Next Article in Journal
Viburnum opulus L. Juice Phenolics Inhibit Mouse 3T3-L1 Cells Adipogenesis and Pancreatic Lipase Activity
Previous Article in Journal
The Role of Iron in Brain Development: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Recommendations Based on Evidence by the Andalusian Group for Nutrition Reflection and Investigation (GARIN) for the Pre- and Postoperative Management of Patients Undergoing Obesity Surgery

by
Antonio J. Martínez-Ortega
1,2,
Gabriel Olveira
3,4,5,*,
José L. Pereira-Cunill
1,2,
Carmen Arraiza-Irigoyen
6,
José M. García-Almeida
4,7,
José A. Irles Rocamora
8,
María J. Molina-Puerta
9,10,
Juan B. Molina Soria
11,
Juana M. Rabat-Restrepo
12,
María I. Rebollo-Pérez
13,
María P. Serrano-Aguayo
1,2,
Carmen Tenorio-Jiménez
14,
Francisco J. Vílches-López
15 and
Pedro P. García-Luna
1,2,16
1
Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen del Rocío, 41013 Sevilla, Spain
2
Endocrine Diseases Research Group, Institute of Biomedicine of Seville (IBiS), 41007 Sevilla, Spain
3
Unidad de Gestión Clínica de Endocrinología y Nutrición, Hospital Regional Universitario de Málaga/Universidad de Málaga, 29010 Málaga, Spain
4
Instituto de Investigación Biomédica de Málaga (IBIMA), 29010 Málaga, Spain
5
CIBERDEM (CB07/08/0019), Instituto de Salud Carlos III, 28029 Madrid, Spain
6
Servicio de Endocrinología y Nutrición, Complejo Hospitalario de Jaén, 23007 Jaén, Spain
7
Unidad de gestión Clínica de Endocrinología y Nutrición, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
8
UGC Endocrinología y Nutrición, Hospital Universitario Valme, 41014 Sevilla, Spain
9
UGC Endocrinología y Nutrición, Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
10
Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain
11
U. Nutrición y Dietética. Hospital General, Linares, 23700 Jaén, Spain
12
Endocrinología y Nutrición, Hospital Universitario Virgen Macarena, 41009 Sevilla, Spain
13
Servicio de Endocrinología y Nutrición, Hospital Juan Ramón Jiménez, 21005 Huelva, Spain
14
Endocrinology and Nutrition Clinical Management Unit, University Hospital Virgen de las Nieves, 18014 Granada, Spain
15
Servicio de Endocrinología y Nutrición, Hospital Puerta del Mar, 11009 Cádiz, Spain
16
GARIN Group Coordinator, 41007 Seville, Spain
*
Author to whom correspondence should be addressed.
Nutrients 2020, 12(7), 2002; https://doi.org/10.3390/nu12072002
Submission received: 12 May 2020 / Revised: 24 June 2020 / Accepted: 2 July 2020 / Published: 6 July 2020
(This article belongs to the Section Clinical Nutrition)

Abstract

:
In order to develop evidence-based recommendations and expert consensus for nutrition management of patients undergoing bariatric surgery and postoperative follow-up, we conducted a systematic literature search using PRISMA methodology plus critical appraisal following the SIGN and AGREE-II procedures. The results were discussed among all members of the GARIN group, and all members answered a Likert scale questionnaire to assess the degree of support for every recommendation. Patients undergoing bariatric surgery should be screened preoperatively for some micronutrient deficiencies and treated accordingly. A VLCD (Very Low-Calorie Diet) should be used for 4–8 weeks prior to surgery. Postoperatively, a liquid diet should be maintained for a month, followed by a semi-solid diet also for one month. Protein requirements (1–1.5 g/kg) should be estimated using adjusted weight. Systematic use of specific multivitamin supplements is encouraged. Calcium citrate and vitamin D supplements should be used at higher doses than are currently recommended. The use of proton-pump inhibitors should be individualised, and vitamin B12 and iron should be supplemented in case of deficit. All patients, especially pregnant women, teenagers, and elderly patients require a multidisciplinary approach and specialised follow-up. These recommendations and suggestions regarding nutrition management when undergoing bariatric surgery and postoperative follow-up have direct clinical applicability.

1. Introduction

Obesity is a pathology that has reached epidemic proportions in recent years. Apart from treatment using hygienic-dietary measures and drug therapy, surgery is also highly effective in the case of grade 3 obesity (BMI > 40 Kg/m2), or grade 2 obesity (BMI > 35 Kg/m2) with associated comorbidities. The main techniques currently in use are divided into: restrictive practices (those that restrict gastric net volume, known as the gastric sleeve, SG, sleeve gastrectomy or Laparoscopic Sleeve Gastrectomy, LSG); malabsorptive practices (those that achieve a malabsorption of nutrients, such as Biliopancreatic Diversion); and mixed practices (those that combine both procedures, as in the case of the gastric bypass, RYGBP) [1,2]. Currently, the almost universal access route is Laparoscopic. These techniques are highly effective in terms of weight loss; however, there are aspects that still do not have clear answers, such as indications in special groups, pre- and postoperative nutritional management, supplementation needs and the necessary tests in the follow-up of these patients. The Andalusian Group for Nutrition Reflection and Investigation (GARIN) aims to answer these questions, in an attempt to try to improve care for these patients and to standardize routine clinical practice.

2. Materials and Methods

The GARIN group members held a virtual meeting to propose and select questions related to the clinical practice and management of obese patients undergoing obesity surgery. In their opinion, these questions were of interest either because clinical practice guidelines (CPGs) did not provide a response or the response provided was unclear. Once selected, a systematic bibliographic search was carried out in PUBMED, Web of Science (WOS) and SCOPUS, which was narrowed down to systematic review articles, meta-analyses, controlled Clinical Trials (CTs), case series and CPGs, published in the last 10 years, limited to human beings, in English and Spanish. The keywords searched for were “bariatric surgery”, “gastric sleeve”, “Roux-en-Y gastric bypass”, in combination with “elderly”, “very low calorie diet”, “calcium”, “iron”, “micronutrients”, “vitamins”, “deficiency”, “proton-pump inhibitors”, “type 2 diabetes”, and “metabolic syndrome”. Unsystematic reviews, studies with less than 5 participants and publications in other languages were excluded.
A total of 286 results were obtained, of which 193 results met the search criteria, after eliminating duplicates. Figure 1 specifies the process according to the PRISMA (Preferred Reporting Items for Systematic reviews and Meta-Analyses) methodology [3]. The critical evaluation of each article was performed using the SIGN methodology (Scottish Intercollegiate Guidelines Network), with the check list suited to each type of article and classified accordingly (Table 1 and Table 2) [4]. In the case of CPGs, the “AGREE” tool (Appraisal of Guidelines Research and Evaluation) was used in its second validated version in Spanish (AGREE-II). The articles were reviewed by two independent reviewers (AJMO and JLPC). In the case of doubt or discrepancy between reviewers, a third reviewer (PPGL) was used.
The results were presented and discussed in two face-to-face meetings held with the members of the GARIN group. A consensus was reached regarding the responses to the previously selected questions, taking into consideration the evidence available. After the discussion, the group members electronically evaluated the consensus using a Likert-type scale [5,6]. The evaluation form had five possible answers to evaluate each recommendation (“totally disagree” with an assigned value of 1, “disagree” with an assigned value of 2, “neither agree nor disagree” with an assigned value of 3, “agree” with an assigned value of 4, and “totally agree” with an assigned value of 5). The consensus level of each recommendation was calculated by adding the total value resulting from the responses obtained, dividing it by the maximum value, and then multiplying it by 100. Finally, the draft of the article was circulated among all the members until the final version was achieved.

3. Results

The article includes the responses obtained from the above-described process to twenty questions framed in three groups, according to the clinical situation of the bariatric patient, in the Preoperative period, during the Postoperative period or in the Obesity Surgery period in special situations. A list of the study articles used to respond to these questions, and the results of the critical evaluation of each article, is shown in Table 3.
All the GARIN group members responded to the survey. No “disagree” or “strongly disagree” responses were obtained. The questions and the results obtained are specified below.

4. Discussion

4.1. Preoperative Management

4.1.1. Should Micronutrient and Vitamin Levels Be Measured Preoperatively in Patients Who Are Candidates for Obesity Surgery?

The GARIN group is a group that seeks improvement to obese patient management. One area of concern is the ‘preoperative’ evaluation of potential candidates for surgery. Many of the reviewed guidelines concentrate on the ‘postoperative’ management of micronutrient deficits, but there is a lack of evidence for the preoperative setup. Vitamin and micronutrient deficiencies are often present prior to surgery, as several studies have reported [34,62,147,153,161]; however, the routine preoperative evaluation of these elements is a rarely seen requirement in local clinical protocols. Therefore, should these deficits be routinely screened for and treated if present? The recently published AACE/TOS/ASMBS/OMA/ASA 2019 guideline [161] (a high-quality guideline which was not included in the reviewed papers as it was published in April 2020, after the bibliography search was concluded) supports preoperative screening of vitamin B12, D and folate. However, what about chromium or other trace elements?
The GARIN group considers that it is convenient to determine the levels of certain micronutrients and vitamins preoperatively in patients who are candidates for Obesity Surgery. In some studies (Gregory DM et al., De Luís et al., Gomes de Lima et al., Wolf et al., Van Rutte et al.) it has been found that there is a deficiency of micronutrients and vitamins not only postoperatively but also preoperatively (Level of Evidence 3, grade of recommendation D) [34,62,147,153,162]. The GARIN group recommends that a vitamin D (25-Hydroxycholecalciferol) study and an iron metabolism study are performed at least once preoperatively on all patient-candidates for Obesity Surgery, plus vitamin B12 and folic acid studies in the risk population (elderly patients, those with a history of atrophic gastritis, patients with a low consumption of fruits and vegetables, or patients with DM treated with metformin, among others), and its repetition until surgery should be individualized as required by routine clinical practice (grade of recommendation D). This recommendation is in line with the proposal by American guidelines [93,161].
Consensus level: 93%.

4.1.2. Should Deficits Be Supplemented? Are There Any Deficits that Have Been Shown to Be Critical?

If deficits are present, both clinical practice and the potential benefits indicate the reasonableness of treating them. The GARIN group experts advocate the use of supplements if any preoperative deficits exist (Level of Evidence 4, grade of recommendation D).
Vitamin D deficiency is highly frequent in the obese population preoperatively (De Luís et al., Pereira-Santos et al., Van Rutte et al., etc.), so it must be assessed and treated (Level of Evidence 1++, grade of recommendation A) [34,113,147]. Regarding vitamin D supplementation, given the lack of solid evidence about the value to treat, the GARIN group experts advocate supplementation in all patients with values below 20 ng/mL, and in an individualized manner for values between 21 and 30 ng/mL. The pleiotropic effects of vitamin D at the level of carbohydrate metabolism and its possible role in the prevention of infectious processes, in addition to it reducing the risk of fractures and improving bone mineral density must be taken into account.
Iron, folate and ferritin deficiency are also frequent (Level of Evidence 3, grade of recommendation D). If deficits are detected the GARIN group experts recommend following the current treatment recommendations [163].
Consensus level: 100%

4.1.3. Is the VLCD (Very Low-Calorie Diet) Recommended Preoperatively in All Patients? Is Liquid VLCD Better Than Solid VLCD?

The use of VLCDs (Very Low-Calorie Diets) in the preparatory preoperative phase to achieve rapid weight loss (and thereby reduce total liver volume) is still a cause of debate. Should liquid or solid VLCDs be used? And for how long?
A VLCD, as its name indicates, is a diet with an important restriction in its caloric content (between 450 and 800 kcal per day). Its objective is to achieve weight loss in patients who due to their circumstances are unable to achieve satisfactory weight loss with standard restrictive diets, or in situations that require significant weight loss in a short period of time. This type of diet not only includes 0.8–1.5 g of proteins of high biological value per kilogram of ideal weight, but also the recommended daily doses of vitamins, minerals, trace elements and essential fatty acids, in order to obtain an adequate nutrition with conservation of lean mass. The VLCDs play a key role both pre and postoperatively in Obesity Surgery [15,79,146,164].
The GARIN group believes that a VLCD is useful preoperatively, as long as: it is managed by specialists with sufficient experience in its use; it provides a sufficient quantity of high biological value proteins; and that the subsequent control follows the specific recommendations in force (ECGs, monitoring of glucose metabolism and hypoglycaemic and hypotensive treatment, and regular analytical control, among others). Table 4 shows the main characteristics of a preoperative VLCD [165].
Both solid and liquid VLCDs allow similar weight losses, but patients who receive a preoperative liquid VLCD appear to have a greater loss of visceral fat mass, which is accompanied by a shorter surgical time, although this does not reflect on a reduction of complications (Leite-Faria et al., Level of Evidence 1−, grade of recommendation B) [79]. Although VLCD is not mentioned in European or American guidelines, the latter guidelines do highlight that a preoperative weight loss might decrease liver volume, and this in turn may help improve the technical aspects of surgery in patients with an enlarged liver or fatty liver disease [161].
The preoperative VLCD diet is not associated with greater postsurgical deficits (Benassar Remolar et al., Level of Evidence 3, grade of recommendation D) [15].
Given this evidence, the GARIN group experts advocate the use of a liquid VLCD diet preoperatively, for a recommended 4–8 weeks minimum prior to surgery and ideally for a longer length of time in patients with a higher BMI and/or associated metabolic-type comorbidities (such as HBP, DM, etc.), especially when taking into consideration its effects on better surgical results and greater visceral fat loss [79,166,167,168,169].
Consensus level: 91%.
References of the reviewed study articles in this section: [15,20,34,43,61,62,76,77,78,79,89,93,101,102,108,113,115,116,128,129,136,146,147,150,153].

4.2. Postoperative Management

When is it possible to resume solid food intake after Obesity Surgery?
The timing for resuming the intake of solid food after a successful surgical procedure is a matter of debate. The clinical opinion varies greatly, with time ranges that span from weeks to months; and as the aforementioned 2019 guidelines highlight, there is even a lack of consensus regarding the number of phases to be included [161].
The GARIN group advocates a liquid diet during the first postoperative phase, followed by a phase of purées, low in carbohydrates and rich in fibre. An adequate supply of liquids and at least 1–1.5 g of protein per kg of adjusted weight per day, together with micronutrients, vitamins and trace elements, must be ensured in both phases. There is controversy regarding the duration of each phase, with a lack of strong evidence in any direction. Although the rapid start of a solid diet in RYGBP (within the first postoperative week) was not associated with an increase in severe surgical complications in one of the studies considered (Edholm D, Level of Evidence 3) [38], which would reinforce the safety of the early start of the post-surgical diet, the degree of evidence is still insufficient to recommend a change in routine clinical practice. Thus, the GARIN group advocates maintaining a liquid diet for about 4 weeks, and then a semi-solid diet for another 4 weeks (grade of recommendation D).
Consensus level: 84%.

