Open access peer-reviewed chapter

Endoscopic Methods in Obesity Treatment

Written By

Anıl Ergin and Cihan Şahan

Submitted: 28 January 2024 Reviewed: 01 February 2024 Published: 07 March 2024

DOI: 10.5772/intechopen.1004427

From the Edited Volume

Weight Loss - A Multidisciplinary Perspective

Hubertus Himmerich

Chapter metrics overview

49 Chapter Downloads

View Full Metrics

Abstract

Obesity is a multifactorial, chronic disease that occurs with a pathologic increase in the body fat ratio and significantly increases mortality and morbidity. It has become a global health problem with increasing prevalence day by day. Methods used in the treatment of obesity are classified as diet, exercise, lifestyle changes, medical treatments, surgical treatments and endoscopic treatments. Endoscopic treatments are classified as intragastric balloon, transpyloric shuttle, endoscopic sleeve gastroplasty, gastric aspiration, small bowel procedures, duodenal mucosal resurfacing, intragastric botulinum toxin A injection. Although surgical procedures are known as the most effective methods in the fight against obesity today, the frequency and effectiveness of endoscopic treatments are increasing day by day. Endoscopic methods in obesity treatment are promising. There is a need for new methods with high efficacy and reliability, easy application, low complication rate and low cost in the treatment of obesity.

Keywords

  • obesity
  • treatment
  • endoscopy
  • intragastric baloon
  • transpyloric shuttle
  • endoscopic sleeve gastroplasty
  • gastric aspiration
  • duodenal mucosal resurfacing
  • intragastric botox

1. Introduction

Obesity is a multifactorial, chronic disease that occurs with a pathologic increase in the body fat ratio and significantly increases mortality and morbidity. It has become a global health problem with increasing prevalence day by day [1]. Between 1980 and 2002, the prevalence of obesity doubled in adults (aged over 19 years) and tripled in children and adolescents (aged between 6 and 19 years) [2]. Obesity has complex etiologic causes that cannot be defined as overeating alone. Genetic and environmental interactions, behavioral disorders, metabolic and endocrine disorders play an intertwined role in the etiology of obesity. Most of them occur as a result of a series of vicious cyclical events that can be explained in a cause and effect relationship. Although genetic and neuroendocrine factors are rarely encountered, environmental factors constitute the basis of the etiology of obesity [3].

In the diagnosis and evaluation of obesity, the patient’s chronological history of weight gain, the age at which the patient started to gain weight, the presence of overweight people in the family, the ages of pregnancy, child rearing and menopause, which are favorable periods for weight gain for female patients, how overweight affects the patient’s health, and the patient’s expectations from treatment should be questioned. During physical examination, diseases that may cause obesity and the main accompanying comorbidities; coronary artery disease, peripheral artery disease, type 2 diabetes mellitus, obstructive sleep apnea syndrome should be questioned [4].

Anthropometric measurements are used to define and classify obesity. These include methods such as body mass index, skinfold thickness and circumference measurements [5, 6].

Advertisement

2. Obesity treatment methods

2.1 Diet, exercise, lifestyle changes

First of all, realistic goals should be set. Changes should be made in the person’s eating habits and physical activity habits. In addition, activity and diet planning should be increased gradually. Access to food intake should be made difficult and self-control should be ensured. Alternative behaviors against eating stimuli should be tried to be developed. Regular weight checks should be performed and social support should be provided if necessary [7].

Diet therapy is applied to ensure a healthy and balanced diet, taking into account the gender, lifestyle, age and physical activity status of the person. It is very important in diet therapy to regulate the wrong eating habits in the process of becoming obese. After the body weight reaches the desired level in diet therapy, it is also necessary to maintain this level and to maintain eating habits for life. In diet therapy for children as well as adults, diet programs are made to maintain development and growth [8].

Exercise, which is one of the methods and essentials of effective weight loss, is one of the types of physical activity. Exercise, which is effective in the treatment of obesity, should be done at moderate intensity, on average 5 times a week and each exercise should be at least 30 minutes. Exercises that should be done in a disciplined manner can be shaped according to the age, gender, living conditions and sports history of the person [9].

In addition to moderate exercises, the person should also do resistance exercises that will work muscles such as the abdomen, hips, legs, shoulders and back. These exercises, which will help to lose weight and help the body reach the desired index, should be done 2 or 3 times a week. The energy burning of all these exercises is valid not only during the activity but also after the activity [9].

In addition to diet, weekly exercise programs are one of the factors that prevent the person from gaining weight again. Regular exercise should be continued after reaching the desired weight. In addition to weight loss, exercise also makes people feel good about themselves. Loss of confidence during obesity can be regained with exercise and it is effective in preventing many diseases. Exercise reduces the risk of many diseases such as decreased insulin resistance, cardiovascular, hypertension, diabetes [9].

Lifestyle changes supported by healthy nutrition and exercise are indispensable conditions for the treatment of obesity.

2.2 Medical treatment

Drug therapy can be used in the treatment of overweight and obese patients in addition to diet, exercise and lifestyle changes. However, problems such as the efficacy and safety of drugs, slowing and plateauing of weight loss during treatment, and weight regain when the drug is discontinued are the limiting points of drug treatment in obesity. Drug treatment can be used for those with a BMI >30 kg/m2 and those with a BMI of 27–29.9 kg/m2 with additional morbidity [10].

