Bariatric Surgery
Page 1 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
UnitedHealthcare
®
Commercial and Individual Exchange
Medical
Policy
Bariatric Surgery
Policy Number: 2024T0362MM
Effective Date: March 1, 2024
Instructions for Use
Table of Contents Page
Application ............................................................................. 1
Coverage Rationale .............................................................. 1
Documentation Requirements ............................................... 3
Definitions .............................................................................. 3
Applicable Codes .................................................................. 4
Description of Services ......................................................... 6
Benefit Considerations .......................................................... 8
Clinical Evidence ................................................................... 9
U.S. Food and Drug Administration .................................... 44
References .......................................................................... 45
Policy History/Revision Information .................................... 56
Instructions for Use ............................................................. 58
Application
UnitedHealthcare Commercial
This Medical Policy applies to all UnitedHealthcare Commercial benefit plans.
UnitedHealthcare Individual Exchange
This Medical Policy applies to Individual Exchange benefit plans in all states except for Alabama, Colorado, Florida,
Georgia, Kansas, Louisiana, Mississippi, Missouri, Ohio, Oklahoma, South Carolina, Tennessee, Texas, Virginia,
Washington, and Wisconsin.
Coverage Rationale
See Benefit Considerations
The following bariatric surgical procedures are proven and medically necessary for treating obesity:
Biliopancreatic diversion/biliopancreatic diversion with duodenal switch
Gastric bypass (includes robotic-assisted gastric bypass)
Adjustable gastric banding (using open or laparoscopic approaches) for individuals > 18 years of age. Refer to the
U.S. Food and Drug Administration (FDA) section for additional information
Sleeve Gastrectomy (Vertical Sleeve Gastrectomy)
In adults age 18 years or older, bariatric surgery using one of the procedures identified above
for treating obesity
is proven and medically necessary when all of the following criteria are met:
One of the following
o BMI 40 kg/m
2
(or BMI ≥ 37.5 kg/m
2
in individuals of Asian descent); or
o BMI 35 kg/m
2
39.9 kg/m
2
(or BMI ≥ 32.5 kg/m
2
37.4 kg/m
2
in individuals of Asian descent) in the presence of
one or more of the following co-morbidities:
Insulin resistance or Type 2 diabetes; or
Cardiovascular disease [e.g., history of stroke and/or myocardial infarction, poorly controlled hypertension
(systolic blood pressure greater than 140 mm Hg or diastolic blood pressure 90 mm Hg or greater, despite
pharmacotherapy), coronary artery disease, hyperlipidemia]; or
Related Commercial/Individual Exchange Policies
Minimally Invasive Procedures for Gastric and
Esophageal Diseases
Obstructive and Central Sleep Apnea Treatment
Robotic-Assisted Surgery Policy, Professional
Community Plan Policy
Bariatric Surgery
Bariatric Surgery
Page 2 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
History of cardiomyopathy; or
Obstructive Sleep Apnea (OSA)
confirmed on polysomnography with an AHI or RDI of > 30; or
Evidence of Nonalcoholic Fatty Liver Disease (NAFLD)
Idiopathic intracranial hypertension (pseudotumor cerebri)
and
The individual must also meet the following criteria:
o Both of the following:
Completion of a preoperative evaluation that includes a detailed weight history along with dietary and physical
activity patterns; and
Psychosocial-behavioral evaluation by an individual who is professionally recognized as part of a behavioral
health discipline to provide screening and identification of risk factors or potential postoperative challenges
that may contribute to a poor postoperative outcome
or
o Participation in a Multidisciplinary
surgical preparatory regimen
In adolescents age 12-17 years, the bariatric surgical procedures identified above
are proven and medically
necessary for treating obesity when all of the following criteria are met:
One of the following:
o Class III obesity; or
o Class II obesity in the presence of one or more of the following co-morbidities:
Insulin resistance or Type 2 diabetes; or
Poorly controlled hypertension (systolic blood pressure greater than 140 mm Hg or diastolic blood pressure
90 mm Hg or greater, despite pharmacotherapy)]; or
Hyperlipidemia; or
Obstructive Sleep Apnea confirmed on polysomnography with an AHI or RDI of > 30; or
Evidence of Nonalcoholic Fatty Liver Disease (NAFLD), or
Idiopathic intracranial hypertension (pseudotumor cerebri)
and
The individual must also receive an evaluation at, or in consultation with, a Multidisciplinary center focused on the
surgical treatment of severe childhood obesity. This may include Adolescent centers that have received accreditation
by the Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP) or can
demonstrate similar programmatic components
A planned two-stage procedure is proven and medically necessary when all of the following criteria are met:
Initial BMI 50 kg/m
2
prior to first stage bariatric procedure; and
Second stage occurs within 2 years following the primary bariatric surgery procedure; and
Individual has been compliant with nutrition and exercise; and
Individual meets medical necessity criteria listed above at time of second stage procedure
Revisional Bariatric Surgery using one of the procedures identified above
is proven and medically necessary
when due to a technical failure or major complication from the initial procedure; potential failure/complications
include but are not limited to the following:
Bowel perforation (including adjustable gastric band erosion)
Adjustable gastric band migration (slippage) that cannot be corrected with manipulation or adjustment (records must
demonstrate that manipulation or adjustment to correct band slippage has been attempted)
Leak
Obstruction (confirmed by imaging studies)
Staple-line failure
Mechanical adjustable gastric band failure
Uncontrollable reflux related to sleeve gastrectomy when all the following criteria are met:
o Maximum nonpharmacological medical management failure (e.g., positional, dietary modification and behavioral
changes); and
o Maximum pharmacological medical management failure (e.g., at least one month of double dose PPI, H2 blocker,
and/or sucralfate); and
o Severe esophagitis (grade C or D
) confirmed by endoscopy despite maximum medical management
Removal of adjustable gastric band and all related components which does not result in a revisional surgery is
proven and medically necessary.