4.2.1. What Protein Provisions Should the Diet Contain?

Almost all the guidelines and systematic reviews analysed coincide in the need for a stable protein delivery, but differences arise in the finer details. The protein provision should ensure the prevention of muscle wasting and loss of lean mass. Protein intake is lower in the immediate postoperative period and at 6 and 12 months postoperative with respect to baseline values (and this trend seems to follow medium-term), therefore a higher dietary provision of protein should be recommended (Schiavo et al., Level of Evidence 3, grade of recommendation D) [130]. The GARIN group advises against calculating the protein provision based on a percentage of the diet’s total caloric value, since this method often results in insufficient intake. Instead, it is advisable to use a direct calculation based on the adjusted weight, at least 1 to 1.5 g of high biological value protein per kg of weight and day, as proposed by European and American guidelines (grade of recommendation D) [20,93,161]. Given that the necessary provisions are not normally reached with the usual intake, the use of protein supplements could be beneficial in the 6–12 months after surgery, allowing the maintenance of lean mass and functionality and improving fat loss (Schollenberger et al., Level of Evidence 1+, grade of recommendation B) [133].
Consensus level: 96%

4.2.2. What Contributions of Calcium and Vitamin D Should Be Recommended Postoperatively?

The need for postoperative calcium and vitamin D supplementation is well established, as the evidence is robust. Bone metabolism and available vitamin D and calcium management often change after bariatric surgery, as bone turnover and bone density loss increases. Therefore, the use of these two compounds, either separately or in combination is a must. [31,65,81,104,120,154,156].
Deficits, especially of iron and vitamin D, as well as B12, are frequent in all techniques (the risk is even greater in the case of malabsorptive surgeries; Level of Evidence 1++).
Elevated bone turnover, secondary hyperparathyroidism, and vitamin D deficiency, together with lower bone mineral density, have been observed in patients undergoing both LSG and RYGBP, with the deficit being greater in the latter group (Chakhtoura et al., Hsin et al., Liu et al., Level of Evidence 1++) [26,27,65,81]. Likewise, the risk of fracture is clearly increased (Zhang et al., Rousseau et al., Level of Evidence 1++) [122,160]. Thus, the use of calcium (1000 mg of calcium element at least) and vitamin D (880 IU of cholecalciferol) supplements is a standard postoperative recommendation (Flores et al., Liu et al., Level of Evidence 1++, grade of recommendation A) [44,45,81]. However, the current recommendations for calcium and vitamin D intake (1200–2000 mg/day of elemental calcium combined with 400–800 U of vitamin D, according to the European guideline [20]) seem to be insufficient for avoiding secondary hyperparathyroidism, so higher doses should be considered, especially after malabsorptive surgeries (Obinwanne et al., White et al., Wei et al., Level of Evidence 3, grade of recommendation D) [104,154,156]. In biliopancreatic diversion/Scopinaro surgeries the GARIN group recommends a higher intake of calcium (2000 mg/d) and especially a higher intake of vitamin D (2000 IU/d) (grade of recommendation D).
Consensus level: 91%.

4.2.3. Should Iron Supplementation Be a Routine Postoperative Necessity?

Iron deficiency and anaemia in the postoperative situation is frequent and a cause for concern especially in younger female patients with polymenorrhagia or other medical conditions that alter the iron metabolism. This complication could, theoretically, be averted if we systematically supplement iron, but… is it worth it? Should we indiscriminately provide iron salts to every patient? Or should we perhaps adopt a more conservative approach, monitoring iron metabolism, with treatment being subject to having established the existence of iron deficiency? The 2017 European guidelines recommend prophylactic empiric iron supplementation after gastric bypass, biliopancreatic diversion, duodenal switch and sleeve gastrectomy [20]. However, providing specific iron supplementation in a systematic way does not seem to reduce the risk of anaemia (Kwon et al., Level of Evidence 1++, grade of recommendation grade A) [75]. However, since more than 40% of patients may have severe iron deficiencies, their levels should be monitored and treated appropriately, either by using oral iron or intravenous iron in the case of refractoriness (Obinwanne et al., Level of Evidence 3, grade of recommendation D) [105]. The GARIN group proposes periodic monitoring of iron levels and treatment in the case of deficit, coinciding with the American guidelines [93,161].
Consensus level: 85%.

4.2.4. Should Vitamin B12 Supplementation Be a Routine Postoperative Necessity?

Insufficient vitamin B12 is a common occurrence in almost all clinical techniques. It can lead to neurological and hematological complications if left unchecked. The routine postoperative use of B12, to prevent the development of B12-induced anaemia, is supported by some authors and studies [75]. Several guidelines recommend the use of oral vitamin B12 supplements after gastric bypass, sleeve gastrectomy, biliopancreatic diversion and duodenal switch, with the possibility of parenteral administration of variable regimes [20,161].
The use of postoperative prophylactic vitamin B12 seems to prevent the development of anaemia (Kwon et al., Level of Evidence 1++, grade of recommendation A) [75]. The GARIN group only supports the parenteral treatment of vitamin B12 if the deficit is major (grade of recommendation D).
Consensus level: 85%.

4.2.5. Should an Increase of the Dietary Intake of Other Micronutrients Be a Routine Postoperative Necessity?

The various surgical techniques intended for obesity treatment, and especially those that cause a degree of intestinal malabsorption, can lead to a deficit in trace elements, as previously stated [2,93]. This fact brings into play the need to improve the plasma profile of these elements. A dietary means to resolve this is a supplement of multivitamin pills and/or pharmacological preparations. We did not find any evidence regarding this matter, and there is no recommendation in any of the revised guidelines. Although there is no scientific evidence, a consideration of the pathophysiological mechanism of Obesity Surgery, especially malabsorptive surgery, would make an increase of the dietary intake of other micronutrients, including supplements recommendable (Level of Evidence 4, grade of recommendation D).
Consensus level: 89%.

4.2.6. What Iron and Calcium Preparations Should Be Used?

The physiopathological changes in the digestive process can alter the absorption of several pharmacological compounds including calcium and iron salts and reduce their bioavailability. However, there are few differences between these formulations in terms of pharmacokinetic behaviour. This leads to the question of whether any form of iron or calcium salt is less influenced by these changes, meaning that it would be superior to the rest in achieving therapeutic goals? There is little evidence available regarding which iron preparation to use, and routine clinical practice is highly heterogeneous, so we cannot make a recommendation in this regard. Regarding calcium, the citrate preparations seem to have a higher bioavailability, especially in patients undergoing RYGBP (Sakhaee et al., evidence 1+, grade of recommendation B) [124,125]. The GARIN group recommends their use, as proposed in the European guidelines and in the American consensus (although therein the grade of recommendation is lower) [20,161].
Consensus level: 93%.

4.2.7. What Analytical Determinations Should Be Made in the Follow-Up of Patients Who Have Undergone Obesity Surgery? How Often?

Bariatric patient follow-up is currently based on expert opinions, and can vary to a high degree. Most guidelines draw a clear line between restrictive and malabsorptive surgeries with specific recommendations, but while some procedures and visit schedules might be similar in the short-term after surgery, in the long-term (6–12 months), the positions of the different groups start to diverge. For example, 24-hour urine calcium is recommended in the follow-up of RYGBP in the 2017 European Association for the Study of Obesity statement, while in the American 2019 guidelines it is not recommended [20,161]. Therefore, what is the best course of action? To do or not to do? Should we follow one lead or the other? What evidence do we have to support or reject any of them?
There is no evidence available to make a specific recommendation (expert consensus in most CPGs) about the frequency of determinations or what determinations to request. The various experts seem to agree that they differ depending on the technique used.
Taking into account the postoperative deficits observed in the different studies (Alexandrou et al., Chakhtoura M et al., Kornerup, Kwon, Obinwanne) together with the consensus of experts, the GARIN group advocates the following analytical determinations, as shown in Table 5 [10,26,74,75,105]:
Consensus level: 89%

4.2.8. Is the Systematic Use of Proton Pump Inhibitors (PPIs) Necessary?

A frequent complication after bariatric surgery is the development of anastomotic ulcers. Its incidence varies according to different reports (0.6 to 25 %). Patients may present with perforation or massive bleeding after an asymptomatic onset. Other, less acute symptoms are epigastric burn and/or vomiting [28]. Proton pump inhibitors are one of the first steps in the treatment of these ulcers, but… should we use them systematically?
According to the latest American consensus, prophylactic therapy with proton-pump inhibitors should be considered for 90 days to 1 year, depending on risk; there is no specific recommendation in the 2017 European guidelines [20,161]. In RYGBP, the prophylactic use of PPIs significantly reduces the risk of ulcers, so it should be advised in all patients (Coblijn et al., Wu Chao et al., Level of Evidence 1++, grade of recommendation A) [28,157].
In LSG, we have found no evidence for or against its systematic use, so the GARIN group recommends individualising its use (Level of Evidence 4, grade of recommendation D).
We have not found any evidence regarding the duration of treatment with PPI for either technique, so we again recommend individualising its use (Level of Evidence 4, grade of recommendation D).
Consensus level: 96%.

4.2.9. Is Systematic Supplementation with Multivitamin Complexes and Micronutrients Necessary?

According to the 2017 European guidelines and the newest American consensus, systematic supplementation with multivitamin complexes is recommended (grade of recommendation D and B, respectively) [20,161].
The GARIN group advocates the systematic use of multivitamin complexes. Taking into account the high prevalence of postsurgical deficits of micronutrients both in RYGBP and in LSG, even after 13 years of surgery (Ben-Porat et al., Gesquiere et al., Gillon et al., Gobato et al., Moizé et al., Obeid et al., Level of Evidence 3) [14,51,52,57,103] and that multivitamin complexes seem to reduce them (Gregory et al., Schijns et al., Level of Evidence 2+) [62,131], it seems reasonable to recommend the systematic use of multivitamin and mineral complexes (Level of Evidence 2+, grade of recommendation C).
Consensus level: 98%.

4.2.10. Does the Composition of Currently Available Multivitamin Supplements Fit the Recommendations?

All guidelines support the use of multivitamin supplements in the bariatric patient, and in view of the aforementioned discussion, this seems reasonable. However, are all these supplements the same? Do they have similar composition and characteristics? And more importantly, do they comply with the current recommendations?
Although supplementation reduces micronutrient deficits, there are still patients who present deficits despite them (Ben-Porat et al., Dogan et al., Gesquiere et al., Guillon et al., Gobato et al., Moizé et al., Obeid et al., Level of Evidence 3) [14,36,51,52,57,98,103]. Therefore, we can assume that although in many cases it is sufficient, in others the supplements are insufficient, fundamentally because in many cases the composition does not adapt to the international recommendations (Table 6). Specific supplements appear to be more effective than non-specific supplements, so they should be recommended (Schijns et al., Level of Evidence 2+, grade of recommendation D) [131]. The GARIN group recommends, whenever available, those supplements that are specifically designed for patients undergoing Obesity Surgery.
Consensus level: 85%.

4.2.11. Is It Necessary to Keep an Eye Out for the Appearance of Kidney Stones? What Measures Should Be Used to Prevent Their Appearance?

Another frequent and sometimes forgotten complication of bariatric surgery is the development of kidney stones, especially in RYGBP (11%); in LSG the incidence is significantly lower (1.5%). The underlying pathophysiological mechanisms of kidney stone formation following bariatric surgery are complex and diverse, including hyperoxaluria, hypocitraturia, and abnormally acid urine [170]. Currently, only 24-hour urinary calcium is recommended as a screening tool in most CPGs, with no specific indications for screening or routine detection of kidney stones in patients undergoing RYGBP. Therefore, we discussed how could we face this clinical problem.
At least in the RYGBP, a higher incidence of kidney stones and calcium supersaturation in urine has been found, so it is necessary to keep an eye out for its appearance (Upala et al., Level of Evidence 1+, grade of recommendation B) [145]. There is no evidence available for LSG.
There are no specific recommendations for screening or routine detection of kidney stones in patients undergoing RYGBP. It would be advisable to monitor the kidney function with the use of serum creatinine and formulas to estimate glomerular filtration, occasional 24-hourour urine calcium, and, in selected cases, imaging tests (Level of Evidence 4, grade of recommendation D) would be desirable.
Consensus level: 84%.

4.2.12. Is Long-Term Follow-Up of Obesity Patients Who Have Undergone Restrictive Techniques Necessary? For How Long? Is Long-Term Follow-Up of Obesity Patients Who Have Undergone Malabsorptive Techniques Necessary? For How Long? Is Long-Term Follow-Up of Obesity Patients Who Have Undergone Mixed Techniques Necessary? For How Long?

Restrictive techniques have a significantly lower risk of deficiencies in comparison with malabsorptive techniques. Most guidelines advocate almost lifelong specialised follow-up of these patients, at least once a year. Nevertheless, in LSG patients who show no evident deficits, adequate clinical evolution and no severe comorbidities, this may not be necessary. These patients can be monitored by a general practitioner, thus reducing the burden on healthcare systems.
There is no high-quality evidence in this regard. After one year post-surgery, the GARIN group recommends annual check-ups in Specialised Care for at least five years of all patients who underwent Obesity Surgery. After this time, it is advisable to maintain annual check-ups in patients who underwent malabsorptive techniques, while those patients without complications who underwent purely restrictive techniques do not require specialised follow-up, except in selected cases (Level of Evidence 4, grade of recommendation D).
Consensus level: 87%.
References of the studies reviewed in this section: [7,10,11,12,13,14,16,17,18,20,21,22,25,26,28,30,31,32,35,36,37,38,39,42,43,44,45,50,51,52,57,62,65,67,71,72,73,74,75,77,80,81,82,83,84,85,87,89,90,93,95,96,97,98,99,102,103,104,105,108,110,111,115,120,121,122,123,124,125,126,127,129,130,131,132,133,134,141,142,143,145,147,148,151,152,154,155,156,157,160].

4.3. Obesity Surgery in Special Situations

4.3.1. What Technique Should We Recommend in Patients with Type 2 Diabetes Mellitus (T2DM) and Other Metabolic Comorbidities?

Type 2 Diabetes Mellitus (T2DM) is a great burden associated with obesity and perhaps one of the great pandemics of our time. One out of four morbidly obese patients have had T2DM, which is linked to higher cardiovascular risk and comorbidities. Bariatric surgery is a treatment option that can achieve diabetes remission, as several studies have demonstrated [159]. Both restrictive and malabsorptive techniques are effective in achieving this goal, but it is unclear if one is superior to the other.
Apparently, RYGBP and LSG are effective in terms of diabetes remission (especially in patients with a shorter duration of T2DM), but RYGBP seems to be superior in terms of probability of T2DM remission (RR 43.1 versus 16.6, Yska JP et al.), so it could be recommended as the first option (Level of Evidence 3, grade of recommendation D) [159]. The GARIN group recommends that, whatever the technique used, it should be performed by a Surgical Team with experience in that technique.
Consensus level: 91%.