The rationality of the targets set is of great importance in the success of treatment. Targeting the ideal normal body weight is not a realistic approach. Therefore, physicians and patients should have a realistic approach to weight loss treatment at the beginning. Success in treatment is measured by the amount of weight lost and improvement in existing risk factors. Weight loss exceeding 2 kg in the first month of drug treatment, losing 5% of the basal weight in the 3rd–6th months and staying at this level can be considered an effective treatment. A 5–10% weight loss significantly reduces the risk for diabetes and cardiovascular disease [10, 11].

Drug treatment does not provide a curative result in obesity. During treatment, weight loss stops after maximal treatment response is achieved. After drug treatment is discontinued, weight gain may occur again. Obese patients with concomitant diabetes, depression and cardiovascular diseases should be advised to choose treatments that have a weight loss effect rather than weight gain when planning drug treatment for these conditions. Currently, the drugs approved by the US Food and Drug Administration (FDA) for the treatment of obesity are phentermine, diethylpropion, phendimetrazine, benzfetamine, orlistat, lorcaserin, phentermine/topiramate-Extended Release (ER) combination. Of these drugs, only orlistat, lorcaserin, phentermine/topiramate-ER are approved by the FDA for long-term use. The others are only approved for short-term (a few weeks) use. All drugs are approved for use in patients with a BMI ≥ 30 kg/m2. Benzfetamine and diethylpropion can also be used in patients with a BMI ≥ 27 kg/m2 and an additional obesity-related comorbidity (hypertension, diabetes). Recently, drugs with the active ingredients semaglutide and liraglutide have also been used in the treatment of obesity and show promise [12].

2.3 Surgical treatment

In 1991, the US National Institutes of Health Consensus Conference decided that surgery should be considered as a treatment in selected patients when it became clear that non-surgical treatments resulted in little weight loss and subsequent weight regain. At the same time, the committee recommended that these patients should be evaluated by a multidisciplinary team before surgery and followed up for life after the operation is performed under appropriate conditions [13, 14, 15].

If we look at the historical development of bariatric surgery, the first operations were performed in the 1950s to treat severe hyperlipidemia associated with obesity. These operations were ileocolic bypass operations to limit absorption and caused severe postoperative nutritional complications and liver failure. The jejunoileal bypass was then developed and became popular in the mid-1970s [13, 14].

In 1969, Mason and Ito performed the first gastric bypass, which they described as the connection of a jejunum ring to a transverse proximal gastric pouch. Postoperative bile reflux esophagitis was severe and Griffin et al. described the roux-n-Y modification of the gastric bypass in 1977 [14, 15, 16, 17].

In 1980, Mason performed the first VBG. This surgery initially achieved excellent weight loss with low mortality and morbidity and became the most commonly performed bariatric operation in the US in the 1980s. However, from the early 1990s, patients who underwent VBG surgery regained weight. In the long term, weight loss was small and by the 1990s, the roux-n-Y gastirk bypass became the preferred operation of bariatric surgery in the US [18, 19].

In Italy, Scopinaro developed biliopancreatic diversion in the 1980s. This operation, together with its modification including duodenal switch, has been the only malabsorptive operation with long-term success. It represents less than 5% of the operations performed in the USA [19, 21].

Laparoscopic bariatric surgery started in the 1990s. Belachew performed the first laparoscopic adjustable gastric band operation in 1994. Wittgrove and Clark performed the first laparoscopic RYGB in the same year. In 2001, the application of LAGB was approved in the USA. Its popularity increased until 2009, but has declined since then. Laparoscopic sleeve gastrectomy (LSG) has enjoyed a rapidly increasing popularity in the US and internationally since 2008 [21].

The most commonly used bariatric surgery methods today are Laparoscopic Sleeve Gastrectomy (LSG), Roux n Y Gastric Bypass (RYGB), Single Anastomosis Gastric Bypass (OAGB), Transit Bipartition (TB), Biliopancreatic diversion and duodenal switch (BPD-DS), Single Anastomosis Duodeno-ileal bypass (SADI-S), Single Anastomosis Sleeve-Ileal Bypass (SASI Bypass) [18, 19, 20, 21].

2.4 Endoscopic treatment methods

The advantages of endoscopic methods used in the treatment of obesity are that they are minimally invasive, have a reversible effect, are reproducible, are a cost-effective treatment option, and can be used for bridging before surgical treatment. Endoscopic bariatric procedures are divided into two categories: interventions in the stomach and interventions in the small bowel. Gastric interventions include gastric balloon, transpyloric shuttle, endoscopic sleeve gastroplasty, pose and gastric aspiration. Interventions in the small intestine include duodenojejunal diversion, gastroduodenojejunal bypass, jejunoileal diversion and duodenomucosal resurfacing/remodeling (Figure 1) [22].

Figure 1.

View of endoscopic treatment methods [22].

2.4.1 Intragastric balloon

Intragastric balloon application is one of the most commonly used endoscopic bariatric methods today. In 2015, it received FDA approval for use in the treatment of obesity. While the peripheral effects of intragastric balloon application are known as reducing gastric volume and slowing gastric emptying; central effects can be counted as activation of gastric tension receptors, signal transmission to the hypothalamus via vagal nerve and early satiety. In 2022, indications for use were specified in the Indications for Metabolic and Bariatric Surgery guidelines published by ASMBS and IFSO. In addition, the areas and conditions of use are also specified in the Spanish and Brazilian intragastric balloon application consensus [23, 24, 25].