Bariatric Surgery
Page 3 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
The following procedures are unproven and not medically necessary for treating obesity due to insufficient
evidence of efficacy:
Revisional Bariatric Surgery for any other indication than those listed above
Bariatric surgery as the primary treatment for any condition other than obesity
Bariatric interventions for the treatment of obesity including but not limited to:
o Bariatric artery embolization (BAE)
o Gastric electrical stimulation with an implantable gastric stimulator (IGS)
o Intragastric balloon
o Laparoscopic greater curvature plication, also known as total gastric vertical plication
o Mini-gastric bypass (MGB)/laparoscopic mini-gastric bypass (LMGBP)/One-Anastomosis Gastric Bypass (OAGB)
o Single-anastomosis duodenal switch [also known as duodenal switch with single anastomosis, or stomach
intestinal pylorus sparing surgery (SIPS)]
o Stomach aspiration therapy
o Transoral endoscopic surgery [includes TransPyloric Shuttle
®
(TPS
®
) Device, endoscopic sleeve gastroplasty]
o Vagus nerve blocking (VBLOC
®
)
o Gastrointestinal liners
Documentation Requirements
Benefit coverage for health services is determined by the member specific benefit plan document and applicable laws that
may require coverage for a specific service. The documentation requirements outlined below are used to assess whether
the member meets the clinical criteria for coverage but do not guarantee coverage of the service requested.
CPT Codes*
Bariatric Surgery
43644, 43645,
43647, 43648,
43659, 43770,
43771, 43772,
43773, 43774,
43775, 43843,
43845, 43846,
43847, 43848,
43860, 43865,
43881, 43882,
43886, 43887,
43888, 64590,
64595.
Medical notes documenting the following, when applicable:
Member height
Member weight
Preoperative evaluation that includes a detailed weight and Body Mass Index (BMI) history
along with dietary and physical activity patterns
Provider attestation of Asian ancestry, when applicable, if individual is of Asian descent
Co-morbidities
Treatments tried, failed, or contraindicated; include the dates and reason for discontinuation
(e.g., medications, diet, exercise, etc.)
Psychosocial-behavioral evaluation by a licensed behavioral health professional
Nutritional consult
Name of the facility where the procedure will be performed
In addition to the above, for staged bariatric surgery for BMI > 50, please include plan
For subsequent bariatric surgery, also provide the following, when applicable:
o Previous unsuccessful medical treatment
o Initial bariatric surgery performed and date and subsequent results or complications that
*For code descriptions, refer to the Applicable Codes section.
Definitions
Asian: Refers to a person having origins from the Far East, Southeast Asia, or the Indian subcontinent (e.g., Cambodia,
China, India, Japan, Korea, Malaysia, Pakistan, the Philippine Islands, Thailand, and Vietnam) (United States Census
Bureau, 2012).
Body Mass Index (BMI): A person's weight in kilograms divided by the square of height in meters. BMI can be used as a
screening tool but is not diagnostic of the body fatness or health of an individual (Centers for Disease Control and
Prevention [CDC], 2017).
The National Heart, Lung, and Blood Institute’s (NHLBI) Practical Guide Identification, Evaluation and Treatment of
Overweight and Obesity in Adults classifies the ranges of BMI in adults as follows:
< 18.5 - Underweight
Bariatric Surgery
Page 4 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
18.5 to 24.9 kg/m
2
Normal Weight
25-29.9 kg/m
2
Overweight
30-34.9 kg/m
2
Obesity Class I
35-39.9 kg/m
2
Obesity Class II
> 40 kg/m
2
Obesity Class III
The American Society of Metabolic and Bariatric Surgeons (ASMBS; Pratt et al., 2018), classifies severe obesity in
adolescents as follows:
Class II obesity 120% of the 95
th
percentile height, or an absolute BMI of 35-39.9 kg/m
2
, whichever is lower*
Class III obesity 140% of the 95
th
percentile height, or an absolute BMI of > 40 kg/m
2
, whichever is lower
*Also as defined by the American Heart Association (Kelly et al., 2013).
Los Angeles (LA) Classification of Oesophagitis:
Grade A: One (or more) mucosal break no longer than 5 mm that does not extend between the tops of two mucosal
folds
Grade B: One (or more) mucosal break more than 5 mm long that does not extend between the tops of two mucosal
folds
Grade C: One (or more) mucosal break that is continuous between the tops of two or more mucosal folds but which
involve less than 75% of the circumference
Grade D: One (or more) mucosal break which involves at least 75% of the esophageal circumference
(Lundell, et al. 1999)
Multidisciplinary: Bariatric center or regimen combining or involving several academic disciplines or professional
specializations in an approach to create a well-trained, safe, and effective environment for the complex bariatric patient.
Building the Multidisciplinary team includes staff such as the bariatric surgeon, obesity medicine specialist, registered
dietician, specialized nursing, behavioral health specialist, exercise specialist and support groups (American Society for
Metabolic and Bariatric Surgery (ASMBS) textbook of bariatric surgery).