4.3.2. Pregnancy after Obesity Surgery

In women of childbearing potential, weight loss after bariatric surgery not only improves metabolic status, it also allows restoration of fertility, as this is often impaired in obese women. This phenomenon is thought to be associated with a steep reduction of insulin resistance and a decrease in androgen endogenous production [20]. However, bariatric surgery also causes different degrees of malabsorption and/or nutritional deficiencies, which could in turn cause foetal growth impairment, and in severe cases, miscarriage. As it is a complex situation, the intervention of several healthcare professionals such as endocrinologists, obstetricians, dietitians and/or specialised nurses, is required, to mention a few. Thus, proper preparation and close follow-up before and during pregnancy is of utmost importance for both mother and child [171].
Pregnancy after bariatric surgery is an increasingly frequent situation which does not have a large amount of scientific evidence available due to its peculiarities; therefore, the grade of recommendation is limited. However, it is a situation of risk for both the foetus (increased risk of small for gestational age (Adams et al., Level of Evidence 3) [9] and iron and vitamin A deficiency (Gimenes et al., Level of Evidence 3) [54]) and the mother (higher risk of anaemia (Gimenes et al., Level of Evidence 3) [53] and vitamin K deficiency (Jans G et al., Level of Evidence 3) [69]). Pregnant women who have undergone obesity surgery should be considered as high risk, not only due to nutrient deficiency but also due to complications of obesity prior to the surgery (Level of Evidence 4, grade of recommendation D). In experienced centres, however, the risk of complications is low and even comparable to the general population (González-Navarro et al., Jans G et al., Mead et al., Level of Evidence 3) [60,68,92].
All women who undergo Obesity Surgery when of childbearing age and who want to have a child should program their pregnancy in a regulated manner (Level of Evidence 4, grade of recommendation D), and receive specific supplementation with multivitamins and vitamin B12 in addition to folic acid and iodine (Jans G et al., Level of Evidence 2++, grade of recommendation C) [68]. There is no evidence regarding the minimum time to wait before programming a pregnancy after Obesity Surgery; the general opinion is at least one year after surgery and/or variations in body weight not exceeding 5% (Level of Evidence 4, grade of recommendation D). During this waiting time, effective contraceptive measures should be recommended (Level of Evidence 4, grade of recommendation D).
All women, regardless of the type of surgery performed, should be screened for micronutrient deficiencies (Vitamin A, D, E, K, B12, folic acid and iron) at least every six-months prior to pregnancy, every three months during pregnancy (or sooner if any type of deficit is detected) and at 6-8 weeks post-partum, especially in the case of breastfeeding (Jans G et al., Level of Evidence 2++, grade of recommendation C) [68].
There is not enough evidence to make a recommendation in favour of or against the determination of zinc, iodine, magnesium, or calcium, but its monitoring seems reasonable (Level of Evidence 4, grade of recommendation D).
In addition to the standard supplementation it is advisable to include vitamin K supplementation, since low circulating levels of vitamin K have been observed in women who undergo Obesity Surgery (and even in pregnant women who do not undergo surgery) (Jans G et al., Level of Evidence 3, grade of recommendation D) [69].
There is no evidence found in the literature that contraindicates breastfeeding after delivery, so it can and should be recommended due to its benefits (Level of Evidence 4, grade of recommendation D).
In light of the above, the GARIN group recommends that:
(a)
Pregnancy should be avoided in the first year after Obesity Surgery, and contraceptive measures should be routinely recommended.
(b)
All women of childbearing age undergoing Obesity Surgery should program their gestation at an experienced centre.
(c)
Possible micronutrient deficits (Vitamin A, D, E, K, B12, folic, iron) should be detected (AND treated) at least every six months prior to gestation, in each trimester of pregnancy (or sooner if any type of deficit is detected) and at 6-8 weeks post-partum, especially in case of breastfeeding.
(d)
Breastfeeding is advocated.
Consensus level: 87%.

4.3.3. Obesity Surgery in the Two Non-Working Age Groups: Adolescents and over 65 Years

The burden of obesity is not restricted to those aged between 18 and 65 years old. Sadly, we are witnessing a steep increase in childhood obesity, to an extent that we have never seen before nor could have predicted several years ago. According to the United States data, the rates of childhood obesity have tripled since the 1980s, and the prevalence of obesity in adolescents has quadrupled; 8.5% of youths meet the criteria of severely obese (BMI ≥ 120% of the 95th percentile), representing approximately 4.5 million children, who are at risk of developing T2DM, hypertension, dyslipidaemia or even sudden cardiac arrest in severe cases. This dramatic situation calls for immediate and vigorous measures to prevent devastating consequences for these children, and bariatric surgery, while not being the best solution, as it falls within the domain of tertiary prevention, is nevertheless a powerful weapon in our therapeutic arsenal [118].
Both LSG (Elhag et al., Level of Evidence 3) [41] and RYGBP (Souza-Silva et al., Olbers T et al., Level of Evidence 2++) [106,138] seems to be an effective technique for losing weight in adolescents between 13 and 18 years, and RYGBP seems to be effective in inducing remission of type 2 diabetes mellitus and resolving pre-diabetes, in the remission of hypertension and dyslipidaemia, and in the improvement of the quality of life (Souza-Silva et al., Olbers T et al., Level of Evidence 2++), meaning that it could be recommended in these patients (Level of Evidence 3, grade of recommendation D for LSG; Level of Evidence 2++, grade of recommendation C for RYGBP) [106,138].
There are no specific recommendations for the follow-up of this type of patient through to adulthood. It is advisable to maintain the same guidelines as for adults, in addition to monitoring the growth rate (Level of Evidence 4, grade of recommendation D). Special attention should be paid to the adolescent’s mental health, changes in their physique, and promotion of their integration (Level of Evidence 4, grade of recommendation D).
Obese patients older than 65 years of age are also a cause of concern. In Europe, in the year 2015, an estimated 32 million elderly people were reported to be obese, meaning that approximately 1.1 to 1.2% of the total elderly population suffered morbid obesity. These numbers, far from decreasing are currently on the rise. Although higher BMI value seems to predict a lower relative mortality in older adults (known as the “Obesity paradox”), up to the age of 75 the absolute mortality risk increases with increasing BMI. This could be explained by the presence of several confounding factors (survival effect, competing mortalities, shortened life expectancy, etc.) as highlighted in different studies, thus misleading to a false perception of obesity in the elderly [172,173].
Medical complications of obesity in this population group are mainly concentrated around metabolic syndrome (impaired glucose tolerance, hypertension, dyslipidaemia and cardiovascular disease). Bariatric surgery, as an effective tool for inducing remission of these conditions, is a very interesting option. However, the results in terms of weight loss seems to be less impressive than in younger patients, and the surgical risk is higher. As a result, the dilemma arises. Should we indicate bariatric surgery in elderly obese patients or not?
In those over 65 years, Obesity Surgery (both LSG and RYGBP) seems to have a greater number of complications, especially RYGBP, and to be associated with a lower rate of resolution of comorbidities (Giordano S, Haywood C, Marczuk P, Level of Evidence 1++) [55,63,88]. RYGBP also seems to be associated with higher mortality (Giordano S, Marczuk P, Level of Evidence 1++) [55,88]. Weight loss appears to be significantly less than in younger RYGBP patients (Giordano S, Marczuk P, Level of Evidence 1++) [55,88], although with LSG it seems that the loss is similar (Haywood C, Level of Evidence 1−) [63]. Consequently, RYGBP should be discouraged in people over 65, especially in cases of high cardiovascular risk (Giordano S, Marczuk P, Level of Evidence 1++, grade of recommendation A) [55,88], and LSG could be considered as an option (Haywood C, Level of Evidence 1−, grade of recommendation B) [63].
Consensus level: 87%.

4.3.4. Obesity Surgery Re-operation. To Whom and with What Technique?

Although obesity surgery is highly effective, about 20% of bariatric patients fail to achieve a significant amount of weight loss (greater than 50% of their preoperative excessive weight), and/or experience medical or surgical complications (gastro-gastric fistula, recurrent ulcers), leading to a secondary surgical procedure [117].
There is no universal definition of surgery failure in terms of weight loss, so the various studies are quite heterogeneous, which in turn makes it difficult to generate high-level evidence (Mann P et al., degree of evidence 1+) [86]. The GARIN group must decide its own definition and extend its use to standardise criteria.
However, it seems that the RYGBP after LSG is an effective and safe option, allowing additional weight loss and improvement of comorbidities (Tran D et al., Casillas RA et al., Ianelli A et al., Brethauer et al., Level of Evidence 1−, grade of recommendation B) [19,23,66,144], although weight loss is less when compared to patients who underwent RYGBP in the first instance (Pędziwiatr M et al., Level of Evidence 1++) [109]. With regard to RYGBP, the biliopancreatic diversion seems to be the most favourable technique, although evidence is scarce (Brethauer et al., Level of Evidence 1−, grade of recommendation B).
Consensus level: 75%.
References of the studies reviewed in this section: [8,9,19,20,23,24,29,33,40,41,46,47,48,49,53,54,55,56,60,63,64,66,68,69,72,86,88,89,91,92,93,94,100,102,106,107,108,109,112,114,117,118,119,135,137,139,140,144,149,158,159].
A brief summary of all recommendations can be found in Table 7.

5. Conclusions

The GARIN group, after reviewing the evidence available, recommend that patients undergoing bariatric surgery should be screened preoperatively for micronutrient deficiencies and treated accordingly. Additionally, a VLCD should be used for 4–8 weeks prior to surgery. Postoperatively, a liquid diet should be maintained for one month, followed by a semi-solid diet for one month. Protein requirements (1–1.5 g/kg) should be estimated using adjusted weight. Systematic use of specific multivitamin supplements is encouraged. Calcium citrate and vitamin D supplements should be used at higher doses than are currently recommended. The use of proton-pump inhibitors should be individualised, and vitamin B12 and iron should be supplemented in the case of deficiencies. All patients, especially pregnant women, teenagers, and elderly patients require a multidisciplinary approach and specialised follow-up. These recommendations and suggestions regarding nutritional management when undergoing bariatric surgery and postoperative follow-up have direct clinical applicability. Nevertheless, new studies are needed in order to increase the quality of evidence and provide concrete answers to questions that are still unclear.