According to the Brazilian intragastric balloon consensus agreement, indications are defined as being over 12 years of age (no upper age limit (signed by both parents)), body mass index over 25 and failure of diet, exercise and lifestyle changes, and bridge treatment before bariatric surgery. Contraindications were defined as the presence of active peptic ulcer, presence of gastric or esophageal varices, presence of hiatal hernia larger than 5 cm, and previous gastric surgery. According to this consensus, the intragastric balloon should be removed in case of intolerance (severe nausea and vomiting attacks, severe pain), recurrent fluid-electrolyte disturbance, balloon deflation (methylene blue—72 hours), gastrointestinal bleeding, moderate to severe pancreatitis and pregnancy (2nd trimester) during treatment [24, 25].

Ideal balloon characteristics are defined as a material with a smooth surface, made of durable material, inflatable with liquid or air, adjustable in volume, with a radio-opaque marker to facilitate follow-up [23, 24, 25]. The most commonly used intragastric balloon types today are Orbera, ReShape Duo, Obalon, Heliosphere, Spatz and Elipse. Some characteristics of intragastric balloon types are summarized in Table 1.

  • Obese patients undergoing IGB treatment require concomitant dietary and lifestyle modification.

  • PPI prophylaxis should be performed in individuals undergoing IGB therapy.

  • Intraoperative anesthetics associated with the lowest incidence of nausea should be used during the procedure.

  • A planned anti-emetic regimen is recommended for 2 weeks post-procedure.

  • Multivitamin supplementation at a daily dose of 1–2 adult doses (Figure 2) [23, 24, 25].

Summary of Intragastric balloon characteristics
FDA/CE approvedCE approved
Balloon typeOrberaReShape DuoObalonHeliosphereSpatzElipse
ManufacturerApollo EndosurgeryReShape MedicalObalon TherapeuticsHelioscopie Medical ImplantsSpatz FGIAAllurion Technologies
Filled withSalineSalineNitrogen gasAirSalineLiquid
Capacity (mL)400-700450 × 2250 × 3900–1000300–900550
Number of balloons12Up to 3111
InsertionEndoscopyEndoscopySwallowedEndoscopyEndoscopySwallowed
RemovalEndoscopyEndoscopyEndoscopyEndoscopyEndoscopyNatural pass
Duration6666124
AdjustableNoNoNoNoYesNo

Table 1.

Characteristics of intragastric balloon types [22].

Figure 2.

Intragastric balloon material.

2.4.2 Trans Pyloric Shuttle

Trans Pyloric Shuttle (TPS) is an endoscopic, minimally invasive method for the treatment of obesity. The TPS is an apparatus that has two ends and is connected to each other with flexible silicone. When the larger part is inflated and filled, it takes the shape of a bulb. The large part of the TPS apparatus, which is delivered to the stomach with an endoscopic application device, is inflated and filled with a coil-like mechanism. After the TPS is endoscopically inserted into the stomach, it remains free in the stomach. Since the small part can easily pass through the pylorus, it passes through the pylorus with peristaltic movements and delays gastric emptying due to the intermittent formation of the large part in the pylorus and at the same time accelerates the filling time of the stomach. This device can stay in the stomach for 12 months and at the end of this period, the internal coil in the large bulb-like part of the TPS is unlocked endoscopically, the part inside the coil is removed first, and the large part is taken out of the stomach after it is narrowed (Figures 3 and 4).

Figure 3.

Transpyloric shuttle [26].

Figure 4.

Image of the transpyloric shuttle in the stomach [26].

After the results of the Endobesity II study were published, TPS received FDA approval in 2019. According to this study, TWL was 9.5% in patients who underwent TPS and 2.8% in the control group after 12 months of follow-up. In terms of serious side effects, 4.69% of patients who underwent TPS experienced side effects, while this rate was 1.12% in the control group [26].

2.4.3 Endoscopic sleeve gastroplasty

Endoscopic sleeve gastropilasty (ESG) is a globally accepted method for the treatment of obesity. Although bariatric surgery is the most effective method in the treatment of obesity, many patients avoid surgery because it is perceived as invasive and fear of complications. ESG is a less invasive and organ-sparing option in the treatment of obesity. Endoscopic gastroplasty alters the anatomy and physiology of the stomach by folding the gastric walls and reducing the intragastric volume by up to 75%. Functional exclusion of part of the stomach results in a reduction of both gastric volume and motility [27].

ESG is performed with two types of FDA-approved devices: Overstitch platform (Apollo Endosurgery, Austin, TX), Primary Obesity Surgery Endoluminal (POSE) (USGI Medical, San Clemente, CA).

The Overstitch platform (Apollo Endosurgery, Austin, TX) is used to endoscopically perform gastroplasty with full-thickness suturing and plication of the greater curvature from the inside. This procedure is the most commonly performed endoscopic gastroplasty in recent years. ESG has been shown to be beneficial for weight loss by both reducing gastric volume and gastric emptying [27]. It also has effects on gut hormones and has been shown to reduce ghrelin levels [27]. The most common side effects are nausea and vomiting and abdominal pain. Leakage and free fluid around the stomach, gastric bleeding, some cases of venous thromboembolism and other complications such as pneumotrax, pneumperitnyum have also been seen [27, 28].