Nonalcoholic Fatty Liver Disease (NAFLD): Condition that is evidenced by hepatic steatosis (HS) diagnosed either by
imaging or histology without a secondary cause of hepatic fat accumulation such as significant alcohol consumption, long-
term use of steatogenic medication, or monogenic hereditary disorders. (Chalasani et al., 2018)
Obstructive Sleep Apnea (OSA): The American Academy of Sleep Medicine (AASM) defines OSA as a sleep related
breathing disorder that involves a decrease or complete halt in airflow despite an ongoing effort to breathe. OSA severity
is defined as:
Mild for AHI or RDI ≥ 5 and < 15
Moderate for AHI or RDI ≥ 15 and ≤ 30
Severe for AHI or RDI > 30/hr
For additional information, refer to the Medical Policy titled Obstructive and Central Sleep Apnea Treatment.
Revisional Bariatric Surgery:
Conversion A second bariatric procedure that changes the bariatric approach from one procedure to a different type
of procedure (e.g., sleeve gastrectomy or adjustable gastric band converted to Roux-en-Y [RYGB]). Note: This is not
the same as an intraoperative conversion (e.g., converting from laparoscopic approach to an open procedure).
Corrective A procedure that corrects or modifies anatomy of a previous bariatric procedure to achieve the original
desired outcome or correct a complication. These procedures also address device manipulation (e.g., gastric pouch
resizing, re-sleeve gastrectomy, limb length adjustments in RYGB and gastric band replacement).
Reversal A procedure that restores original anatomy. (Mirkin, et al. 2021)
Applicable Codes
The following list(s) of procedure and/or diagnosis codes is provided for reference purposes only and may not be all
inclusive. Listing of a code in this policy does not imply that the service described by the code is a covered or non-covered
health service. Benefit coverage for health services is determined by the member specific benefit plan document and
applicable laws that may require coverage for a specific service. The inclusion of a code does not imply any right to
reimbursement or guarantee claim payment. Other Policies and Guidelines may apply.
Coding Clarification: Utilize CPT code 43775 to report laparoscopic sleeve gastrectomy rather than the unlisted CPT
code 43659.
Bariatric Surgery
Page 5 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
CPT Code
Description
0813T
Esophagogastroduodenoscopy, flexible, transoral, with volume adjustment of intragastric bariatric
balloon
43290
Esophagogastroduodenoscopy, flexible, transoral; with deployment of intragastric bariatric balloon
43291
Esophagogastroduodenoscopy, flexible, transoral; with removal of intragastric bariatric balloon(s)
43644
Laparoscopy, surgical, gastric restrictive procedure; with gastric bypass and Roux-en-Y
gastroenterostomy (roux limb 150 cm or less)
43645
Laparoscopy, surgical, gastric restrictive procedure; with gastric bypass and small intestine
reconstruction to limit absorption
43647
Laparoscopy, surgical; implantation or replacement of gastric neurostimulator electrodes, antrum
43648
Laparoscopy, surgical; revision or removal of gastric neurostimulator electrodes, antrum
43659
Unlisted laparoscopy procedure, stomach
43770
Laparoscopy, surgical, gastric restrictive procedure; placement of adjustable gastric restrictive
device (e.g., gastric band and subcutaneous port components)
43771
Laparoscopy, surgical, gastric restrictive procedure; revision of adjustable gastric restrictive device
component only
43772
Laparoscopy, surgical, gastric restrictive procedure; removal of adjustable gastric restrictive device
component only
43773
Laparoscopy, surgical, gastric restrictive procedure; removal and replacement of adjustable gastric
restrictive device component only
43774
Laparoscopy, surgical, gastric restrictive procedure; removal of adjustable gastric restrictive device
and subcutaneous port components
43775
Laparoscopy, surgical, gastric restrictive procedure; longitudinal gastrectomy (i.e., sleeve
gastrectomy)
43843
Gastric restrictive procedure, without gastric bypass, for morbid obesity; other than vertical-banded
gastroplasty
43845
Gastric restrictive procedure with partial gastrectomy, pylorus-preserving duodenoileostomy and
ileoileostomy (50 to 100 cm common channel) to limit absorption (biliopancreatic diversion with
duodenal switch)
43846
Gastric restrictive procedure, with gastric bypass for morbid obesity; with short limb (150 cm or less)
Roux-en-Y gastroenterostomy
43847
Gastric restrictive procedure, with gastric bypass for morbid obesity; with small intestine
reconstruction to limit absorption
43848
Revision, open, of gastric restrictive procedure for morbid obesity, other than adjustable gastric
restrictive device (separate procedure)
43860
Revision of gastrojejunal anastomosis (gastrojejunostomy) with reconstruction, with or without
partial gastrectomy or intestine resection; without vagotomy
43865
Revision of gastrojejunal anastomosis (gastrojejunostomy) with reconstruction, with or without
partial gastrectomy or intestine resection; with vagotomy
43881
Implantation or replacement of gastric neurostimulator electrodes, antrum, open
43882
Revision or removal of gastric neurostimulator electrodes, antrum, open
43886
Gastric restrictive procedure, open; revision of subcutaneous port component only
43887
Gastric restrictive procedure, open; removal of subcutaneous port component only
43888
Gastric restrictive procedure, open; removal and replacement of subcutaneous port component only
64590
Insertion or replacement of peripheral, sacral, or gastric neurostimulator pulse generator or receiver,
requiring pocket creation and connection between electrode array and pulse generator or receiver
64595
Revision or removal of peripheral, sacral, or gastric neurostimulator pulse generator or receiver,
with detachable connection to electrode array
64999
Unlisted procedure, nervous system
CPT
®
is a registered trademark of the American Medical Association
Bariatric Surgery
Page 6 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Description of Services
Obesity
Obesity is defined clinically using the Body Mass Index (BMI). Obesity is a significant health concern due to its high
prevalence and associated health risks.
