reconstruction: review of the optimal nontransplant surgical options for adults and children with short bowel syndrome. Nutr Clin Pract. 2013;28:65-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 22]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
162.  Rege A. The Surgical Approach to Short Bowel Syndrome - Autologous Reconstruction versus Transplantation. Viszeralmedizin. 2014;30:179-189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
163.  Abu-Elmagd K. The concept of gut rehabilitation and the future of visceral transplantation. Nat Rev Gastroenterol Hepatol. 2015;12:108-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 44]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
164.  Kim HB, Fauza D, Garza J, Oh JT, Nurko S, Jaksic T. Serial transverse enteroplasty (STEP): a novel bowel lengthening procedure. J Pediatr Surg. 2003;38:425-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 253]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
165.  Kahn AB, Tulla KA, Tzvetanov IG. Indications of Intestinal Transplantation. Gastroenterol Clin North Am. 2019;48:575-583.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]