Contraindications for ESG, as with other endoscopic options for the treatment of obesity, include large hiatal hernia, gastric ulcer, gastric tumors or intestinal metaplasia, previous surgical procedure, pregnancy, continuous monitoring and anticoagulant use [27, 28].

There are many studies in the literature on this technique. Seven studies including 6775 patients (3413 with ESG and 3362 with LSG) were included in Beran et al. ESG and LSG were compared in the short and long term and significant differences in TBWL% were seen in favor of LSG. The trend towards a lower incidence of adverse events in ESG compared with LSG did not reach statistical significance (RR 0.51, 95% CI 0.23–1.11, P = 0.09). The incidence of new-onset gastroesophageal reflux disease (GERD) was significantly lower after ESG compared to LSG, 1.3% versus 17.9%, respectively (RR 0.10, 95% CI 0.02–0.53, P = 0.006). ESG provided clinically adequate but lower short- and medium-term weight loss and resulted in fewer adverse events, including GERD, compared to LSG. Given the gastroprotective nature of ESG and its acceptable safety profile, it may be considered as an alternative to LSG for patients with mild to moderate obesity [28].

In a study by Lopez-Nava et al. with 248 patients, TBWL was 18.5% after 24 months of follow-up in patients who underwent ESG [29].

In the 91-patient study by Sharaiha et al. TBWL was 17.6% after 1-year follow-up (76% patient follow-up rate). At the same time, the reduction in medical comorbidities, HgA1 C, Blood Pressure, Triglyceride and ALT values were statistically significant [30].

In another study of 148 patients, endolumenal sleeve gastrolasty resulted in a TWBL of 17.53 ± 7.57% at 1 year and 18.5 ± 9% at 18 months. Leptin hormone levels unexpectedly decreased in every patient. As a complication, only 1 patient had a hemorrhage during the passage of the suture helix, which was resolved by sclerotherapy. In this study by Moreles et al., a continuous suture line was created using 4 parallel “Z” suture patterns to minimize the gastric cavity. The suture pattern aimed to eliminate the effect of the distorting force on the suture and to provide a homogeneous distribution between all suture points [31].

Comparison of ESG studies is very difficult due to the difference in technical procedures. However, many studies have been conducted by describing continuous sutures.

Primary obesity surgery endoluminal (POSE) uses an incisionless surgical system to create full-thickness plications in the gastric fundus and body. Many studies have demonstrated the safety and efficacy of the original POSE for the treatment of obesity.

In the meta-analysis by Singh et al. 7 studies were included, 2 of these studies were randmized controlled studies while the others were observational studies. At the end of 12–15 months follow-up, EWL was 48.86% and TWL was 12.68%. Serious adverse events included GI bleeding, extra-gastric bleeding, hepatic abscess, severe pain, severe nausea and severe vomiting; the overall incidence of these serious adverse events was only 2.84% [32] (Figure 5).

Figure 5.

Illustration showing the differences between the POSE 1 and POSE 2 procedures.

POSE 1 anatomically reduces the gastric fundus and places a plication line horizontally into the distal gastric body. In POSE 2, by placing plications in the greater curvature of the stomach starting from the level of the incisura and extending proximally to the corpus, the volume of the stomach is reduced so that a small reservoir of food remains in the fundus and the stomach is somewhat reduced and shortened. Both ESG and POSE have shown both gastric volume reduction and hormonal efficacy when compared with other endoscopic procedures. In addition, both ESG and POSE procedures have similar efficacy [33].

2.4.4 Gastric aspiration therapy

The AspireAssist device (Aspire Bariatrics, King of Prussia, PA) is a type of endoscopic gastrostomy tube. It works by partially aspirating ingested food. It is reversible and does not cause significant changes in the anatomy of the stomach. First, the AspireAssist device is inserted. Gastric content is aspirated by feeding water through the part connected to the external apparatus.

In the pilot study conducted by Sullivan et al. in 18 individuals with obesity, 11 patients randomly assigned to aspiration therapy and lifestyle therapy, and 7 patients to lifestyle therapy only. Lifestyle therapy consisted of a 15-session dietary and behavioral education program. After 1 year, TBWL % was 18.6% ± 2.3% in the aspiration therapy group and TWBL % was 5.9% ± 5.0% in the lifestyle therapy group. There was no significant difference between the two groups (p < 0.4) (Figure 6) [34].

Figure 6.

Aspire Bariatrics AspireAssist. Notes: (A) aspiration tube and skin port in place and (B) external device connected for aspiration [34].

2.4.5 Small bowel procedures

Duodenal bypass sheaths are closed sheaths placed between the mucosa and the GI lumen to block the digestion and absorption of nutrients. Currently, these devices are not FDA approved. Most of the literature mentions the EndoBarrier, a duodenal-jejunal bypass sleeve (DJBS) with a length of 60 cm. During insertion, a self-expanding nitinol anchor is placed inside the duodenal bulb and an impermeable fluoropolymer liner is placed distal to the proximal jejunum. Similarly, the ValenTx is a 120 cm gastro-duodenal-jejunal bypass that is fixed at the gastroesophageal junction and extends into the jejunum. Laparoscopic assistance is required for the placement of this second device [35, 36, 37, 38].