Health consequences associated with obesity include hypertension, Type II diabetes, hyperlipidemia, atherosclerosis,
heart disease, stroke, diseases of the gallbladder, liver disease, osteoarthritis, Obstructive Sleep Apnea, and other
respiratory problems. In addition, certain cancers are more prevalent in obese individuals, including endometrial, ovarian,
breast, prostate, colon cancer, renal cell carcinoma, and non-Hodgkin's lymphoma.
The U.S. Preventive Services Task Force (USPSTF) recommends screening all adults for obesity. Clinicians should offer
or refer patients with a BMI of 30 kg/m2 or higher to intensive, multicomponent behavioral interventions (USPSTF, 2012).
Bariatric Surgery in the Adolescent Population
For Adolescents, physical development and maturation may be determined utilizing the gender specific growth chart and
BMI chart developed by the CDC, National Center for Health Statistics.
First-Line Treatments for Obesity
First-line treatments for obesity include dietary therapy, physical activity, behavior modification, and medication
management; all of which have often been unsuccessful in long-term weight management for obese individuals (Lannoo
and Dillemans, 2014).
Bariatric Surgical Procedures
The goal of surgical treatment for obesity is to induce significant weight loss and, thereby, reduce the incidence or
progression of obesity-related comorbidities, as well as to improve quality of life. The purpose of performing bariatric
surgery in Adolescent patients is to reduce the lifelong impact of severe obesity.
Surgical treatment of obesity offers two main weight-loss approaches: restrictive and malabsorptive. Restrictive methods
are intended to cause weight loss by restricting the amount of food that can be consumed by reducing the size of the
stomach. Malabsorptive methods are intended to cause weight loss by limiting the amount of food that is absorbed from
the intestines into the body. A procedure can have restrictive features, malabsorptive features, or both. The surgical
approach can be open or laparoscopic. The clinical decision on which surgical procedure to use is made based on a
medical assessment of the patient's unique situation.
Roux-en-y Bypass (RYGB)/Gastric Bypass
The RYGB procedure involves creating a stomach pouch out of a small portion of the stomach and attaching it directly to
the small intestine, bypassing a large part of the stomach and duodenum.
Laparoscopic Adjustable Gastric Banding (LAGB)
The laparoscopic adjustable gastric banding procedure involves placing an inflatable silicone band around the upper
portion of the stomach. The silicone band contains a saline reservoir that can be filled or emptied under fluoroscopic
guidance to change the caliber of the gastric opening.
Vertical Sleeve Gastrectomy (VSG)
VSG can be performed as part of a two-staged approach to surgical weight loss or as a stand-alone procedure. A VSG
involves the removal of 60-75% of the stomach, leaving a narrow gastric “tube” or “sleeve.” This small remaining “tube”
cannot hold as much food and produces less of the appetite-regulating hormone ghrelin, lessening a patient’s desire to
eat. VSG is not a purely malabsorptive procedure, so there is no requirement for lifetime nutritional supplementation
(California Technology Assessment Forum, 2015).
Biliopancreatic Diversion with Duodenal Switch (BPD/DS) (also known as the
Scopinaro Procedure)
BPD is primarily malabsorptive but has a temporary restrictive component. As in RYGB, three "limbs" of intestine are
created: one through which food passes, one that permits emptying of fluids (e.g., bile) from digestive organs, and a
common limb through which both food and digestive fluids pass. This procedure involves removal of the greater curvature
Bariatric Surgery
Page 7 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
of the stomach instead of the distal portion. The two limbs meet in a common channel measuring only 50 to 100 cm,
thereby permitting relatively little absorption.
Robotic-Assisted Surgery
Robotic surgery provides surgeons with three-dimensional vision, increased dexterity and precision by downscaling
surgeon's movements enabling a fine tissue dissection and filtering out physiological tremor. It overcomes the restraint of
torque on ports from thick abdominal wall and minimizes port site trauma by remote center technology (Bindal et al.,
2015).
Transoral Endoscopic Surgery
Transoral endoscopic surgery is an option being explored for bariatric surgery. Natural orifice transluminal endoscopic
surgery (NOTES) is performed via a natural orifice (e.g., mouth, vagina, etc.), and in some cases eliminates the need for
abdominal incisions. This form of surgery is being investigated as an alternative to conventional surgery.
Transoral restorative obesity surgery (ROSE) is another endoscopic procedure. The endoscope with four channels is
inserted into the esophagus and then the stomach. Specialized instruments are placed through the channels to create
multiple folds around the existing stoma to reduce the diameter.
The Transpyloric Shuttle
®
(TPS
®
) device is a non-balloon, space occupying device with a 12-month treatment duration
that is proposed as a new endoscopic bariatric therapy. The TPS device is comprised of a spherical silicone bulb
connected to a smaller cylindrical silicone bulb by a flexible tether; it is delivered to and removed from the stomach using
transluminal endoscopic procedures in the outpatient setting (Marinos, 2014). The device was granted FDA premarket
approval on April 16, 2019, and was approved for up to 12 months weight loss therapy in patients with a BMI of 35.0
kg/m2 to 40.0 kg/m2 or a BMI of 30.0 kg/m2 to 34.9 kg/m2 with 1 or more obesity-related comorbid condition. The device
is intended to be used in conjunction with a diet and behavior modification program (ECRI, 2019).
Endoscopic Sleeve Gastroplasty (ESG) is a minimally invasive technique through the mouth that uses an endoscopic
suturing device (e.g., OverStitch) to reduce gastric capacity by sealing off most of the stomach, forcing ingested food
through an open tube of stomach tissue that connects the esophagus to the small intestine. ESG is similar to a
laparoscopic sleeve gastrectomy in which the stomach is manipulated to create a tube-shape, however no stomach tissue
is removed.