Funding

Funding for the face-to-face meeting and the translation into English was provided by NESTLE HEALTHCARE. This entity did not participate at any time in the choice of topic to be discussed, nor in the choice of authors or in any scientific aspect of the article. Only in the official translation of the same.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Buchwald, H. The evolution of metabolic/bariatric surgery. Obes. Surg. 2014, 24, 1126–1135. [Google Scholar] [CrossRef] [PubMed]
  2. Colquitt, J.L.; Pickett, K.; Loveman, E.; Frampton, G.K. Surgery for weight loss in adults. In Cochrane Database of Systematic Reviews; John Wiley & Sons, Ltd.: Chichester, UK, 2014. [Google Scholar]
  3. Moher, D.; Liberati, A.; Tetzlaff, J.; Altman, D.G. Preferred reporting items for systematic reviews and meta-analyses: The PRISMA statement. BMJ 2009, 339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. SIGN 50: A Guideline Developer’s Handbook, 3rd ed.; Scottish Intercollegiate Guidelines Network: Edinburg, UK, 2015; ISBN 978-1-909103-30-6.
  5. Carifio, J.; Perla, R. Resolving the 50-year debate around using and misusing Likert scales. Med. Educ. 2008, 42, 1150–1152. [Google Scholar] [CrossRef] [PubMed]
  6. Hartley, J. Some thoughts on Likert-type scales. Int. J. Clin. Health Psychol. 2014, 14, 83–86. [Google Scholar] [CrossRef] [Green Version]
  7. Aaseth, E.; Fagerland, M.W.; Aas, A.-M.; Hewitt, S.; Risstad, H.; Kristinsson, J.; Bøhmer, T.; Mala, T.; Aasheim, E.T. Vitamin concentrations 5 years after gastric bypass. Eur. J. Clin. Nutr. 2015, 69, 1249–1255. [Google Scholar] [CrossRef]
  8. Abdemur, A.; Han, S.-M.; Lo Menzo, E.; Szomstein, S.; Rosenthal, R. Reasons and outcomes of conversion of laparoscopic sleeve gastrectomy to Roux-en-Y gastric bypass for nonresponders. Surg. Obes. Relat. Dis. 2016, 12, 113–118. [Google Scholar] [CrossRef]
  9. Adams, T.D.; Hammoud, A.O.; Davidson, L.E.; Laferrère, B.; Fraser, A.; Stanford, J.B.; Hashibe, M.; Greenwood, J.L.J.; Kim, J.; Taylor, D.; et al. Maternal and neonatal outcomes for pregnancies before and after gastric bypass surgery. Int. J. Obes. 2015, 39, 686–694. [Google Scholar] [CrossRef] [Green Version]
  10. Alexandrou, A.; Armeni, E.; Kouskouni, E.; Tsoka, E.; Diamantis, T.; Lambrinoudaki, I. Cross-sectional long-term micronutrient deficiencies after sleeve gastrectomy versus Roux-en-Y gastric bypass: A pilot study. Surg. Obes. Relat. Dis. 2014, 10, 262–268. [Google Scholar] [CrossRef]
  11. Aron-Wisnewsky, J.; Verger, E.O.; Bounaix, C.; Dao, M.C.; Oppert, J.-M.; Bouillot, J.-L.; Chevallier, J.-M.; Clément, K. Nutritional and protein deficiencies in the short term following both gastric bypass and gastric banding. PLoS ONE 2016, 11, e0149588. [Google Scholar] [CrossRef] [Green Version]
  12. Bailly, L.; Schiavo, L.; Sebastianelli, L.; Fabre, R.; Pradier, C.; Iannelli, A. Anemia and bariatric surgery: Results of a national french survey on administrative data of 306,298 consecutive patients between 2008 and 2016. Obes. Surg. 2018. [Google Scholar] [CrossRef]
  13. Basfi-Fer, K.; Rojas, P.; Carrasco, F.; Valencia, A.; Inostroza, J.; Codoceo, J.; Pizarro, F.; Olivares, M.; Papapietro, K.; Csendes, A.; et al. Evolución de la ingesta y del estado nutricional de zinc, hierro y cobre. Nutr. Hosp. 2012, 27, 1527–1535. [Google Scholar] [CrossRef] [PubMed]
  14. Ben-Porat, T.; Elazary, R.; Goldenshluger, A.; Sherf Dagan, S.; Mintz, Y.; Weiss, R. Nutritional deficiencies four years after laparoscopic sleeve gastrectomy—Are supplements required for a lifetime? Surg. Obes. Relat. Dis. 2017, 13, 1138–1144. [Google Scholar] [CrossRef] [PubMed]
  15. Bennasar Remolar, M.Á.; Martínez Ramos, D.; Ortega Serrano, J.; Salvador Sanchís, J.L. Nutritional alterations after very low-calorie diet before bariatric surgery. Cir. Esp. Engl. Ed. 2016, 94, 159–164. [Google Scholar] [CrossRef]
  16. Botella-Carretero, J.I.; Lafuente, C.; Montes-Nieto, R.; Balsa, J.; Vega-Piñero, B.; Garcia-Moreno, F.; Peromingo, R.; Galindo, J.; San-Millan, J.L.; Escobar-Morreale, H. Serum bioavailable vitamin D concentrations and bone mineral density in women after obesity surgery. Obes. Surg. 2016, 26, 2732–2737. [Google Scholar] [CrossRef]
  17. Botella Romero, F.; Milla Tobarra, M.; Alfaro Martínez, J.J.; García Arce, L.; García Gómez, A.; Ángeles Salas Sáiz, M.; Soler Marín, A. Bariatric surgery in duodenal switch procedure: Weight changes and associated nutritional deficiencies. Endocrinol. Nutr. Engl. Ed. 2011, 58, 214–218. [Google Scholar] [CrossRef] [PubMed]
  18. Boyce, S.G.; Goriparthi, R.; Clark, J.; Cameron, K.; Roslin, M.S. Can composite nutritional supplement based on the current guidelines prevent vitamin and mineral deficiency after weight loss surgery? Obes. Surg. 2016, 26, 966–971. [Google Scholar] [CrossRef] [PubMed]
  19. Brethauer, S.A.; Kothari, S.; Sudan, R.; Williams, B.; English, W.J.; Brengman, M.; Kurian, M.; Hutter, M.; Stegemann, L.; Kallies, K.; et al. Systematic review on reoperative bariatric surgery. Surg. Obes. Relat. Dis. 2014, 10, 952–972. [Google Scholar] [CrossRef]
  20. Busetto, L.; Dicker, D.; Azran, C.; Batterham, R.L.; Farpour-Lambert, N.; Fried, M.; Hjelmesæth, J.; Kinzl, J.; Leitner, D.R.; Makaronidis, J.M.; et al. Practical recommendations of the obesity management task force of the European association for the study of obesity for the post-bariatric surgery medical management. Obes. Facts 2017, 10, 597–632. [Google Scholar] [CrossRef]
  21. Cabral, J.A.V.; de Souza, G.P.; Nascimento, J.d.A.; Simoneti, L.F.; Marchese, C.; Sales-Peres, S.H.d.C. Impact of vitamin D and calcium deficiency in the bones of patients undergoing bariatric surgery: A systematic review. ABCD Arq. Bras. Cir. Dig. São Paulo 2016, 29, 120–123. [Google Scholar] [CrossRef] [Green Version]
  22. Caron, M.; Hould, F.S.; Lescelleur, O.; Marceau, S.; Lebel, S.; Julien, F.; Simard, S.; Biertho, L. Long-term nutritional impact of sleeve gastrectomy. Surg. Obes. Relat. Dis. 2017, 13, 1664–1673. [Google Scholar] [CrossRef]
  23. Casillas, R.A.; Um, S.S.; Zelada Getty, J.L.; Sachs, S.; Kim, B.B. Revision of primary sleeve gastrectomy to Roux-en-Y gastric bypass: Indications and outcomes from a high-volume center. Surg. Obes. Relat. Dis. 2016, 12, 1817–1825. [Google Scholar] [CrossRef] [PubMed]
  24. Chagas, C.; Saunders, C.; Pereira, S.; Silva, J.; Saboya, C.; Ramalho, A. Vitamin A status and its relationship with serum zinc concentrations among pregnant women who have previously undergone Roux-en-Y gastric bypass. Int. J. Gynecol. Obstet. 2016, 133, 94–97. [Google Scholar] [CrossRef] [PubMed]
  25. Chakhtoura, M.T.; Nakhoul, N.; Akl, E.A.; Mantzoros, C.S.; El Hajj Fuleihan, G.A. Guidelines on vitamin D replacement in bariatric surgery: Identification and systematic appraisal. Metabolism 2016, 65, 586–597. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Chakhtoura, M.T.; Nakhoul, N.N.; Shawwa, K.; Mantzoros, C.; El Hajj Fuleihan, G.A. Hypovitaminosis D in bariatric surgery: A systematic review of observational studies. Metabolism 2016, 65, 574–585. [Google Scholar] [CrossRef] [Green Version]
  27. Chakhtoura, M.; Rahme, M.; El-Hajj Fuleihan, G. Vitamin D metabolism in bariatric surgery. Endocrinol. Metab. Clin. North Am. 2017, 46, 947–982. [Google Scholar] [CrossRef]
  28. Coblijn, U.K.; Goucham, A.B.; Lagarde, S.M.; Kuiken, S.D.; van Wagensveld, B.A. Development of ulcer disease after roux-en-y gastric bypass, incidence, risk factors, and patient presentation: A systematic review. Obes. Surg. 2014, 24, 299–309. [Google Scholar] [CrossRef]
  29. Coblijn, U.K.; Verveld, C.J.; van Wagensveld, B.A.; Lagarde, S.M. Laparoscopic roux-en-y gastric bypass or laparoscopic sleeve gastrectomy as revisional procedure after adjustable gastric band—A systematic review. Obes. Surg. 2013, 23, 1899–1914. [Google Scholar] [CrossRef]
  30. Menegati, G.C.; de Oliveira, L.C.; Santos, A.L.A.; Cohen, L.; Mattos, F.; Mendonça, L.M.C.; Carneiro, J.R.I.; Farias, M.L.F.; Rosado, E.L. Nutritional status, body composition, and bone health in women after bariatric surgery at a university hospital in Rio de Janeiro. Obes. Surg. 2016, 26, 1517–1524. [Google Scholar] [CrossRef]
  31. Costa, T.L.; Paganotto, M.; Radominski, R.B.; Kulak, C.M.; Borba, V.C. Calcium metabolism, vitamin D and bone mineral density after bariatric surgery. Osteoporos. Int. 2015, 26, 757–764. [Google Scholar] [CrossRef]
  32. Sherf Dagan, S.; Tovim, T.B.; Keidar, A.; Raziel, A.; Shibolet, O.; Zelber-Sagi, S. Inadequate protein intake after laparoscopic sleeve gastrectomy surgery is associated with a greater fat free mass loss. Surg. Obes. Relat. Dis. 2017, 13, 101–109. [Google Scholar] [CrossRef]
  33. Daigle, C.R.; Andalib, A.; Corcelles, R.; Cetin, D.; Schauer, P.R.; Brethauer, S.A. Bariatric and metabolic outcomes in the super-obese elderly. Surg. Obes. Relat. Dis. 2016, 12, 132–137. [Google Scholar] [CrossRef] [PubMed]
  34. de Luis, D.A.; Pacheco, D.; Izaola, O.; Terroba, M.C.; Cuellar, L.; Cabezas, G. Micronutrient status in morbidly obese women before bariatric surgery. Surg. Obes. Relat. Dis. 2013, 9, 323–327. [Google Scholar] [CrossRef] [PubMed]
  35. del Villar Madrigal, E.; Neme-Yunes, Y.; Clavellina-Gaytan, D.; Sanchez, H.A.; Mosti, M.; Herrera, M.F. Anemia after Roux-en-Y gastric bypass. How feasible to eliminate the risk by proper supplementation? Obes. Surg. 2015, 25, 80–84. [Google Scholar] [CrossRef]
  36. Dogan, K.; Homan, J.; Aarts, E.O.; de Boer, H.; van Laarhoven, C.J.H.M.; Berends, F.J. Long-term nutritional status in patients following Roux-en-Y gastric bypass surgery. Clin. Nutr. 2018, 37, 612–617. [Google Scholar] [CrossRef]
  37. Dunstan, M.J.D.; Molena, E.J.; Ratnasingham, K.; Kamocka, A.; Smith, N.C.; Humadi, S.; Irukulla, S. Variations in oral vitamin and mineral supplementation following bariatric gastric bypass surgery: A national survey. Obes. Surg. 2015, 25, 648–655. [Google Scholar] [CrossRef] [PubMed]
  38. Edholm, D. Early intake of solid food after Roux-en-Y gastric bypass and complications. A cohort study from the Scandinavian obesity surgery registry. Surg. Obes. Relat. Dis. 2018. [Google Scholar] [CrossRef]
  39. Edholm, D.; Svensson, F.; Näslund, I.; Karlsson, F.A.; Rask, E.; Sundbom, M. Long-term results 11 years after primary gastric bypass in 384 patients. Surg. Obes. Relat. Dis. 2013, 9, 708–713. [Google Scholar] [CrossRef] [PubMed]
  40. Elbahrawy, A.; Bougie, A.; Loiselle, S.-E.; Demyttenaere, S.; Court, O.; Andalib, A. Medium to long-term outcomes of bariatric surgery in older adults with super obesity. Surg. Obes. Relat. Dis. 2018, 14, 470–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Elhag, W.; El Ansari, W.; Abdulrazzaq, S.; Abdullah, A.; Elsherif, M.; Elgenaied, I. Evolution of 29 anthropometric, nutritional, and cardiometabolic parameters among morbidly obese adolescents 2 years post sleeve gastrectomy. Obes. Surg. 2018, 28, 474–482. [Google Scholar] [CrossRef]
  42. Fashandi, A.Z.; Mehaffey, J.H.; Hawkins, R.B.; Schirmer, B.; Hallowell, P.T. Bariatric surgery increases risk of bone fracture. Surg. Endosc. 2018, 32, 2650–2655. [Google Scholar] [CrossRef] [PubMed]
  43. Ferreira Nicoletti, C.; Morandi Junqueira-Franco, M.V.; dos Santos, J.E.; Sergio Marchini, J.; Junior, W.S.; Nonino, C.B. Protein and amino acid status before and after bariatric surgery: A 12-month follow-up study. Surg. Obes. Relat. Dis. 2013, 9, 1008–1012. [Google Scholar] [CrossRef] [PubMed]
  44. Flores, L.; Moizé, V.; Ortega, E.; Rodríguez, L.; Andreu, A.; Filella, X.; Vidal, J. Prospective study of individualized or high fixed doses of vitamin D supplementation after bariatric surgery. Obes. Surg. 2015, 25, 470–476. [Google Scholar] [CrossRef]
  45. Flores, L.; Martínez Osaba, M.J.; Andreu, A.; Moizé, V.; Rodríguez, L.; Vidal, J. Calcium and vitamin D supplementation after gastric bypass should be individualized to improve or avoid hyperparathyroidism. Obes. Surg. 2010, 20, 738–743. [Google Scholar] [CrossRef]
  46. Froylich, D.; Sadeh, O.; Mizrahi, H.; Kafri, N.; Pascal, G.; Daigle, C.R.; Geron, N.; Hazzan, D. Midterm outcomes of sleeve gastrectomy in the elderly. Surg. Obes. Relat. Dis. 2018, 14, 1495–1500. [Google Scholar] [CrossRef] [PubMed]
  47. Fulton, C.; Sheppard, C.; Birch, D.; Karmali, S.; de Gara, C. A comparison of revisional and primary bariatric surgery. Can. J. Surg. 2017, 60, 205–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Gadgil, M.D.; Chang, H.-Y.; Richards, T.M.; Gudzune, K.A.; Huizinga, M.M.; Clark, J.M.; Bennett, W.L. Laboratory testing for and diagnosis of nutritional deficiencies in pregnancy before and after bariatric surgery. J. Womens Health 2014, 23, 129–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Gebhart, A.; Young, M.T.; Nguyen, N.T. Bariatric surgery in the elderly: 2009–2013. Surg. Obes. Relat. Dis. 2015, 11, 393–398. [Google Scholar] [CrossRef]
  50. Gesquiere, I.; Lannoo, M.; Augustijns, P.; Matthys, C.; Van der Schueren, B.; Foulon, V. Iron deficiency after Roux-en-Y gastric bypass: Insufficient iron absorption from oral iron supplements. Obes. Surg. 2014, 24, 56–61. [Google Scholar] [CrossRef] [Green Version]
  51. Gesquiere, I.; Foulon, V.; Augustijns, P.; Gils, A.; Lannoo, M.; Van der Schueren, B.; Matthys, C. Micronutrient intake, from diet and supplements, and association with status markers in pre- and post-RYGB patients. Clin. Nutr. 2017, 36, 1175–1181. [Google Scholar] [CrossRef]
  52. Gillon, S.; Jeanes, Y.M.; Andersen, J.R.; Våge, V. Micronutrient status in morbidly obese patients prior to laparoscopic sleeve gastrectomy and micronutrient changes 5 years post-surgery. Obes. Surg. 2017, 27, 606–612. [Google Scholar] [CrossRef]
  53. Gimenes, J.C.; Nicoletti, C.F.; de Souza Pinhel, M.A.; Cortes-Oliveira, C.; Salgado Júnior, W.; Nonino, C.B. Nutritional status of children from women with previously bariatric surgery. Obes. Surg. 2018, 28, 990–995. [Google Scholar] [CrossRef] [PubMed]
  54. Gimenes, J.C.; Nicoletti, C.F.; de Souza Pinhel, M.A.; de Oliveira, B.A.P.; Salgado Júnior, W.; Marchini, J.S.; Nonino, C.B. Pregnancy after Roux en Y gastric bypass: Nutritional and biochemical aspects. Obes. Surg. 2017, 27, 1815–1821. [Google Scholar] [CrossRef] [PubMed]
  55. Giordano, S.; Victorzon, M. Laparoscopic Roux-en-Y gastric bypass in elderly patients (60 years or older): A meta-analysis of comparative studies. Scand. J. Surg. 2018, 107, 6–13. [Google Scholar] [CrossRef] [PubMed]
  56. Victorzon, M.; Giordano, S. Bariatric surgery in elderly patients: A systematic review. Clin. Interv. Aging 2015, 1627. [Google Scholar] [CrossRef] [Green Version]
  57. Gobato, R.C.; Seixas Chaves, D.F.; Chaim, E.A. Micronutrient and physiologic parameters before and 6 months after RYGB. Surg. Obes. Relat. Dis. 2014, 10, 944–951. [Google Scholar] [CrossRef]
  58. Goldberg, H.R.; Chin, V.L.; Zitsman, J.L.; Zhang, C.; Williams, K.M.; Oberfield, S.; Fennoy, I. Bariatric surgery in adolescents: Is routine nutrient supplementation sufficient to avoid anemia following bariatric surgery? Nutr. Clin. Pract. 2017, 32, 502–507. [Google Scholar] [CrossRef]
  59. Lima, K.V.G.; Lima, R.P.A.; Gonçalves, M.C.R.; Faintuch, J.; Morais, L.C.S.L.; Asciutti, L.S.R.; Costa, M.J.C. High frequency of serum chromium deficiency and association of chromium with triglyceride and cholesterol concentrations in patients awaiting bariatric surgery. Obes. Surg. 2014, 24, 771–776. [Google Scholar] [CrossRef]
  60. González Navarro, I.; Pereira Cunill, J.L.; Serrano Aguayo, P.; Morales Conde, S.; Martos Martínez, J.M.; García Luna, P.P. Resultados materno-fetales de la gestación tras cirugía bariátrica. Nutr. Hosp. 2011, 376–383. [Google Scholar] [CrossRef]