A rapid improvement in T2DM and glycemic control, as well as hormonal changes, has been noted after DJBS implantation [39, 40]. Munoz et al. [41] suggest additional weight-independent glycemic control mechanisms. Jirapinyo et al. [42] published a meta-analysis of 4 RCTs showing a 1.3% reduction in HbA1c and a significant reduction of 0.9% compared to control. This reduction was maintained after 6 months. Pooled data from observational and RCTs showed a TWL of 18% that remained significant after 1 year; GLP-1, PYY and ghrelin increased while glucose-dependent insulinotropic polypeptide decreased [41, 42].

There are still safety issues in using these methods that need to be addressed. Betzel et al. [43] published a systematic review of 1057 patients, of whom 33 had serious adverse events, or 3.7%. These included 11 hepatic abscesses, 8 gastrointestinal bleeding, 4 esophageal perforation and 3 acute pancreatitis. 8 patients required surgery but no deaths were reported. Despite these risks, Laubner et al. [44] showed a possible positive risk-benefit ratio in favor of DJBL for the treatment of T2DM.

Currently, bypass procedures are performed laparoscopically with stapler or manual anastomosis. Similarly, performing anastomoses is endoscopically challenging. For the treatment of obesity, type II diabetes and malignant obstructions, a completely endoscopic method of creating a durable reliable bowel bypass is desirable [45, 46, 47].

Ryou et al. performed endoscopic jejuno-ileal anastomosis in an animal experiment on 8 pigs. Since the anatomy of the pigs was not suitable, ileal magnets were placed laparoscopically and anastomoses were checked after 3 months in 8 pigs and it was observed that the anastomoses were successful [48]. In the pilot study conducted by Sullivan et al., enteroscopy and colonoscopy were performed in 10 people with obesity and magnets were placed in jejunum and ileum. With the help of laparoscopy, the correct position of the magnets was controlled and adhesion was assisted. Successful anastomoses were performed in normal form. Common side effects were vomiting and diarrhea. At the end of 6 months, TWL was 10.6% (EWL was 28%). HbA1C levels decreased in 4 patients with diabetes [49].

2.4.6 Dudenal mucosal surfacing

Duodenal mucosal resurfacing is performed using specially designed catheters (Fractyl Laboratories) advanced through a guidewire next to the endoscope. Duodenal mucosal resurfacing (DMR) is a single, minimally invasive endoscopic procedure that involves circumferential hydrothermal ablation of the duodenal mucosa followed by regeneration of the mucosa. Before ablation, the mucosa is removed with saline to protect the outer layers of the duodenum. A first-in-human study showed significant improvements in glycemia in T2D patients after DMR and suggested a positive correlation between the length of the ablated segment and efficacy [50]. Baar et al. evaluated 36 patients and found that DMR is a feasible and safe endoscopic procedure that provides durable glycemic improvement in suboptimally controlled T2D patients on oral glucose-lowering medication regardless of weight loss [51].

2.4.7 Intragastric botulinum toxin a (BTA) injection

Botulinum toxin A (BTA) injection may temporarily inhibit gastric peristalsis by paralyzing the muscular layer of the stomach. This can delay gastric emptying and prolong the duration of satiety. Conflicting evidence on efficacy has been published. Early studies showed weight loss through delayed gastric emptying without side effects [52, 53, 54]. In 2015, a meta-analysis of 8 studies concluded that BTA is effective in the treatment of obesity [55]. A recent meta-analysis concluded that intragastric BTA injection is not effective [56]. In another study, intragastric injection of BTX-A does not seem to be an effective method to achieve preoperative weight loss in super obese patients [57]. In a systemic meta-analysis conducted by Yen et al. [58], 192 individuals were evaluated in 6 randomized controlled studies. Intragastric BTA injection was found to be effective in the treatment of obesity, adequate dose (≥200 U), multiple gastric injection sites and combined diet control are very important. However, caution should be exercised due to the small sample size and limited power (Figure 7) [58].

Figure 7.

Endoscopic interventions for obesity and dysmetabolic conditions. Endoscopic interventions: (a) duodenal mucosa resurfacing, (b) intragastric botox injection, (c) gas-filled intragastric balloon, (d) liquid-filled intragastric balloon, (e) transpyloric shuttle, (f) duodenal-jejunal bypass liner, (g) gastro-duodenal-jejunal bypass liner, (h) endoscopic sleeve gastroplasty, (i) primary obesity surgery endoluminal, (l) aspiration therapy, and (m) partial jejunal diversion [36].

Advertisement

3. Conclusion

Although surgical procedures are known as the most effective methods in the fight against obesity today, the frequency and effectiveness of endoscopic treatments are increasing day by day. Endoscopic methods in obesity treatment are promising. There is a need for new methods with high efficacy and reliability, easy application, low complication rate and low cost in the treatment of obesity.