Laparoscopic Mini Gastric Bypass (LMGBP)/One-Anastomosis Gastric Bypass
(OAGB)
LMGBP/OAGB involves the construction of a gastric tube by dividing the stomach vertically, down to the antrum. As in the
RYGB, food does not enter the distal stomach. However, unlike gastric bypass surgery, digestive enzymes and bile are
not diverted away from the stomach after LMGBP/OAGB. This can lead to bile reflux gastritis which can cause pain that is
difficult to treat.
Implantable Gastric Stimulator (IGS)
IGS is a small, battery-powered device similar to a cardiac pacemaker, in a small pocket, created beneath the skin of the
abdomen using laparoscopy. The IGS is programmed externally using a controller that sends radiofrequency signals to
the device. Although the exact mechanism of action is not yet understood, gastric stimulation is thought to target ghrelin,
an appetite-related peptide hormone (Gallas and Fetissov, 2011).
Vagus Nerve Blocking Neurostimulation Therapy (VBLOC)
VBLOC uses an implanted subcutaneous neurostimulator to deliver electrical pulses to the vagus nerve, which may
suppress appetite (ECRI, 2016).
VBLOC therapy () is designed to target the multiple digestive functions under control of the vagus nerves and to affect the
perception of hunger and fullness.
Intragastric Balloon (IGB)
IGBs are acid-resistant balloons that are inserted into the stomach and expanded with saline or air. These space-
occupying devices promote weight loss by creating a feeling of fullness, which can lead to reduced consumption of food.
The devices are intended as an adjunct to diet, exercise, and behavioral counseling for the treatment of obesity (Hayes,
2021). Available clinical data and manufacturer recommendations indicate 6 months to be the current standard duration of
therapy from insertion to removal (ASMBS, 2016).
Bariatric Surgery
Page 8 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Laparoscopic Greater Curvature Plication (LGCP) [also known as Total Gastric
Vertical Plication (TGVP)]
LGCP is a restrictive procedure that involves folding and suturing the stomach onto itself to decrease the size of the
stomach and requires no resection, bypass, or implantable device. This procedure is a modification of the gastric sleeve
which requires surgical resection of stomach.
Stomach Aspiration Therapy
Stomach aspiration therapy, such as with the AspireAssist
®
, uses a surgically placed tube (endoluminal device) designed
to aspirate a portion of the stomach contents after every meal (Hayes, 2021). The AspireAssist
®
is intended for long-term
use in conjunction with lifestyle therapy (to help patients develop healthier eating habits and reduce caloric intake) and
continuous medical monitoring. Patients must be monitored regularly for weight loss progress, stoma site heath, and
metabolic and electrolyte balance.
Bariatric Artery Embolization (BAE)
BAE is a minimally invasive procedure which is the percutaneous, catheter-directed, trans-arterial embolization of the left
gastric artery (LGA). The procedure is performed by an interventional radiologist and targets the fundus that produces the
majority of the hunger-controlling hormone ghrelin. Beads placed inside the vessels purportedly help decrease blood flow
and limit the secretion of ghrelin to minimize feelings of hunger to initiate weight loss.
Gastrointestinal Liners
Gastrointestinal liners, such as the EndoBarrier
system, utilize an endoscopically implanted sleeve into the stomach to
reduce the stomach size. The sleeve is then removed after weight loss has been achieved. The EndoBarrier is not
approved for use by the U.S. Food and Drug Administration (FDA) in the United States; it is limited by federal law to
investigational use only.
Single-Anastomosis Duodenal Switch (SADS)
SADS is also called single-anastomosis loop duodenal switch, single-anastomosis duodenoileal bypass with sleeve
gastrectomy, or stomach intestinal pylorus-sparing surgeryis a modification of biliopancreatic diversion with duodenal
switch (BPD-DS). SADS consists of a sleeve gastrectomy to remove most of the stomach and an intestinal bypass to
shorten the length of the small intestine and to allow bile and pancreatic digestive juices to mix with the food. SADS is
typically performed laparoscopically as an inpatient procedure.
Revisional Surgery
The indications for Revisional Bariatric Surgery vary greatly depending on the index procedure performed and the nature
of the complication. Some complications may be encountered during the acute postoperative recovery period (leaks,
abscesses, fistulae, etc.). Prior to revisional surgery, patients should undergo a thorough Multidisciplinary assessment
and consideration of their individual risks and benefits from revisional surgery (Brethauer et al., 2014). It is important to
determine if the poor response to primary bariatric surgery is due to anatomic causes that led to inadequate weight loss or
weight regain or to the patient’s postoperative behavior, such as not following the prescribed diet and lifestyle changes
(e.g., consuming large portions, high-calorie foods, and/or snacks between meals; not exercising). Uncontrollable reflux
may be a complication experienced by some patients; first-line therapy for patients who experience GERD after bariatric
surgery includes dietary and lifestyle modification, alcohol, and smoking cessation, followed by acid-reducing medications
(King et al. 2021).
The Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP) is a national
accreditation standard for bariatric surgery centers. In 2012, the American College of Surgeons (ACS) and the American
Society for Metabolic and Bariatric Surgery (ASMBS) combined their individual accreditation programs into a single unified
program. MBSAQIP works to advance safe, high-quality care for bariatric surgical patients through the accreditation of
bariatric surgical centers. A bariatric surgical center achieves accreditation following a rigorous review process during
which it proves that it can maintain certain physical resources, human resources, and standards of practice. All accredited
centers report their outcomes to the MBSAQIP database (MBSAQIP, 2019).