  61. Grace, C.; Vincent, R.; Aylwin, S.J. High prevalence of vitamin D insufficiency in a United Kingdom urban morbidly obese population: Implications for testing and treatment. Surg. Obes. Relat. Dis. 2014, 10, 355–360. [Google Scholar] [CrossRef]
  62. Gregory, D.M.; Twells, L.K.; Lester, K.K.; Midodzi, W.K.; Pedersen, M.R.; Pace, D.; Smith, C.; Boone, D.; Randell, E.W.; Kovacs, C.S. Preoperative and postoperative assessments of biochemical parameters in patients with severe obesity undergoing laparoscopic sleeve gastrectomy. Obes. Surg. 2017. [Google Scholar] [CrossRef]
  63. Haywood, C.; Sumithran, P. Treatment of obesity in older persons-A systematic review. Obes. Rev. 2019. [Google Scholar] [CrossRef] [PubMed]
  64. Homan, J.; Betzel, B.; Aarts, E.O.; van Laarhoven, K.J.H.M.; Janssen, I.M.C.; Berends, F.J. Secondary surgery after sleeve gastrectomy: Roux-en-Y gastric bypass or biliopancreatic diversion with duodenal switch. Surg. Obes. Relat. Dis. 2015, 11, 771–777. [Google Scholar] [CrossRef] [PubMed]
  65. Hsin, M.-C.; Huang, C.-K.; Tai, C.-M.; Yeh, L.-R.; Kuo, H.-C.; Garg, A. A case-matched study of the differences in bone mineral density 1 year after 3 different bariatric procedures. Surg. Obes. Relat. Dis. 2015, 11, 181–185. [Google Scholar] [CrossRef] [PubMed]
  66. Iannelli, A.; Debs, T.; Martini, F.; Benichou, B.; Ben Amor, I.; Gugenheim, J. Laparoscopic conversion of sleeve gastrectomy to Roux-en-Y gastric bypass: Indications and preliminary results. Surg. Obes. Relat. Dis. 2016, 12, 1533–1538. [Google Scholar] [CrossRef] [PubMed]
  67. James, H.; Lorentz, P.; Collazo-Clavell, M.L. Patient-reported adherence to empiric vitamin/mineral supplementation and related nutrient deficiencies after Roux-en-Y gastric bypass. Obes. Surg. 2016, 26, 2661–2666. [Google Scholar] [CrossRef]
  68. Jans, G.; Matthys, C.; Bogaerts, A.; Lannoo, M.; Verhaeghe, J.; Van der Schueren, B.; Devlieger, R. Maternal micronutrient deficiencies and related adverse neonatal outcomes after bariatric surgery: A systematic review. Adv. Nutr. 2015, 6, 420–429. [Google Scholar] [CrossRef] [Green Version]
  69. Jans, G.; Guelinckx, I.; Voets, W.; Galjaard, S.; Van Haard, P.M.M.; Vansant, G.M.; Devlieger, R. Vitamin K1 monitoring in pregnancies after bariatric surgery: A prospective cohort study. Surg. Obes. Relat. Dis. 2014, 10, 885–890. [Google Scholar] [CrossRef]
  70. Jáuregui-Lobera, I. Iron deficiency and bariatric surgery. Nutrients 2013, 5, 1595–1608. [Google Scholar] [CrossRef] [Green Version]
  71. Kalani, A.; Bami, H.; Tiboni, M.; Jaeschke, R.; Adachi, J.D.; Lau, A.N. The effect of bariatric surgery on serum 25-OH vitamin D levels: A systematic review and meta-analysis: Impact on bone health in bariatric surgery. Obes. Sci. Pract. 2017, 3, 319–332. [Google Scholar] [CrossRef]
  72. Kim, M.K.; Kim, W.; Kwon, H.-S.; Baek, K.-H.; Kim, E.K.; Song, K.-H. Effects of bariatric surgery on metabolic and nutritional parameters in severely obese Korean patients with type 2 diabetes: A prospective 2-year follow up. J. Diabetes Investig. 2014, 5, 221–227. [Google Scholar] [CrossRef] [Green Version]
  73. Ito, M.K.; Gonçalves, V.S.S.; Faria, S.L.C.M.; Moizé, V.; Porporatti, A.L.; Guerra, E.N.S.; De Luca Canto, G.; de Carvalho, K.M.B. Effect of protein intake on the protein status and lean mass of post-bariatric surgery patients: A systematic review. Obes. Surg. 2017, 27, 502–512. [Google Scholar] [CrossRef] [PubMed]
  74. Kornerup, L.S.; Hvas, C.L.; Abild, C.B.; Richelsen, B.; Nexo, E. Early changes in vitamin B12 uptake and biomarker status following Roux-en-Y gastric bypass and sleeve gastrectomy. Clin. Nutr. 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Kwon, Y.; Kim, H.J.; Lo Menzo, E.; Park, S.; Szomstein, S.; Rosenthal, R.J. Anemia, iron and vitamin B12 deficiencies after sleeve gastrectomy compared to Roux-en-Y gastric bypass: A meta-analysis. Surg. Obes. Relat. Dis. 2014, 10, 589–597. [Google Scholar] [CrossRef] [PubMed]
  76. Krzizek, E.-C.; Brix, J.M.; Herz, C.T.; Kopp, H.P.; Schernthaner, G.-H.; Schernthaner, G.; Ludvik, B. Prevalence of micronutrient deficiency in patients with morbid obesity before bariatric surgery. Obes. Surg. 2018, 28, 643–648. [Google Scholar] [CrossRef] [PubMed]
  77. Lecube, A.; Zafon, C.; Gromaz, A.; Fort, J.M.; Caubet, E.; Baena, J.A.; Tortosa, F. Iodine deficiency is higher in morbid obesity in comparison with late after bariatric surgery and non-obese women. Obes. Surg. 2015, 25, 85–89. [Google Scholar] [CrossRef]
  78. Lefebvre, P.; Letois, F.; Sultan, A.; Nocca, D.; Mura, T.; Galtier, F. Nutrient deficiencies in patients with obesity considering bariatric surgery: A cross-sectional study. Surg. Obes. Relat. Dis. 2014, 10, 540–546. [Google Scholar] [CrossRef]
  79. Faria, S.L.; Faria, O.P.; Cardeal, M.d.A.; Ito, M.K. Effects of a very low calorie diet in the preoperative stage of bariatric surgery: A randomized trial. Surg. Obes. Relat. Dis. 2015, 11, 230–237. [Google Scholar] [CrossRef]
  80. Li, Z.; Zhou, X.; Fu, W. Vitamin D supplementation for the prevention of vitamin D deficiency after bariatric surgery: A systematic review and meta-analysis. Eur. J. Clin. Nutr. 2017. [Google Scholar] [CrossRef]
  81. Liu, C.; Wu, D.; Zhang, J.-F.; Xu, D.; Xu, W.-F.; Chen, Y.; Liu, B.-Y.; Li, P.; Li, L. Changes in bone metabolism in morbidly obese patients after bariatric surgery: A meta-analysis. Obes. Surg. 2016, 26, 91–97. [Google Scholar] [CrossRef] [PubMed]
  82. Soares, F.L.; Bissoni de Sousa, L.; Corradi-Perini, C.; Ramos da Cruz, M.R.; Nunes, M.G.J.; Branco-Filho, A.J. Food quality in the late postoperative period of bariatric surgery: An evaluation using the bariatric food pyramid. Obes. Surg. 2014, 24, 1481–1486. [Google Scholar] [CrossRef] [PubMed]
  83. Luger, M.; Kruschitz, R.; Kienbacher, C.; Traussnigg, S.; Langer, F.B.; Prager, G.; Schindler, K.; Kallay, E.; Hoppichler, F.; Trauner, M.; et al. Vitamin D3 loading is superior to conventional supplementation after weight loss surgery in vitamin D-deficient morbidly obese patients: A double-blind randomized placebo-controlled trial. Obes. Surg. 2017, 27, 1196–1207. [Google Scholar] [CrossRef] [Green Version]
  84. Majumder, S.; Soriano, J.; Louie Cruz, A.; Dasanu, C.A. Vitamin B12 deficiency in patients undergoing bariatric surgery: Preventive strategies and key recommendations. Surg. Obes. Relat. Dis. 2013, 9, 1013–1019. [Google Scholar] [CrossRef] [PubMed]
  85. Malone, M.; Alger-Mayer, S.; Lindstrom, J.; Bailie, G.R. Management of iron deficiency and anemia after Roux-en-Y gastric bypass surgery: An observational study. Surg. Obes. Relat. Dis. 2013, 9, 969–974. [Google Scholar] [CrossRef] [PubMed]
  86. Mann, J.P.; Jakes, A.D.; Hayden, J.D.; Barth, J.H. Systematic review of definitions of failure in revisional bariatric surgery. Obes. Surg. 2015, 25, 571–574. [Google Scholar] [CrossRef]
  87. Manousou, S.; Carlsson, L.M.S.; Eggertsen, R.; Hulthén, L.; Jacobson, P.; Landin-Wilhelmsen, K.; Trimpou, P.; Svensson, P.-A.; Nyström, H.F. Iodine status after bariatric surgery—A prospective 10-year report from the swedish obese subjects (SOS) study. Obes. Surg. 2018, 28, 349–357. [Google Scholar] [CrossRef] [Green Version]
  88. Marczuk, P.; Kubisa, M.J.; Święch, M.; Walędziak, M.; Kowalewski, P.; Major, P.; Pędziwiatr, M.; Paśnik, K.; Janik, M.R. Effectiveness and safety of Roux-en-Y gastric bypass in elderly patients—Systematic review and meta-analysis. Obes. Surg. 2018. [Google Scholar] [CrossRef] [Green Version]
  89. Martín García-Almenta, E.; Ruiz-Tovar, J.; Sánchez Santos, R.; Garcia-Moreno, F. (Eds.) Vía Clínica de Cirugía Bariátrica, 1st ed.; Im3diA Comunicación S.L: Albacete, Spain, 2017; ISBN 978-84-697-7104-4. [Google Scholar]
  90. McCracken, E.; Wood, G.C.; Prichard, W.; Bistrian, B.; Still, C.; Gerhard, G.; Rolston, D.; Benotti, P. Severe anemia after Roux-en-Y gastric bypass: A cause for concern. Surg. Obes. Relat. Dis. 2018. [Google Scholar] [CrossRef] [PubMed]
  91. McGlone, E.R.; Bond, A.; Reddy, M.; Khan, O.A.; Wan, A.C. Super-obesity in the elderly: Is bariatric surgery justified? Obes. Surg. 2015, 25, 1750–1755. [Google Scholar] [CrossRef]
  92. Mead, N.C.; Sakkatos, P.; Sakellaropoulos, G.C.; Adonakis, G.L.; Alexandrides, T.K.; Kalfarentzos, F. Pregnancy outcomes and nutritional indices after 3 types of bariatric surgery performed at a single institution. Surg. Obes. Relat. Dis. 2014, 10, 1166–1173. [Google Scholar] [CrossRef]
  93. Mechanick, J.I.; Youdim, A.; Jones, D.B.; Timothy Garvey, W.; Hurley, D.L.; Molly McMahon, M.; Heinberg, L.J.; Kushner, R.; Adams, T.D.; Shikora, S.; et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery patient—2013 update: Cosponsored by American Association of Clinical Endocrinologists, The Obesity Society, and American Society for Metabolic & Bariatric Surgery. Surg. Obes. Relat. Dis. 2013, 9, 159–191. [Google Scholar] [CrossRef]
  94. Mendes-Filho, A.M.; Godoy, E.S.N.; Alhinho, H.C.A.W.; Galvão-Neto, M.D.P.; Ramos, A.C.; Ferraz, Á.A.B.; Campos, J.M. Fundoplication conversion in Roux-en-Y gastric bypass for control of obesity and gastroesophageal reflux: Systematic review. ABCD Arq. Bras. Cir. Dig. São Paulo 2017, 30, 279–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Mingrone, G.; Bornstein, S.; Le Roux, C.W. Optimisation of follow-up after metabolic surgery. Lancet Diabetes Endocrinol. 2018, 6, 487–499. [Google Scholar] [CrossRef]
  96. Mischler, R.A.; Armah, S.M.; Craig, B.A.; Rosen, A.D.; Banerjee, A.; Selzer, D.J.; Choi, J.N.; Gletsu-Miller, N. Comparison of oral iron supplement formulations for normalization of iron status following Roux-en-Y gastric bypass surgery: A randomized trial. Obes. Surg. 2018, 28, 369–377. [Google Scholar] [CrossRef] [PubMed]
  97. Mischler, R.A.; Armah, S.M.; Wright, B.N.; Mattar, S.G.; Rosen, A.D.; Gletsu-Miller, N. Influence of diet and supplements on iron status after gastric bypass surgery. Surg. Obes. Relat. Dis. 2016, 12, 651–658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Moizé, V.; Andreu, A.; Flores, L.; Torres, F.; Ibarzabal, A.; Delgado, S.; Lacy, A.; Rodriguez, L.; Vidal, J. Long-term dietary intake and nutritional deficiencies following sleeve gastrectomy or Roux-en-Y gastric bypass in a mediterranean population. J. Acad. Nutr. Diet. 2013, 113, 400–410. [Google Scholar] [CrossRef]
  99. Moore, C.E.; Sherman, V. Effectiveness of B vitamin supplementation following bariatric surgery: Rapid increases of serum vitamin B12. Obes. Surg. 2015, 25, 694–699. [Google Scholar] [CrossRef]
  100. Morales, M.P.; Wheeler, A.A.; Ramaswamy, A.; Scott, J.S.; de la Torre, R.A. Laparoscopic revisional surgery after Roux-en-Y gastric bypass and sleeve gastrectomy. Surg. Obes. Relat. Dis. 2010, 6, 485–490. [Google Scholar] [CrossRef]
  101. Nicoletti, C.F.; Lima, T.P.; Donadelli, S.P.; Salgado, W.; Marchini, J.S.; Nonino, C.B. New look at nutritional care for obese patient candidates for bariatric surgery. Surg. Obes. Relat. Dis. 2013, 9, 520–525. [Google Scholar] [CrossRef]
  102. O’Kane, M.; Parretti, H.M.; Hughes, C.A.; Sharma, M.; Woodcock, S.; Puplampu, T.; Blakemore, A.I.; Clare, K.; MacMillan, I.; Joyce, J.; et al. Guidelines for the follow-up of patients undergoing bariatric surgery: Follow-up of bariatric surgery patients. Clin. Obes. 2016, 6, 210–224. [Google Scholar] [CrossRef] [Green Version]
  103. Obeid, N.R.; Malick, W.; Concors, S.J.; Fielding, G.A.; Kurian, M.S.; Ren-Fielding, C.J. Long-term outcomes after Roux-en-Y gastric bypass: 10- to 13-year data. Surg. Obes. Relat. Dis. 2016, 12, 11–20. [Google Scholar] [CrossRef]
  104. Obinwanne, K.M.; Riess, K.P.; Kallies, K.J.; Mathiason, M.A.; Manske, B.R.; Kothari, S.N. Effects of laparoscopic Roux-en-Y gastric bypass on bone mineral density and markers of bone turnover. Surg. Obes. Relat. Dis. 2014, 10, 1056–1062. [Google Scholar] [CrossRef] [PubMed]
  105. Obinwanne, K.M.; Fredrickson, K.A.; Mathiason, M.A.; Kallies, K.J.; Farnen, J.P.; Kothari, S.N. Incidence, treatment, and outcomes of iron deficiency after laparoscopic Roux-en-Y gastric bypass: A 10-year analysis. J. Am. Coll. Surg. 2014, 218, 246–252. [Google Scholar] [CrossRef] [PubMed]
  106. Olbers, T.; Beamish, A.J.; Gronowitz, E.; Flodmark, C.-E.; Dahlgren, J.; Bruze, G.; Ekbom, K.; Friberg, P.; Göthberg, G.; Järvholm, K.; et al. Laparoscopic Roux-en-Y gastric bypass in adolescents with severe obesity (AMOS): A prospective, 5-year, Swedish nationwide study. Lancet Diabetes Endocrinol. 2017, 5, 174–183. [Google Scholar] [CrossRef] [Green Version]
  107. Parmar, C.; Mahawar, K.K.; Carr, W.R.J.; Schroeder, N.; Balupuri, S.; Small, P.K. Bariatric surgery in septuagenarians: A comparison with <60 year olds. Obes. Surg. 2017, 27, 3165–3169. [Google Scholar] [CrossRef]
  108. Parrott, J.; Frank, L.; Rabena, R.; Craggs-Dino, L.; Isom, K.A.; Greiman, L. American Society for Metabolic and Bariatric Surgery integrated health nutritional guidelines for the surgical weight loss patient 2016 update: Micronutrients. Surg. Obes. Relat. Dis. 2017, 13, 727–741. [Google Scholar] [CrossRef] [PubMed]
  109. Pędziwiatr, M.; Małczak, P.; Wierdak, M.; Rubinkiewicz, M.; Pisarska, M.; Major, P.; Wysocki, M.; Karcz, W.K.; Budzyński, A. Revisional gastric bypass is inferior to primary gastric bypass in terms of short- and long-term outcomes—Systematic review and meta-analysis. Obes. Surg. 2018, 28, 2083–2091. [Google Scholar] [CrossRef]
  110. Pellitero, S.; Martínez, E.; Puig, R.; Leis, A.; Zavala, R.; Granada, M.L.; Pastor, C.; Moreno, P.; Tarascó, J.; Puig-Domingo, M. Evaluation of vitamin and trace element requirements after sleeve gastrectomy at long term. Obes. Surg. 2017, 27, 1674–1682. [Google Scholar] [CrossRef]
  111. Pereira, S.; Saboya, C.; Ramalho, A. Impact of different protocols of nutritional supplements on the status of vitamin A in class III obese patients after Roux-en-Y gastric bypass. Obes. Surg. 2013, 23, 1244–1251. [Google Scholar] [CrossRef]
  112. Da Cruz, S.P.; Matos, A.; Pereira, S.; Saboya, C.; da Cruz, S.P.; Ramalho, A. Roux-en-Y gastric bypass aggravates vitamin A deficiency in the mother-child group. Obes. Surg. 2018, 28, 114–121. [Google Scholar] [CrossRef]
  113. Pereira-Santos, M.; Costa, P.R.F.; Assis, A.M.O.; Santos, C.A.S.T.; Santos, D.B. Obesity and vitamin D deficiency: A systematic review and meta-analysis: Obesity and vitamin D. Obes. Rev. 2015, 16, 341–349. [Google Scholar] [CrossRef]
  114. Quirante, F.P.; Montorfano, L.; Rammohan, R.; Dhanabalsamy, N.; Lee, A.; Szomstein, S.; Lo Menzo, E.; Rosenthal, R.J. Is bariatric surgery safe in the elderly population? Surg. Endosc. 2017, 31, 1538–1543. [Google Scholar] [CrossRef] [PubMed]
  115. Peterson, L.A.; Zeng, X.; Caufield-Noll, C.P.; Schweitzer, M.A.; Magnuson, T.H.; Steele, K.E. Vitamin D status and supplementation before and after bariatric surgery: A comprehensive literature review. Surg. Obes. Relat. Dis. 2016, 12, 693–702. [Google Scholar] [CrossRef] [PubMed]
  116. Peterson, L.A.; Cheskin, L.J.; Furtado, M.; Papas, K.; Schweitzer, M.A.; Magnuson, T.H.; Steele, K.E. Malnutrition in bariatric surgery candidates: Multiple micronutrient deficiencies prior to surgery. Obes. Surg. 2016, 26, 833–838. [Google Scholar] [CrossRef] [PubMed]
  117. Pinto-Bastos, A.; Conceição, E.M.; Machado, P.P.P. Reoperative bariatric surgery: A systematic review of the reasons for surgery, medical and weight loss outcomes, relevant behavioral factors. Obes. Surg. 2017, 27, 2707–2715. [Google Scholar] [CrossRef] [PubMed]
  118. Pratt, J.S.A.; Browne, A.; Browne, N.T.; Bruzoni, M.; Cohen, M.; Desai, A.; Inge, T.; Linden, B.C.; Mattar, S.G.; Michalsky, M.; et al. ASMBS pediatric metabolic and bariatric surgery guidelines, 2018. Surg. Obes. Relat. Dis. 2018. [Google Scholar] [CrossRef]