References

  1. 1. Babinska Z, Bandosz P, Zdrojewski T, Wyrzykowski B. Epidemiologia otyłosci i otyłosci brzusznej w Polsce, Europie Zachodniej i USA. Kardiologia w Praktyce. 2004;5:3-7
  2. 2. Ogden CL, Carroll MD, Curtin LR, McDowell MA, Tabak CJ, Flegal KM. Prevalence of overweight and obesity in the United States, 1999-2004. JAMA. 2006;295:1549-1555
  3. 3. Gülay H. Temel ve sistematik cerrahi, Güven Kitabevi, İzmir, Turkey. 2005. ISBN: 9759241285, 9789759241285
  4. 4. Baştürk S. Sleeve gastrektominin orta ve uzun dönem sonuçları, obeziteye eşlik eden komorbiditelere Etkileri. Pamukkale Üniversitesi: Uzmanlık tezi; 2015
  5. 5. James WP. What are health risks? The medical consequences of obesity and its health risks. Experimental and Clinical Endocrinology & Diabetes. 1998;106(Suppl. 2):1-6
  6. 6. Kenney WL, Humphrey RH, Bryant CX, Mahler DA. ACSM’s Guidelines for Exercise Testing and Prescription. 5th ed. Baltimore: A Waverly Company; 1995. pp. 53-63
  7. 7. Xavier Pi-Sunyer F, Becker DM, Bouchard C, Carleton RA, Colditz GA, William H, et al. Clinical guidelines on the identification, evaluation, and treatment of overweight and obesity in adults. The Evidence Report NIH Publication No. 98-4083. National Institutes Of Health National Heart, Lung, And Blood Institute In Cooperation With The National Institute Of Diabetes And Digestive And Kidney Diseases. Sep 1998
  8. 8. Kaila B, Raman M. Obesity: A review of pathogenesis and management strategies. Canadian Journal of Gastroenterology. 2008;22:61-68
  9. 9. Jakicic JM, Otto AD. Physical activity considerations for the treatment and prevention of obesity. The American Journal of Clinical Nutrition. 2005;82(suppl):226-229
  10. 10. Knowler WC, Barret-Connor E, Fowler SE. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. The New England Journal of Medicine. 2002;346:393. DOI: 10.1056/NEJMoa012512
  11. 11. Douketis JD, Macie C, Thabane L, Williamson DF. Systematic review of long-term weight loss studies in obese adults: Clinical significance and applicability to clinical practice. International Journal of Obesity. 2005;29:1153-1167
  12. 12. Yanovski ZS, Yanovski AJ. Long-term drug treatment for obesity: A systematic and clinical review. JAMA. 2014;311:74-86
  13. 13. Gastrointestinal surgery for severe obesity: National Institutes of Health consensus development conference statement. American Journal of Clinical Nutrition. 1992;52(Suppl. 2):615-619
  14. 14. Schauer PR, Schirmer B. The surgical management of obesity. In: Brunicardi F, Andersen DK, Billiar TR, Dunn DL, Hunter JG, Matthews JB, et al, editors. Schwartz’s Principles of Surgery. 10th ed. McGraw-Hill Education; 2015. Available from: https://accessmedicine.mhmedical.com/content.aspx?bookid=980&sectionid=59610869 [Accessed: March 02, 2024]
  15. 15. Richards WO. Sabiston textbook of surgery. The Biological Basis of Modern Surgical Practice . 21st ed. Morbid Obesity, Chapter 69. Elsevier; Apr 2021. pp. 1160-1187. Hardback ISBN: 9780323640626, eBook ISBN: 9780323640640
  16. 16. Gloy VL, Briel M, Bhatt DL, Kashyap SR, Schauer PR, Mingrone G. Bariatric surgery versus non-surgical treatment for obesity: A systematic review and meta-analysis of randomised controlled trials. British Medical Journal. 2013;347:1-16
  17. 17. Pories WJ. Bariatric surgery: Risks and rewards. The Journal of Clinical Endocrinology and Metabolism. 2008;93(11):89-96
  18. 18. Buchwald H. Buchwald’s Atlas of Metabolic & Bariatric Surgical Techniques and Procedures. Philadelphia, PA: Elsevier/Saunders; 2012. DOI: 10.1016/C2009-0-34090-8
  19. 19. Buchwald H, Buchwald JN. Evolution of operative procedures for the management of morbid obesity 1950- 2000. Obesity Surgery. 2002;12(5):705-717
  20. 20. O'Brien P. Bariatric surgery: Mechanisms, indications and outcomes. Journal of Gastroenterology and Hepatology. 2010;25(8):1358-1365
  21. 21. Schneider BE, Mun EC. Surgical management of morbid obesity. Diabetes Care. Feb 2005;28(2):475-480. DOI: 10.2337/diacare.28.2.475. PMID: 15677820
  22. 22. Dayyeh BKA, Edmundowicz S, Thompson CC. Clinical practice update: Expert review on endoscopic bariatric therapies. Gastroenterology. Mar 2017;152(4):716-729
  23. 23. Neto MG, Silva LB, Grecco E, et al. Brazilian intragastric balloon consensus statement (BIBC): Practical guidelines based on experience of over 40,000 cases. Surgery for Obesity and Related Diseases. 2018;14(2):151-159