Benefit Considerations
Most Certificates of Coverage and many Summary Plan Descriptions explicitly exclude benefit coverage for bariatric
surgery.
Bariatric Surgery
Page 9 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Some states may require coverage for bariatric surgery. Refer to the member specific benefit plan document to determine
availability of benefits for these procedures. As in all benefit adjudication, state legislated mandates must be followed.
Therefore, the applicable state-specific requirements and the member specific benefit plan document must be reviewed to
determine what benefits, if any, exist for bariatric surgery.
For Fully Insured Group Policies in Maryland Only
Use the following criteria as specified in the Code of Maryland Regulations COMAR 31.10.33.03B (Accessed on
November 27, 2023):
A Body Mass Index (BMI) above 40 kg/m2 without co-morbidity; or
A BMI of 35 kg/m2 or greater with obesity-related co-morbid medical conditions including:
o Hypertension
o Cardiopulmonary condition
o Sleep apnea
o Diabetes
o Any life threatening or serious medical condition that is weight induced
Age 18 years or older
Completion of a structured diet program, such as Weight Watchers or Jenny Craig. Either of the following in the two-
year period that immediately precedes the request for the surgical treatment of morbid obesity meets the indication:
o One structured diet program for six consecutive months; or
o Two structured diet programs for three consecutive months
A carrier or a private review agent acting on behalf of a carrier shall use flexibility with regard to defining a structured
diet program
Completion of a psychological examination of the member's readiness and fitness for surgery and the necessary
postoperative lifestyle changes
Use the following criteria as specified in the Code of Maryland Regulations COMAR 31.10.33.04
(Accessed on November
27, 2023):
Documentation of completion of a structured diet program should include:
o Physician Notes
o Notes of health care providers, other than physicians
o Receipts of payment for a structured diet program; or
o Diet or weight loss logs from a structured diet program
Clinical Evidence
The criteria for patient selection for bariatric surgery are relatively uniform among clinical studies published in the peer-
reviewed literature and broadly correspond to criteria recommended by the American Association of Clinical
Endocrinologists (AACE), the Obesity Society, and American Society for Metabolic & Bariatric Surgery (ASMB)
(Mechanick et al., 2019):
Patients with a BMI 40 kg/m2 (Obesity Class III) with or without coexisting medical problems and for whom bariatric
surgery would not be associated with excessive risk.
Patients with a BMI 35 kg/m2 (Obesity Class II) and one or more severe obesity-related co-morbidities.
Demonstration that a multidisciplinary approach with dietary, other lifestyle modifications (such as exercise and
behavioral modification), and pharmacological therapy, if appropriate, have been unsuccessful.
Refer to the Clinical Practice Guidelines
section below for additional information.
Kapeluto et al. (2020) assessed long-term glycemic outcomes in 132 patients with type 2 diabetes (T2D) that received
Biliopancreatic Diversion with Duodenal Switch (BPD/DS) surgery versus other bariatric surgeries. Inclusion criteria
consisted of patients with diagnosis of T2D and those that had underwent BPD/DS surgical procedure. Patient follow up
consisted of post-surgical assessments at week 3 and then at 4, 8, 12, 18, and 24 months and annually thereafter. Fifteen
patients were lost to death during the 10 years follow-up and two more beyond 10 years. 90% of the patients had clinical
remission of their diabetes; 3 patients had partial remission, 21 had improvement and 3 were unchanged in their status.
The authors found that BPD-DS maintained a remission rate of 10 years postop in the vast majority of patients with
advanced diabetes. The authors concluded patients that underwent BPD-DS had positive results for long-term benefits for
remission of T2D and that earlier referral for this type of surgery should be made. Limitations included late arrival of the
standard use of the HbA1C test, incomplete weight parameters due to lack of self-reported weights and retrospective
analysis.
Bariatric Surgery
Page 10 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Khalaj et al. (2020) conducted a cohort study comparing gastric bypass (GB) to sleeve gastrectomy (SG) and the
effectiveness and safety of these two procedures. The authors evaluated 2,202 patients that underwent laparoscopic SG
and 1,085 patients who underwent laparoscopic GB. The SG procedure was performed over a 36-F bougie and reinforced
with an omental pouch; the GB procedure was performed as either RYGB or one anastomosis (OAGB). Evaluation of
weight loss included body mass index change, percent of total weight loss, and percentage of excess weight loss. Type 2
diabetes mellitus (T2DM), hypertension (HTN), and dyslipidemia, as obesity-associated comorbidities were assessed in
all patients. There were no major complications identified which was recognized by a return to the operating room,
prolonged hospital stays beyond 7 days, or the need for re-admission. Quality of life (QoL) was assessed using the
Iranian version of the Short-Form Health Survey which measured physical, social, and mental aspects of health. Patient
follow up for both types of procedures occurred at 6, 12, and 24 months after surgery. The authors found no significant
differences between the two surgical groups; patients that underwent SG had a lower FPG and HbA1C when compared to
the GB group. BMI was not significantly different between the two groups. Excess weight loss (EWL)% was 61.9 ±15.7,
74.8 ±19.1, and 75.0 ±21.9 in the SG group and 62.7 ±15.3, 77.5 ±18.4, and 80.1 ±20.8 in the GB group at 6-, 12-, and
24-month follow-ups, respectively. All patient comorbidities and QoL improved. The authors concluded that bariatric
surgery is effective and safe for treatment of obesity; while both procedures are effective for weight loss, remission of
obesity-associated comorbidities, and QoL, SG is associated with fewer complications and nutritional deficiencies.