  119. Quezada, N.; Hernández, J.; Pérez, G.; Gabrielli, M.; Raddatz, A.; Crovari, F. Laparoscopic sleeve gastrectomy conversion to Roux-en-Y gastric bypass: Experience in 50 patients after 1 to 3 years of follow-up. Surg. Obes. Relat. Dis. 2016, 12, 1611–1615. [Google Scholar] [CrossRef]
  120. Rodríguez-Carmona, Y.; López-Alavez, F.J.; González-Garay, A.G.; Solís-Galicia, C.; Meléndez, G.; Serralde-Zúñiga, A.E. Bone mineral density after bariatric surgery. A systematic review. Int. J. Surg. 2014, 12, 976–982. [Google Scholar] [CrossRef] [Green Version]
  121. Rottenstreich, A.; Barkai, A.; Arad, A.; Raccah, B.H.; Kalish, Y. The effect of bariatric surgery on direct-acting oral anticoagulant drug levels. Thromb. Res. 2018, 163, 190–195. [Google Scholar] [CrossRef]
  122. Rousseau, C.; Jean, S.; Gamache, P.; Lebel, S.; Mac-Way, F.; Biertho, L.; Michou, L.; Gagnon, C. Change in fracture risk and fracture pattern after bariatric surgery: Nested case-control study. BMJ 2016, i3794. [Google Scholar] [CrossRef] [Green Version]
  123. Ruiz-Tovar, J.; Oller, I.; Priego, P.; Arroyo, A.; Calero, A.; Diez, M.; Zubiaga, L.; Calpena, R. Short- and Mid-term Changes in Bone Mineral Density After Laparoscopic Sleeve Gastrectomy. Obes. Surg. 2013, 23, 861–866. [Google Scholar] [CrossRef]
  124. Sakhaee, K.; Pak, C. Superior calcium bioavailability of effervescent potassium calcium citrate over tablet formulation of calcium citrate after Roux-en-Y gastric bypass. Surg. Obes. Relat. Dis. 2013, 9, 743–748. [Google Scholar] [CrossRef] [PubMed]
  125. Sakhaee, K.; Griffith, C.; Pak, C.Y.C. Biochemical control of bone loss and stone-forming propensity by potassium-calcium citrate after bariatric surgery. Surg. Obes. Relat. Dis. 2012, 8, 67–72. [Google Scholar] [CrossRef]
  126. Salgado, W.; Modotti, C.; Nonino, C.B.; Ceneviva, R. Anemia and iron deficiency before and after bariatric surgery. Surg. Obes. Relat. Dis. 2014, 10, 49–54. [Google Scholar] [CrossRef] [PubMed]
  127. Sallé, A.; Demarsy, D.; Poirier, A.L.; Lelièvre, B.; Topart, P.; Guilloteau, G.; Bécouarn, G.; Rohmer, V. Zinc deficiency: A frequent and underestimated complication after bariatric surgery. Obes. Surg. 2010, 20, 1660–1670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Sánchez, A.; Rojas, P.; Basfi-fer, K.; Carrasco, F.; Inostroza, J.; Codoceo, J.; Valencia, A.; Papapietro, K.; Csendes, A.; Ruz, M. Micronutrient deficiencies in morbidly obese women prior to bariatric surgery. Obes. Surg. 2016, 26, 361–368. [Google Scholar] [CrossRef]
  129. Santarpia, L.; Grandone, I.; Alfonsi, L.; Sodo, M.; Contaldo, F.; Pasanisi, F. Long-term medical complications after malabsorptive procedures: Effects of a late clinical nutritional intervention. Nutrition 2014, 30, 1301–1305. [Google Scholar] [CrossRef]
  130. Schiavo, L.; Scalera, G.; Pilone, V.; De Sena, G.; Quagliariello, V.; Iannelli, A.; Barbarisi, A. A Comparative study examining the impact of a protein-enriched vs. normal protein postoperative diet on body composition and resting metabolic rate in obese patients after sleeve gastrectomy. Obes. Surg. 2017, 27, 881–888. [Google Scholar] [CrossRef]
  131. Schijns, W.; Schuurman, L.T.; Melse-Boonstra, A.; van Laarhoven, C.J.H.M.; Berends, F.J.; Aarts, E.O. Do specialized bariatric multivitamins lower deficiencies after RYGB? Surg. Obes. Relat. Dis. 2018. [Google Scholar] [CrossRef]
  132. Schneider, J.; Peterli, R.; Gass, M.; Slawik, M.; Peters, T.; Wölnerhanssen, B.K. Laparoscopic sleeve gastrectomy and Roux-en-Y gastric bypass lead to equal changes in body composition and energy metabolism 17 months postoperatively: A prospective randomized trial. Surg. Obes. Relat. Dis. 2016, 12, 563–570. [Google Scholar] [CrossRef]
  133. Schollenberger, A.E.; Karschin, J.; Meile, T.; Küper, M.A.; Königsrainer, A.; Bischoff, S.C. Impact of protein supplementation after bariatric surgery: A randomized controlled double-blind pilot study. Nutrition 2016, 32, 186–192. [Google Scholar] [CrossRef]
  134. Shah, M.; Sharma, A.; Wermers, R.A.; Kennel, K.A.; Kellogg, T.A.; Mundi, M.S. Hypocalcemia after bariatric surgery: Prevalence and associated risk factors. Obes. Surg. 2017, 27, 2905–2911. [Google Scholar] [CrossRef] [PubMed]
  135. Sheng, B.; Truong, K.; Spitler, H.; Zhang, L.; Tong, X.; Chen, L. The long-term effects of bariatric surgery on type 2 diabetes remission, microvascular and macrovascular complications, and mortality: A systematic review and meta-analysis. Obes. Surg. 2017, 27, 2724–2732. [Google Scholar] [CrossRef] [PubMed]
  136. Sherf-Dagan, S.; Hod, K.; Buch, A.; Mardy-Tilbor, L.; Regev, Z.; Ben-Porat, T.; Sakran, N.; Goitein, D.; Raziel, A. Health and nutritional status of vegetarian candidates for bariatric surgery and practical recommendations. Obes. Surg. 2018, 28, 152–160. [Google Scholar] [CrossRef] [PubMed]
  137. Sjöström, L.; Peltonen, M.; Jacobson, P.; Ahlin, S.; Andersson-Assarsson, J.; Anveden, Å.; Bouchard, C.; Carlsson, B.; Karason, K.; Lönroth, H.; et al. Association of bariatric surgery with long-term remission of type 2 diabetes and with microvascular and macrovascular complications. JAMA 2014, 311, 2297. [Google Scholar] [CrossRef] [Green Version]
  138. Silva, J.S.; Chaves, G.V.; Stenzel, A.P.; Pereira, S.E.; Saboya, C.J.; Ramalho, A. Improvement of anthropometric and biochemical, but not of vitamin A, status in adolescents who undergo Roux-en-Y gastric bypass: A 1-year follow up study. Surg. Obes. Relat. Dis. 2017, 13, 227–233. [Google Scholar] [CrossRef]
  139. Susmallian, S.; Barnea, R.; Weiss, Y.; Raziel, A. Outcome of bariatric surgery in older patients. Surg. Obes. Relat. Dis. 2018, 14, 1705–1713. [Google Scholar] [CrossRef]
  140. Susmallian, S.; Raziel, A.; Barnea, R.; Paran, H. Bariatric surgery in older adults: Should there be an age limit? Medicine (Baltimore) 2019, 98, e13824. [Google Scholar] [CrossRef]
  141. Tang, L.; Alsulaim, H.A.; Canner, J.K.; Prokopowicz, G.P.; Steele, K.E. Prevalence and predictors of postoperative thiamine deficiency after vertical sleeve gastrectomy. Surg. Obes. Relat. Dis. 2018. [Google Scholar] [CrossRef] [PubMed]
  142. Tondapu, P.; Provost, D.; Adams-Huet, B.; Sims, T.; Chang, C.; Sakhaee, K. Comparison of the absorption of calcium carbonate and calcium citrate after Roux-en-Y gastric bypass. Obes. Surg. 2009, 19, 1256–1261. [Google Scholar] [CrossRef] [Green Version]
  143. Topart, P.; Becouarn, G.; Sallé, A.; Ritz, P. Biliopancreatic diversion requires multiple vitamin and micronutrient adjustments within 2 years of surgery. Surg. Obes. Relat. Dis. 2014, 10, 936–941. [Google Scholar] [CrossRef]
  144. Tran, D.D.; Nwokeabia, I.D.; Purnell, S.; Zafar, S.N.; Ortega, G.; Hughes, K.; Fullum, T.M. Revision of Roux-en-Y gastric bypass for weight regain: A systematic review of techniques and outcomes. Obes. Surg. 2016, 26, 1627–1634. [Google Scholar] [CrossRef] [PubMed]
  145. Upala, S.; Jaruvongvanich, V.; Sanguankeo, A. Risk of nephrolithiasis, hyperoxaluria, and calcium oxalate supersaturation increased after Roux-en-Y gastric bypass surgery: A systematic review and meta-analysis. Surg. Obes. Relat. Dis. 2016, 12, 1513–1521. [Google Scholar] [CrossRef] [PubMed]
  146. Van Nieuwenhove, Y.; Dambrauskas, Z.; Campillo-Soto, A.; van Dielen, F.; Wiezer, R.; Janssen, I.; Kramer, M.; Thorell, A. Preoperative very low-calorie diet and operative outcome after laparoscopic gastric bypass: A randomized multicenter study. Arch. Surg. Chic. Ill. 1960 2011, 146, 1300–1305. [Google Scholar] [CrossRef] [Green Version]
  147. Van Rutte, P.W.J.; Aarts, E.O.; Smulders, J.F.; Nienhuijs, S.W. Nutrient deficiencies before and after sleeve gastrectomy. Obes. Surg. 2014, 24, 1639–1646. [Google Scholar] [CrossRef] [PubMed]
  148. Verger, E.O.; Aron-Wisnewsky, J.; Dao, M.C.; Kayser, B.D.; Oppert, J.-M.; Bouillot, J.-L.; Torcivia, A.; Clément, K. Micronutrient and protein deficiencies after gastric bypass and sleeve gastrectomy: A 1-year follow-up. Obes. Surg. 2016, 26, 785–796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Vinan-Vega, M.; Diaz Vico, T.; Elli, E.F. Bariatric surgery in the elderly patient: Safety and short-time outcome. A case match analysis. Obes. Surg. 2018. [Google Scholar] [CrossRef] [PubMed]
  150. Wang, C.; Guan, B.; Yang, W.; Yang, J.; Cao, G.; Lee, S. Prevalence of electrolyte and nutritional deficiencies in Chinese bariatric surgery candidates. Surg. Obes. Relat. Dis. 2016, 12, 629–634. [Google Scholar] [CrossRef]
  151. Wang, F.-G.; Yu, Z.-P.; Yan, W.-M.; Yan, M.; Song, M.-M. Comparison of safety and effectiveness between laparoscopic mini-gastric bypass and laparoscopic sleeve gastrectomy: A meta-analysis and systematic review. Medicine (Baltimore) 2017, 96, e8924. [Google Scholar] [CrossRef]
  152. Ward, E.K.; Jensen-Otsu, E.; Schoen, J.A.; Rothchild, K.; Mitchell, B.; Austin, G.L. Acid suppression medications are associated with suboptimal weight loss after laparoscopic Roux-en-Y gastric bypass in patients older than 40 years. Surg. Obes. Relat. Dis. 2015, 11, 585–590. [Google Scholar] [CrossRef]
  153. Wolf, E.; Utech, M.; Stehle, P.; Büsing, M.; Stoffel-Wagner, B.; Ellinger, S. Preoperative micronutrient status in morbidly obese patients before undergoing bariatric surgery: Results of a cross-sectional study. Surg. Obes. Relat. Dis. 2015, 11, 1157–1163. [Google Scholar] [CrossRef]
  154. Wei, J.-H.; Lee, W.-J.; Chong, K.; Lee, Y.-C.; Chen, S.-C.; Huang, P.-H.; Lin, S.-J. High incidence of secondary hyperparathyroidism in bariatric patients: Comparing different procedures. Obes. Surg. 2018, 28, 798–804. [Google Scholar] [CrossRef] [PubMed]
  155. Weng, T.-C.; Chang, C.-H.; Dong, Y.-H.; Chang, Y.-C.; Chuang, L.-M. Anaemia and related nutrient deficiencies after Roux-en-Y gastric bypass surgery: A systematic review and meta-analysis. BMJ Open 2015, 5, e006964. [Google Scholar] [CrossRef] [PubMed]
  156. White, M.G.; Ward, M.A.; Applewhite, M.K.; Wong, H.; Prachand, V.; Angelos, P.; Kaplan, E.L.; Grogan, R.H. Rates of secondary hyperparathyroidism after bypass operation for super-morbid obesity: An overlooked phenomenon. Surgery 2017, 161, 720–726. [Google Scholar] [CrossRef] [PubMed]
  157. Ying, V.W.C.; Kim, S.H.H.; Khan, K.J.; Farrokhyar, F.; D’Souza, J.; Gmora, S.; Anvari, M.; Hong, D. Prophylactic PPI help reduce marginal ulcers after gastric bypass surgery: A systematic review and meta-analysis of cohort studies. Surg. Endosc. 2015, 29, 1018–1023. [Google Scholar] [CrossRef] [PubMed]
  158. Yorke, E.; Sheppard, C.; Switzer, N.J.; Kim, D.; de Gara, C.; Karmali, S.; Kanji, A.; Birch, D. Revision of sleeve gastrectomy to Roux-en-Y gastric bypass: A Canadian experience. Am. J. Surg. 2017, 213, 970–974. [Google Scholar] [CrossRef]
  159. Yska, J.P.; van Roon, E.N.; de Boer, A.; Leufkens, H.G.M.; Wilffert, B.; de Heide, L.J.M.; de Vries, F.; Lalmohamed, A. Remission of type 2 diabetes mellitus in patients after different types of bariatric surgery: A population-based cohort study in the United Kingdom. JAMA Surg. 2015, 150, 1126. [Google Scholar] [CrossRef] [Green Version]
  160. Zhang, Q.; Chen, Y.; Li, J.; Chen, D.; Cheng, Z.; Xu, S.; Huang, Y.; Wang, Q. A meta-analysis of the effects of bariatric surgery on fracture risk: Bariatric surgery and fracture. Obes. Rev. 2018, 19, 728–736. [Google Scholar] [CrossRef]
  161. Mechanick, J.I.; Apovian, C.; Brethauer, S.; Timothy Garvey, W.; Joffe, A.M.; Kim, J.; Kushner, R.F.; Lindquist, R.; Pessah-Pollack, R.; Seger, J.; et al. Clinical practice guidelines for the perioperative nutrition, metabolic, and nonsurgical support of patients undergoing bariatric procedures—2019 update: Cosponsored by American Association of Clinical Endocrinologists/American College of Endocrinology, The Obesity Society, American Society for Metabolic and Bariatric Surgery, Obesity Medicine Association, and American Society of Anesthesiologists. Obes. Silver Spring Md. 2020, 28, O1–O58. [Google Scholar] [CrossRef]
  162. Lima, K.V.G.D.; Costa, M.J.D.C.; Gonçalves, M.D.C.R.; Sousa, B.S.D. Deficiências de micronutrientes no pré-operatório de cirurgia bariátrica. ABCD Arq. Bras. Cir. Dig. São Paulo 2013, 26, 63–66. [Google Scholar] [CrossRef] [Green Version]
  163. Amaya García, M.J.; Vílchez López, F.J.; Campos Martín, C.; Sánchez Vera, P.; Pereira Cunill, J.L. Micronutrientes en Cirugía Bariátrica. Nutr. Hosp. 2012, 349–361. [Google Scholar] [CrossRef]
  164. Vilchez López, F.J.; Campos Martín, C.; Amaya García, M.J.; Sánchez Vera, P.; Pereira Cunill, J.L. Very low calorie diets in clinical management of morbid obesity. Nutr. Hosp. 2013, 275–285. [Google Scholar] [CrossRef]
  165. Gargallo Fernández, M.; Basulto Marset, J.; Breton Lesmes, I.; Quiles Izquierdo, J.; Formiguera Sala, X.; Salas-Salvadó, J. Recomendaciones nutricionales basadas en la evidencia para la prevención y el tratamiento del sobrepeso y la obesidad en adultos (consenso FESNAD-SEEDO): Metodología y resumen ejecutivo (I/III). Nutr. Hosp. 2012, 27, 800–832. [Google Scholar] [CrossRef]
  166. Alami, R.S.; Morton, J.M.; Schuster, R.; Lie, J.; Sanchez, B.R.; Peters, A.; Curet, M.J. Is there a benefit to preoperative weight loss in gastric bypass patients? A prospective randomized trial. Surg. Obes. Relat. Dis. Off. J. Am. Soc. Bariatr. Surg. 2007, 3, 141–145. [Google Scholar] [CrossRef] [PubMed]
  167. Van Wissen, J.; Bakker, N.; Doodeman, H.J.; Jansma, E.P.; Bonjer, H.J.; Houdijk, A.P.J. Preoperative methods to reduce liver volume in bariatric surgery: A systematic review. Obes. Surg. 2016, 26, 251–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Gerber, P.; Anderin, C.; Gustafsson, U.O.; Thorell, A. Weight loss before gastric bypass and postoperative weight change: Data from the Scandinavian Obesity Registry (SOReg). Surg. Obes. Relat. Dis. Off. J. Am. Soc. Bariatr. Surg. 2016, 12, 556–562. [Google Scholar] [CrossRef]
  169. Martínez-Ramos, D.; Salvador-Sanchis, J.L.; Escrig-Sos, J. Pérdida de peso preoperatoria en pacientes candidatos a cirugía bariátrica. Recomendaciones basadas en la evidencia. Cir. Esp. 2012, 90, 147–155. [Google Scholar] [CrossRef] [PubMed]
  170. Sakhaee, K.; Poindexter, J.; Aguirre, C. The effects of bariatric surgery on bone and nephrolithiasis. Bone 2016, 84, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Kominiarek, M.A. Preparing for and managing a pregnancy after bariatric surgery. Semin. Perinatol. 2011, 35, 356–361. [Google Scholar] [CrossRef] [Green Version]
  172. Mathus-Vliegen, E.M.H.; Basdevant, A.; Finer, N.; Hainer, V.; Hauner, H.; Micic, D.; Maislos, M.; Roman, G.; Schutz, Y.; Tsigos, C.; et al. Prevalence, pathophysiology, health consequences and treatment options of obesity in the elderly: A guideline. Obes. Facts 2012, 5, 460–483. [Google Scholar] [CrossRef]
  173. Mathus-Vliegen, E.M.H. Obesity and the elderly. J. Clin. Gastroenterol. 2012, 46, 533–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Flow diagram following the PRISMA methodology that reflects the selection and evaluation process of the analysed papers.
Figure 1. Flow diagram following the PRISMA methodology that reflects the selection and evaluation process of the analysed papers.
Nutrients 12 02002 g001
Table 1. Level of Evidence (LoE) assigned to each article according to its quality.
Table 1. Level of Evidence (LoE) assigned to each article according to its quality.
LoEInterpretation
1++High quality meta-analyses, systematic reviews of CTs, or high quality CTs with a very low risk of bias
1+Well conducted meta-analyses, systematic reviews of CTs, or well conducted CTs with a low risk of bias
1−Meta-analyses, systematic reviews of CTs, or CTs with a high risk of bias
2++High quality systematic reviews of case control or cohort studies. Case control or cohort studies with a low risk of bias and a high probability that the relationship is causal
2+Well conducted case control or cohort studies with a low risk of bias and a moderate probability that the relationship is causal