  24. 24. American Society for Metabolic and Bariatric Surgery. Metabolic and Bariatric Surgery: Fact Sheet. 2013. Available from: https://asmbs.org/wp/uploads/2014/05/Metabolic+Bariatric-Surgery.pdf [Accessed: Jul 18, 2016]
  25. 25. Fernandes M, Atallah AN, Soares BGO, et al. Intragastric balloon for obesity. Cochrane Database of Systematic Reviews. 2007;(1):CD004931
  26. 26. Transpyloric Shuttle/Transpyloric Shuttle Delivery Device Instructions for Use. Available from: https://www.clinicaltrials.gov/ct2/show/NCT02518685
  27. 27. Vargas EJ, Rizk M, Gomez-Villa J, Edwards PK, Jaruvongvanich V, Storm AC, et al. Effect of endoscopic sleeve gastroplasty on gastric emptying, motility and hormones: A comparative prospective study. Gut. 2023;72(6):1073-1080. DOI: 10.1136/gutjnl-2022-327816. Epub 2022 Oct 14
  28. 28. Beran A, Matar R, Jaruvongvanich V, Rapaka BB, Alalwan A, Portela R, et al. Comparative effectiveness and safety between endoscopic sleeve gastroplasty and laparoscopic sleeve gastrectomy: A meta-analysis of 6775 individuals with obesity. Obesity Surgery. 2022;32(11):3504-3512. DOI: 10.1007/s11695-022-06254-y. Epub 2022 Sep 2
  29. 29. Lopez-Nava G, Sharaiha RZ, Vargas EJ, Bazerbachi F, Manoel GN, Bautista-Castaño I, et al. Endoscopic sleeve gastroplasty for obesity: A multicenter study of 248 patients with 24 months follow-up. Obesity Surgery. 2017;27(10):2649-2655. DOI: 10.1007/s11695-017-2693-7
  30. 30. Sharaiha RZ, Kumta NA, Saumoy M, Desai AP, Sarkisian AM, Benevenuto A, et al. Endoscopic sleeve gastroplasty significantly reduces body mass index and metabolic complications in obese patients. Clinical Gastroenterology and Hepatology. 2017;15(4):504-510. DOI: 10.1016/j.cgh.2016.12.012. Epub 2016 Dec 23
  31. 31. Graus Morales J, Crespo Pérez L, Marques A, Marín Arribas B, Bravo Arribas R, Ramo E, et al. Modified endoscopic gastroplasty for the treatment of obesity. Surgical Endoscopy. 2018;32(9):3936-3942. DOI: 10.1007/s00464-018-6133-0. Epub 2018 Feb 28
  32. 32. Singh S, Bazarbashi AN, Khan A, Chowdhry M, Bilal M, de Moura DTH, et al. Primary obesity surgery endoluminal (POSE) for the treatment of obesity: A systematic review and meta-analysis. Surgical Endoscopy. 2022;36(1):252-266. DOI: 10.1007/s00464-020-08267-z. Epub 2021 Feb 1
  33. 33. Espinós JC, Turró R, Moragas G, Bronstone A, Buchwald JN, Mearin F, et al. Gastrointestinal physiological changes and their relationship to weight loss following the POSE procedure. Obesity Surgery. 2016;26(5):1081-1089. DOI: 10.1007/s11695-015-1863-8
  34. 34. Sullivan S, Stein R, Jonnalagadda S, Mullady D, Edmundowicz S. Aspiration therapy leads to weight loss in obese subjects: A pilot study. Gastroenterology. 2013;145(6):1245-52.e1-5. DOI: 10.1053/j.gastro.2013.08.056. Epub 2013 Sep 6
  35. 35. Abu Dayyeh BK, Acosta A, Camilleri M, Mundi MS, Rajan E, Topazian MD, et al. Endoscopic sleeve gastroplasty alters gastric physiology and induces loss of body weight in obese individuals. Clinical Gastroenterology and Hepatology. 2017;15(1):37-43.e1. DOI: 10.1016/j.cgh.2015.12.030. Epub 2015 Dec 31
  36. 36. Telese A, Sehgal V, Magee CG, Naik S, Alqahtani SA, Lovat LB, et al. Bariatric and metabolic endoscopy: A new paradigm. Clinical and Translational Gastroenterology. 2021;12(6):e00364. DOI: 10.14309/ctg.0000000000000364
  37. 37. Sandler BJ, Rumbaut R, Swain CP, Torres G, Morales L, Gonzales L, et al. Human experience with an endoluminal, endoscopic, gastrojejunal bypass sleeve. Surgical Endoscopy. 2011;25(9):3028-3033. DOI: 10.1007/s00464-011-1665-6. Epub 2011 Apr 13
  38. 38. Sandler BJ, Rumbaut R, Swain CP, Torres G, Morales L, Gonzales L, et al. One-year human experience with a novel endoluminal, endoscopic gastric bypass sleeve for morbid obesity. Surgical Endoscopy. 2015;29(11):3298-3303. DOI: 10.1007/s00464-015-4081-5. Epub 2015 Jan 29
  39. 39. de Jonge C, Rensen SS, Verdam FJ, Vincent RP, Bloom SR, Buurman WA, et al. Endoscopic duodenal-jejunal bypass liner rapidly improves type 2 diabetes. Obesity Surgery. 2013;23(9):1354-1360. DOI: 10.1007/s11695-013-0921-3
  40. 40. Cohen RV, Neto MG, Correa JL, Sakai P, Martins B, Schiavon CA, et al. A pilot study of the duodenal-jejunal bypass liner in low body mass index type 2 diabetes. The Journal of Clinical Endocrinology and Metabolism. 2013;98(2):E279-E282. DOI: 10.1210/jc.2012-2814. Epub 2013 Jan 21
  41. 41. Muñoz R, Escalona A. Duodenal-jejunal bypass liner to treat type 2 diabetes mellitus in morbidly obese patients. Current Cardiology Reports. 2014;16(3):454. DOI: 10.1007/s11886-013-0454-3