Jung et al. (2020) conducted a systematic review and meta-analysis of 22 studies with 2,141 patients to comprehensively
evaluate the efficacy of different endoscopic bariatric procedures compared to lifestyle modification in the treatment of
morbid obesity. Intragastric balloon, duodenal-jejunal bypass liner (DJBL), aspiration therapy, primary obesity surgery
endoluminal (POSE) procedure, and botulinum toxin injection to the stomach were included and the meta-analysis
determined the percentage of weight loss (%weight loss) and percentage of excess weight loss (%EWL). The results
showed that the Obalon Balloon system was shown to have efficacy for both %weight loss and %EWL, its efficacy was
not proven due to the small number of studies and comparatively low effect size. Aspiration therapy demonstrated
effectiveness for weight reduction when compared to lifestyle modification. Gas-filled balloon and botulinum toxin injection
did not show a significant difference in %weight loss or %EWL compared with the control. The authors concluded that all
bariatric endoscopic procedures, with the exception of a gas filled balloon and botulinum toxin injection show superior
short-term efficacy compared with lifestyle modification. These findings are limited by lack of long-term efficacy and safety
quality data. (The following publications previously cited in this policy, are included in this systematic review: Abu Dayyeh
2015b, Chang 2014, Courcoulas 2017, Gersin 2010, Schouten 2010, Sullivan 2013, Thompson 2017).
O’Brien et al. (2019) performed a systematic review and meta-analysis on 33 reports containing ten or more years of
follow-up for patients that underwent bariatric surgery. The authors evaluated the long-term effectiveness of Roux-en-Y
gastric bypass (RYGB), laparoscopic adjustable gastric banding (LAGB), or BPD/DS. Results for gastric bypass surgery
showed a weighted mean % EWL of 56.7% at 10 or more years with a mean of 55.4% EWL. Eleven reports addressing
BPD/DS showed a mean of 74.1% EWL and two reports for sleeve gastrectomy showed a mean of 57.0% EWL. A
longitudinal cohort study for the patients receiving LAGB showed patient weight loss reached a peak at the 2-year follow-
up and remained relatively stable through the next 18 years with a mean weight loss of 24.8 kg representing 47.2 %EWL.
The authors concluded that RYGB, LAGB and BPD/DS lead to substantial weight loss which continued for at least 10
years. Due to patient education and lap band design changes, revisional surgery has decreased significantly over the past
eleven years. The findings are limited by lack of direct comparison between techniques and lack of comparison groups not
undergoing surgical treatments. (The following publications previously cited in this policy, are included in this systematic
review: Maciejewski 2016, Salminen 2018, Schauer 2017, Sethi 2016, Sheikh 2017, Topart 2017, Vinzens 2017).
Zhao and Jiao (2019) conducted a systematic review to determine whether LRYGB and LSG are equivalent for mid- and
long-term weight loss, resolution of comorbidities and adverse events (AEs). Eleven RCTs were included in the meta-
analysis and the authors found no significant difference in excess weight loss between LRYGB and LSG nor any
significant difference for T2D improvement. This analysis did identify more postoperative early complications for LRYGB,
but no difference between the two procedures in later postoperative period. Future studies should focus on the
comparison of complication and comorbidities. Limitations included the variation in sample size among the included
studies which may have created a bias, variation of patient age and preoperative BMIs which may have led to
heterogeneity, and failure of subgroup analysis for reoperation rate. Additional studies are needed to determine the
relative long-term efficacy of different bariatric surgeries. (Publication by Salminen 2018, which was previously cited in this
policy, is included in this systematic review).
Chaar et al. (2018) reported 30-day outcomes of SG versus RYGB based on the Metabolic and Bariatric Surgery
Accreditation and Quality Improvement Program database in a large retrospective cohort study. The authors’ evaluation
showed that the incidence of postoperative complications in the first 30 days after surgery is low for both RYGB and SG.
However, SG seems to have a better safety profile in the first 30 days postoperatively compared with RYGB. These
Bariatric Surgery
Page 11 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
findings should be considered in the preoperative evaluation and counseling of bariatric patients. Long-term follow-up is
needed to compare safety and efficacy of SG versus RYGB.
Jambhekar et al. (2018) evaluated demographic and socioeconomic factors in the United States that are predictors of
long-term weight loss after LSG in a cohort study. Prospectively collected data on 713 consecutive primary LSG
operations was included in this study. Multiple regression analyses were done to determine if gender, race, or
socioeconomic factors such as insurance and employment status correlated with postoperative weight loss. The presence
of chronic comorbidities affecting quality of life such as T2D and obstructive sleep apnea (OSA) were also recorded and
analyzed. All studied groups had similar preoperative body mass index (BMI) (mean 46 kg/m
2
). Race was not significantly
associated with weight loss at any postoperative interval. Male gender was associated with increased weight loss through
the first three months (48.2 +/- 12.5 lbs. vs. 40.5 +/- 11 lbs.; p = 0.0001). Patients with T2D had significantly less weight
loss at the 6 through 18-month intervals (50.4 +/- 17.9 lbs. vs. 59.6 +/- 15.6 lbs. at six months; p = 0.00032; 53.3 +/-
25.4lbs vs. 80.5 +/- 31.3lbs at 18 months; p = 0.008). Patients with OSA had significantly less weight loss at the two-year
interval (57.5 +/- 29.2 lbs.) vs. those without OSA (69.6 +/- 23.5 lbs.; p = 0.047). Finally, those patients who were students
had the greatest weight loss at two years postoperatively with the least weight loss seen in retired patients followed by
those on disability (108.0 +/-21.5 lbs. vs. 26.0 lbs. vs. 46.0 +/-19.7 lbs.; p = 0.04). Further studies are needed to evaluate
whether demographic differences impact long term weight loss. Limitations included loss to follow-up, identification, and
testing of only selected predictive factors, thus underrepresenting other socioeconomic factors, and conflicting results
were identified between the model variables.