2−Case control or cohort studies with a high risk of bias and a significant risk that the relationship is not causal
3Non-analytic studies, e.g., case reports and case series
4Expert opinion
Abbreviations: CTs: Controlled Clinical Trials.
Table 2. Degree of recommendation applicable to each consensus response according to the supporting evidence.
Table 2. Degree of recommendation applicable to each consensus response according to the supporting evidence.
Grade of RecommendationInterpretation
AAt least one meta-analysis, systematic review, or CT rated as 1++, and directly applicable to the guidelines target population; or A body of scientific evidence consisting of studies rated as 1+ and demonstrating overall consistency of results.
BA body of scientific evidence including studies rated as 2++, directly applicable to the guidelines target population, and demonstrating overall consistency of results; or Extrapolated scientific evidence from studies rated as 1++ or 1+
CA body of scientific evidence including studies rated as 2+, directly applicable to the guidelines target population and demonstrating overall consistency of results; or Extrapolated scientific evidence from studies rated as 2++
DScientific evidence level 3 or 4; or Extrapolated scientific evidence from studies rated as 2+
Table 3. List of study articles/guidelines used to respond to the 20 selected questions.
Table 3. List of study articles/guidelines used to respond to the 20 selected questions.
Articles Used to Answer Each Question
First AuthorType of StudySIGN/AGREE II ScoreLoEReference
Aaseth ECase seriesNo checklist required3[7]
Abdemur ACase seriesNo checklist required3[8]
Adams TDCase-control studyHigh quality (++)2++[9]
Alexandrou ATransversal studyNo checklist required3[10]
Aron-Wisnewsky JCase seriesNo checklist required3[11]
Bailly LCase seriesNo checklist required3[12]
Basfi-Fer KCase seriesNo checklist required3[13]
Ben-Porat TCase seriesNo checklist required3[14]
Benassar Remolar MACase seriesNo checklist required3[15]
Botella-Carretero JICase seriesNo checklist required3[16]
Botella Romero FCase seriesNo checklist required3[17]
Boyce SGCase seriesNo checklist required3[18]
Brethauer SASystematic reviewLow quality (−)1−[19]
Busetto LPractice guideline74.53% Good qualityNA[20]
Cabral JSystematic reviewLow quality (−)1−[21]
Caron MCase seriesNo checklist required3[22]
Casillas RACase seriesNo checklist required3[23]
Chagas CCase seriesNo checklist required3[24]
Chakhtoura MTSystematic reviewAcceptable (+)1+[25]
Chakhtoura MTSystematic reviewHigh quality (++)1++[26]
Chakhtoura MTSystematic reviewHigh quality (++)1++[27]
Coblijn UKSystematic reviewAcceptable (+)1+[28]
Coblijn UKSystematic reviewLow quality (−)1−[29]
Cosendey Menegati GCase-control studyAcceptable (+)2+[30]
Costa TLCase-control studyAcceptable (+)2+[31]
Dagan SSCohort studyAcceptable (+)2+[32]
Daigle CRCase seriesNo checklist required3[33]
De Luis DACase seriesNo checklist required3[34]
Del Villar Madrigal ECase seriesNo checklist required3[35]
Dogan KCase seriesNo checklist required3[36]
Dunstan MCase seriesNo checklist required3[37]
Edholm DCase seriesNo checklist required3[38]
Edholm DCase seriesNo checklist required3[39]
Elbahrawy ACase seriesNo checklist required3[40]
Elhag WCase seriesNo checklist required3[41]
Fashandi AZCase seriesNo checklist required3[42]
Ferreira-Nicoletti CPre-post studyNo checklist required3[43]
Flores LNRCTAcceptable (+)1+[44]
Flores LCase seriesNo checklist required3[45]
Froylich DCase seriesNo checklist required3[46]
Fulton CCase seriesNo checklist required3[47]
Gadgil MDCase-control studyHigh quality (++)2++[48]
Gebhart ACase seriesNo checklist required3[49]
Gesquiere ICase seriesNo checklist required3[50]
Gesquiere ICase seriesNo checklist required3[51]
Gillon SCase seriesNo checklist required3[52]
Gimenes JCTransversal studyNo checklist required3[53]
Gimenes JCCase seriesNo checklist required3[54]
Giordano SSystematic reviewHigh quality (++)1++[55]
Giordano SSystematic reviewAcceptable (+)1+[56]
Gobato RCCase seriesNo checklist required3[57]
Goldberg HRCase seriesNo checklist required3[58]
Gomes de Lima KVCase seriesNo checklist required3[59]
González-Navarro ICase seriesNo checklist required3[60]
Grace, CCase seriesNo checklist required3[61]
Gregory DMPre-post studyNo checklist required3[62]
Haywood CSystematic reviewLow quality (−)1−[63]
Homan JCase seriesNo checklist required3[64]
Hsin MCCohort studyAcceptable (+)2+[65]
Iannelli ACase seriesNo checklist required3[66]
James HCase seriesNo checklist required3[67]
Jans GSystematic reviewLow quality (−)2++[68]
Jans GCohort studyAcceptable (+)2+[69]
Jáuregui-Lobera ISystematic reviewAcceptable (+)1+[70]
Kalani ASystematic reviewHigh quality (++)1++[71]
Kim MKCase seriesNo checklist required3[72]
Kiyomi-Ito MSystematic reviewHigh quality (++)1++[73]
Kornerup LSCase seriesNo checklist required3[74]
Kwon YSystematic reviewHigh quality (++)1++[75]
Krzizek ECCase seriesNo checklist required3[76]
Lecube ATransversal studyNo checklist required3[77]
Lefebvre PCase seriesNo checklist required3[78]
Leite Faria, SOpen RCTAcceptable (+)1−[79]
Li ZSystematic reviewHigh quality (++)1++[80]
Liu CSystematic reviewHigh quality (++)1++[81]
Lucas Soares FCase seriesNo checklist required3[82]
Luger MRCTHigh quality (++)1+[83]
Majumder SSystematic reviewLow quality (−)1−[84]
Malone MCase seriesNo checklist required3[85]
Mann JPSystematic reviewLow quality (−)1−[86]
Manousou SCase-control studyHigh quality (++)2++[87]
Marczuk PSystematic reviewHigh quality (++)1++[88]
Martín García-Almenta EPractice guideline54.03% Low qualityNA[89]
McCracken ECase-control studyHigh quality (++)2++[90]
McGlone ESCase seriesNo checklist required3[91]
Mead NCTransversal studyNo checklist required3[92]
Mechanick JLPractice guideline90.06% Excellent qualityNA[93]
Mendes-Filho AMSystematic reviewLow quality (−)1−[94]
Mingrone GSystematic reviewLow quality (−)1−[95]
Mischler RRCTLow quality (−)1−[96]
Mischler RTransversal studyNo checklist required3[97]
Moizé VCase seriesNo checklist required3[98]
Moore CECase seriesNo checklist required3[99]
Morales MPCase seriesNo checklist required3[100]
Nicoletti CFCase seriesNo checklist required3[101]
O’Kane MPractice guideline77.63% Good qualityNA[102]
Obeid NRCase seriesNo checklist required3[103]
Obinwanne KCase seriesNo checklist required3[104]
Obinwanne KCase seriesNo checklist required3[105]
Olbers TCohort studyHigh quality (++)2+[106]
Parmar CTransversal studyNo checklist required3[107]
Parrot JPractice guideline63.35% Acceptable qualityNA[108]
Pędziwiatr MSystematic reviewHigh quality (++)1++[109]
Pellitero SCase seriesNo checklist required3[110]
Pereira SNRCTAcceptable (+)1−[111]
Pereira Da Cruz SCohort studyHigh quality (++)2+[112]
Pereira-Santos MSystematic reviewHigh quality (++)1++[113]
Pérez Quirante FTransversal studyNo checklist required3[114]
Peterson LASystematic reviewLow quality (−)1−[115]
Peterson LATransversal studyNo checklist required3[116]
Pinto-Bastos ASystematic reviewLow quality (−)1−[117]
Pratt JSAPractice guideline70.8% Good qualityNA[118]
Quezada NCase seriesNo checklist required3[119]
Rodríguez-Carmona YSystematic reviewHigh quality (++)1++[120]
Rottenstreich ACase seriesNo checklist required3[121]
Rousseau CCase-control studyHigh quality (++)2++[122]
Ruíz-Tovar JCase seriesNo checklist required3[123]
Sakhaee KRCTAcceptable (+)1+[124]
Sakhaee KRCTAcceptable (+)1+[125]
Salgado WCase seriesNo checklist required3[126]
Sallé ACase seriesNo checklist required3[127]
Sánchez ATransversal studyNo checklist required3[128]
Santarpia, LCase seriesNo checklist required3[129]
Schiavo LPre-post studyNo checklist required3[130]
Schijns WCohort studyAcceptable (+)2+[131]
Schneider JRCTAcceptable (+)1+[132]
Schollenberger AERCTAcceptable (+)1+[133]
Shah MCase seriesNo checklist required3[134]
Sheng BSystematic reviewHigh quality (++)1+[135]
Sherf-Dagan STransversal studyNo checklist required3[136]
Sjöström LCohort studyAcceptable (+)2+[137]
Souza Silva JPre-post studyNo checklist required3[138]
Susmallian SCase seriesNo checklist required3[139]
Susmallian SCase seriesNo checklist required3[140]
Tang LCase seriesNo checklist required3[141]
Tondapu PRCTAcceptable (+)1+[142]
Topart PCase seriesNo checklist required3[143]
Tran DDSystematic reviewLow quality (−)1−[144]
Upala SSystematic reviewHigh quality (++)1+[145]
Van Nieuwenhove YRCTAcceptable (+)1+[146]
Van Rutte PWJPre-post studyNo checklist required3[147]
Verger EOCase seriesNo checklist required3[148]
Vinan-Vega MCase-control studyAcceptable (+)2+[149]
Wang CCase seriesNo checklist required3[150]
Wang FGSystematic reviewHigh quality (++)1++[151]
Ward EKRetrospective cohort studyAcceptable (+)2+[152]
Wolf ETransversal studyNo checklist required3[153]
Wei JHPre-post studyNo checklist required3[154]
Wang TCSystematic reviewHigh quality (++)1++[155]
White MGCase seriesNo checklist required3[156]
Wu Chao Ying VSystematic reviewHigh quality (++)1++[157]
Yorke ECase seriesNo checklist required3[158]
Yska JPCase seriesNo checklist required3[159]
Zhang QSystematic reviewHigh quality (++)1++[160]
Abbreviations: NA, Not Applicable; NRCT Non-Randomised Controlled Trial; RCT, Randomised Controlled Trial.
Table 4. VLCD main characteristics.
Table 4. VLCD main characteristics.
Total Caloric Value (Kcal/day)450–800
Carbohydrate contentAt least 55 gr/day
Protein content50 gr/day (high biological value)
Lipids7 gr
Linoleic acid3 gr
Alpha-linolenic acid0.5 gr
Fibre 10 gr
Vitamins, micronutrients and trace elements100% of daily requirements
Table 5. Follow-up analytical recommendations.
Table 5. Follow-up analytical recommendations.
Pre-Surgery1 Month3 Months6 Months12 MonthsAnnual
CBC/BiochemistryXXXXXX
AlbuminXXXXXX
PrealbuminXOptionalOptionalOptionalOptionalOptional
CRPXXXXXX
Iron/FerritinX XXXX
Ca/P/MgX XXXX
iPTH XXX
B12/Folic acidXX * XAXAXA
Vitamin DX XXX
Zn/Cu OptionalOptionalOptional
B1 OptionalOptionalOptional
Vitamin A and E XX AX A
* It is advisable to request in case of preoperative deficit. In other cases, optional. A Mandatory in malabsorptive surgery, optional in restrictive. X: must be done.
Table 6. Comparison between the composition of different multivitamins available in Spain.
Table 6. Comparison between the composition of different multivitamins available in Spain.
Recommendations of the American Society for Metabolic and Bariatric Surgery (ASMBS)Multicentrum (Per tablet)Multi-Tenex (Per tablet)Supradyn (Per tablet)Micebrina Complex
Vit A, μg1500–3000800800800450
Vit B1, mg1.21.41.41.110
Vit B12, μg350–5002.512.512
Vit D3, μg7555510
Vit E, mg1515101230
Vit K, μg90–12030-25-
Copper, mg1–20.5212
Iron, mg45–605141418
Zinc, mg8–225141015
Calcium, mg1200–15001621001200.15 (Calcium iodate)
Table prepared according to data provided by the manufacturer.
Table 7. Final recommendations/suggestions reached in the present review.
Table 7. Final recommendations/suggestions reached in the present review.
RecommendationGrade of Recommendation/Consensus Level
It is convenient to determine the levels of certain micronutrients and vitamins preoperatively, at least vitamin D and iron metabolism. Folic acid and B12 vitamin should be included in certain populations D/93%
Specific micronutrient supplements should be used if there is any evidence of any preoperative deficit following the current treatment recommendationsD/100%
We recommend the use of a liquid VLCD diet preoperatively, for at least 4-8 weeks minimum prior to surgery and ideally for a longer length of time in selected patientsB/91%
After surgery, a liquid diet should be maintained for about 4 weeks, and then a semi-solid diet for another 4 weeksD/84%
The GARIN group advises against calculating the protein provision based on a percentage of the diet’s total caloric value, since this method often results in insufficient intake. Instead, it is advisable to use a direct calculation based on the adjusted weight, at least 1 to 1.5 gr of high biological value protein per Kg of weight and dayD/96%
The use of protein supplements could be beneficial in the 6-12 months after surgeryB/96%
The postoperative use of calcium (1000 mg of calcium element at least) and vitamin D (880 IU of cholecalciferol) supplements are recommendedA/91%
In biliopancreatic diversion/Scopinaro surgeries the GARIN group recommends a higher intake of calcium (2000 mg/d) and especially a higher intake of vitamin D (2000 IU/d)D/91%
Periodic monitoring of iron levels after surgery should be performed, and in the case of deficit, treated accordinglyD/85%
Use of parenteral treatment for vitamin B12 deficiency only if the deficit is evidentD/85%
Although there is no scientific evidence, a consideration of the pathophysiological mechanism of Obesity Surgery, especially malabsorptive surgery, would make an increase of the dietary intake of other micronutrients, including supplements recommendableD/89%
Calcium citrate preparations should be recommended above other calcium compounds, especially in RYGBPB/93%
The GARIN group suggest periodic and customised analytical follow-up after surgery. Vitamin A, E, B12 and folic acid are mandatory in malabsorptive surgery (See text for details)D/89%
The GARIN group recommends individualising the use and duration of PPI therapyD/96%
We recommend the systematic use of multivitamin and mineral complexesC/98%
The GARIN group recommends, whenever available, the use of supplements that are specifically designed for patients undergoing Obesity SurgeryD/85%
We recommend periodical kidney function monitoring using serum creatinine and specific formulas to estimate glomerular filtration, occasional 24-hourour urine calcium, and, in selected cases, imaging tests, at least in patients who underwent RYGBP to rule out kidney stones. D/84%
After one year post-surgery, the GARIN group recommends annual check-ups in Specialised Care for at least five years of all patients who underwent Obesity Surgery. After this time, it is advisable to maintain annual check-ups in patients who underwent malabsorptive techniques, while those patients without complications who underwent purely restrictive techniques do not require specialised follow-up, except in selected casesD/87%
The GARIN group recommends that in patients with T2DM, RYGBP should be considered before LSG; independently of the technique used, it should be performed by a Surgical Team with experience in that techniqueD/91%
Pregnancy should be avoided in the first year after Obesity Surgery, and contraceptive measures should be routinely recommendedD/87%
All women of childbearing age undergoing Obesity Surgery should programme their gestation at an experienced centreD/87%
Possible micronutrient deficits (Vitamin A, D, E, K, B12, folic, iron) should be detected (AND treated) at least every six months prior to gestation, in each trimester of pregnancy (or sooner if any type of deficit is detected ) and at 6-8 weeks post-partum, especially in case of breastfeeding.C/87%
Breastfeeding is encouragedD/87%
In selected adolescent population between 13 and 18 years LSG can be considered for weight lossD/87%
In selected adolescent patients between 13 and 18 years, RYGBP can be considered for weight loss, especially in patients with prediabetes/T2DM, hypertension and/or dyslipidaemiaC/87%
BGYR should be discouraged in people over 65, especially in cases of high cardiovascular riskA/87%
In selected patients over 65 years LSG could be considered as an optionB/87%
If re-intervention is needed due to poor results in weight loss, RYGBP after LSG is an effective and safe option, allowing additional weight loss and improvement of comorbidities. If the first surgery was RYGBP, biliopancreatic diversion seems to be the most favourable techniqueB/75%