  42. 42. Jirapinyo P, Haas AV, Thompson CC. Effect of the duodenal-jejunal bypass liner on glycemic control in patients with type 2 diabetes with obesity: A meta-analysis with secondary analysis on weight loss and hormonal changes. Diabetes Care. 2018;41(5):1106-1115. DOI: 10.2337/dc17-1985
  43. 43. Betzel B, Drenth JPH, Siersema PD. Adverse events of the duodenal-jejunal bypass liner: A systematic review. Obesity Surgery. 2018;28(11):3669-3677. DOI: 10.1007/s11695-018-3441-3
  44. 44. Laubner K, Riedel N, Fink K, Holl RW, Welp R, Kempe HP, et al. Comparative efficacy and safety of the duodenal-jejunal bypass liner in obese patients with type 2 diabetes mellitus: A case control study. Diabetes, Obesity & Metabolism. 2018;20(8):1868-1877. DOI: 10.1111/dom.13300. Epub 2018 Apr 23
  45. 45. Ly J, O’Grady G, Mittal A, Plank L, Windsor JA. A systematic review of methods to palliate malignant gastric outlet obstruction. Surgical Endoscopy. 2010;24:290-297
  46. 46. Gentileschi P, Kini S, Catarci M, Gagner M. Evidence-based medicine: Open and laparoscopic bariatric surgery. Surgical Endoscopy. 2002;16:736-744
  47. 47. Duan J, Tan C, Xu H, et al. Side-to-side jejunoileal bypass induces better glucose-lowering effect than end-to-side jejunoileal bypass on nonobese diabetic rats. Obesity Surgery. 2015;25:1458-1467
  48. 48. Ryou M, Aihara H, Thompson CC. Minimally invasive entero-enteral dual-path bypass using self-assembling magnets. Surgical Endoscopy. 2016;30:4533-4538. DOI: 10.1007/s00464-016-4789-x
  49. 49. Sullivan S, Edmundowicz SA, Thompson CC. Endoscopic bariatric and metabolic therapies: New and emerging technologies. Gastroenterology. 2017;152(7):1791-1801. DOI: 10.1053/j.gastro.2017.01.044. Epub 2017 Feb 10
  50. 50. Rajagopalan H, Cherrington AD, Thompson CC, Kaplan LM, Rubino F, Mingrone G, et al. Endoscopic duodenal mucosal resurfacing for the treatment of type 2 diabetes: 6-month interim analysis from the first-in-human proof-of-concept study. Diabetes Care. 2016;39(12):2254-2261. DOI: 10.2337/dc16-0383. Epub 2016 Aug 12
  51. 51. Van Baar ACG, Holleman F, Crenier L, Haidry R, Magee C, Hopkins D, et al. Endoscopic duodenal mucosal resurfacing for the treatment of type 2 diabetes mellitus: One year results from the first international, open-label, prospective, multicentre study. Gut. 2020;69(2):295-303. DOI: 10.1136/gutjnl-2019-318349.Epub 2019 Jul 22
  52. 52. Foschi D, Corsi F, Lazzaroni M, Sangaletti O, Riva P, La Tartara G, et al. Treatment of morbid obesity by intraparietogastric administration of botulinum toxin: A randomized, double-blind, controlled study. International Journal of Obesity. 2007;31(4):707-712. DOI: 10.1038/sj.ijo.0803451. Epub 2006 Sep 26
  53. 53. Foschi D, Lazzaroni M, Sangaletti O, Corsi F, Trabucchi E, Bianchi Porro G. Effects of intramural administration of botulinum toxin A on gastric emptying and eating capacity in obese patients. Digestive and Liver Disease. 2008;40(8):667-672. DOI: 10.1016/j.dld.2008.02.040
  54. 54. Topazian M, Camilleri M, De La Mora-Levy J, Enders FB, Foxx-Orenstein AE, Levy MJ, et al. Endoscopic ultrasound-guided gastric botulinum toxin injections in obese subjects: A pilot study. Obesity Surgery. 2008;18(4):401-407. DOI: 10.1007/s11695-008-9442-x. Epub 2008 Feb 20
  55. 55. Bang CS, Baik GH, Shin IS, Kim JB, Suk KT, Yoon JH, et al. Effect of intragastric injection of botulinum toxin A for the treatment of obesity: A meta-analysis and meta-regression. Gastrointestinal Endoscopy. 2015;81(5):1141-9.e1-7. DOI: 10.1016/j.gie.2014.12.025. Epub 2015 Mar 9
  56. 56. Bustamante F, Brunaldi VO, Bernardo WM, de Moura DTH, de Moura ETH, Galvão M, et al. Obesity treatment with botulinum toxin-a is not effective: A systematic review and meta-analysis. Obesity Surgery. 2017;27(10):2716-2723. DOI: 10.1007/s11695-017-2857-5
  57. 57. de Moura EGH, Ribeiro IB, Frazão MSV, Mestieri LHM, de Moura DTH, Dal Bó CMR, et al. EUS-guided intragastric injection of botulinum toxin a in the preoperative treatment of super-obese patients: A randomized clinical trial. Obesity Surgery. 2019;29(1):32-39. DOI: 10.1007/s11695-018-3470-y
  58. 58. Yen YA, Wang CC, Sung WW, Fang KC, Huang SM, Lin CC, et al. Intragastric injection of botulinum toxin A for weight loss: A systematic review and meta-analysis of randomized controlled trials. Journal of Gastroenterology and Hepatology. 2022;37(6):983-992. DOI: 10.1111/jgh.15847. Epub 2022 Apr 22

Written By

Anıl Ergin and Cihan Şahan

Submitted: 28 January 2024 Reviewed: 01 February 2024 Published: 07 March 2024