Share and Cite

MDPI and ACS Style

Martínez-Ortega, A.J.; Olveira, G.; Pereira-Cunill, J.L.; Arraiza-Irigoyen, C.; García-Almeida, J.M.; Irles Rocamora, J.A.; Molina-Puerta, M.J.; Molina Soria, J.B.; Rabat-Restrepo, J.M.; Rebollo-Pérez, M.I.; et al. Recommendations Based on Evidence by the Andalusian Group for Nutrition Reflection and Investigation (GARIN) for the Pre- and Postoperative Management of Patients Undergoing Obesity Surgery. Nutrients 2020, 12, 2002. https://doi.org/10.3390/nu12072002

AMA Style

Martínez-Ortega AJ, Olveira G, Pereira-Cunill JL, Arraiza-Irigoyen C, García-Almeida JM, Irles Rocamora JA, Molina-Puerta MJ, Molina Soria JB, Rabat-Restrepo JM, Rebollo-Pérez MI, et al. Recommendations Based on Evidence by the Andalusian Group for Nutrition Reflection and Investigation (GARIN) for the Pre- and Postoperative Management of Patients Undergoing Obesity Surgery. Nutrients. 2020; 12(7):2002. https://doi.org/10.3390/nu12072002

Chicago/Turabian Style

Martínez-Ortega, Antonio J., Gabriel Olveira, José L. Pereira-Cunill, Carmen Arraiza-Irigoyen, José M. García-Almeida, José A. Irles Rocamora, María J. Molina-Puerta, Juan B. Molina Soria, Juana M. Rabat-Restrepo, María I. Rebollo-Pérez, and et al. 2020. "Recommendations Based on Evidence by the Andalusian Group for Nutrition Reflection and Investigation (GARIN) for the Pre- and Postoperative Management of Patients Undergoing Obesity Surgery" Nutrients 12, no. 7: 2002. https://doi.org/10.3390/nu12072002

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

Article Metrics

Back to TopTop