Shoar and Saber (2017) conducted a systematic review and meta-analysis to compare long-term and midterm outcomes
of LSG versus laparoscopic RYGB (LRYGB). Fourteen studies comprising 5264 patients were eligible. Follow-up ranged
from 36 months to 75.8 ±8.4 months. The pooled result for weight loss outcomes did not show any significant difference in
midterm weight loss (standardized mean difference = -0.03; 95% confidence interval (CI), -0.38-.33; p = .88) but a
significant difference in the long-term weight loss outcome favoring LRYGB (standardized mean difference = .17; 95% CI,
.05-.28; p = .005). The pooled results demonstrated no significant difference for resolution of T2D, hypertension,
hyperlipidemia, and hypertriglyceridemia. Despite the insignificant difference between LRYGB and LSG in midterm weight
loss, LRYGB produced better weight loss in the long-term. There was no significant difference between the 2 procedures
for co-morbidity resolution. A major limitation of this study was the inclusion of short-term studies in the pooled analysis of
midterm studies but claimed to be a long-term meta-analysis.
Lager et al. (2017) retrospectively studied 30-day postoperative complications as well as changes in weight, blood
pressure, cholesterol, hemoglobin, hemoglobin A1C, and creatinine from baseline to 2, 6, 12, and 24 months
postoperatively in 383 patients undergoing RYGB and 336 patients undergoing SG. Follow-up rates were 706/719 at 2
months, 566/719 at 6 months, 519/719 at 12 months, and 382/719 at 24 months. Baseline characteristics were similar in
both groups except for higher weight and BMI in the SG group. The RYGB group experienced greater total body weight
loss at 6, 12, and 24 months (41.9 vs. 34.6 kg at 24 months, p < 0.0001). Excess weight loss was 69.7 and 51.7 %
following RYGB and SG respectively at 24 months (p < 0.0001). Blood pressure improved significantly in both groups.
Surgical complication rates were greater after RYGB (10.1 vs. 3.5 %, p = 0.0007) with no significant difference in life-
threatening or potentially life-threatening complications. Weight loss was greater following RYGB compared to SG at 2
years. The authors recommend that surgical intervention be tailored to surgical risk, comorbidities, and desired weight
loss. Limitations included retrospective design which may have impacted patient selection and other biases, incomplete
biochemical data as some patients did not return to clinic for routine blood draws and performed at specific institution.
Kang and Le (2017) conducted a systematic review and meta-analysis to determine the effectiveness of bariatric surgical
procedures. Eleven randomized controlled trials (RCTs) that met the criteria were included in the review. Of 9 trials (n = 
765), the differences in mean BMI reduction were -0.76 (95% CI: -3.1 to 1.6) for RYGB versus SG, -5.8 (95% CI: -9.2 to -
2.4) for RYGB versus LAGB, and -5.0 (95% CI: -9.0 to -1.0) for SG versus LAGB. Eight RCTs (n = 656) reported
percentage excess weight-loss (%EWL), the mean differences between RYGB and SG, RYGB and LAGB, and SG and
LAGB were 3.8% (95% CI: -8.5% to 13.8%), -22.2% (95% CI: -34.7% to -6.5%), and -26.0% (95% CI: -40.6% to -6.4%),
respectively. The meta-analysis indicated low heterogeneity between studies, and the node splitting analysis showed that
the studies were consistent between direct and indirect comparisons (p > .05). The authors concluded that the RYGB and
SG were similar in weight-loss effect, and both were superior to LAGB. Other factors such as complications and patient
preference should be considered during surgical consultations.
In a systematic analysis, Osland et al. (2017a) evaluated the postoperative impact on T2D resolution following
laparoscopic vertical sleeve gastrectomy (LVSG) and LRYGB. Seven RCTs involving a total of 732 patients (LVSG n =
365, LRYGB n = 367) met inclusion criteria. Significant diabetes resolution or improvement was reported with both
procedures across all time points. Similarly, measures of glycemic control (HbA1C and fasting blood glucose levels)
improved with both procedures, with earlier improvements noted in LRYGB that stabilized and did not differ from LVSG at
Bariatric Surgery
Page 12 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
12 months postoperatively. Early improvements in measures of insulin resistance in both procedures were also noted in
the studies that investigated this. The authors suggest that both procedures are effective in resolving or improving
preoperative T2Din obese patients during the reported 3-to -5-year follow-up periods. However, further studies are
required before longer-term outcomes can be elucidated. Areas identified that need to be addressed for future studies on
this topic include longer follow-up periods, standardized definitions, and time point for reporting.
Osland et al. (2017b) conducted a systematic review of non-diabetic comorbid disease status following LRYGB and
LVSG. Six RCTs involving a total of 695 patients (LVSG n = 347, LRYGB n = 348) reported on the resolution or
improvement of comorbid disease following LVSG and LRYGB procedures. The authors concluded that this systematic
review of RCTs suggests that both LVSG and LRYGB are effective in resolving or improving preoperative nondiabetic
comorbid diseases in obese patients. While results are not conclusive, in the authors’ opinion, LRYGB may provide
superior results compared to LVSG in mediating the remission and/or improvement in some conditions such as
dyslipidemia and arthritis.
Polega et al. (2017) conducted a matched cohort study of laparoscopic BPD/DS and SG to compare 30-day outcomes. Of
the 741 patients who underwent BPD/DS or SG, 2 cohorts of 167 patients each were matched for age, sex, and BMI.
Length of stay (LOS) was longer in the BPD/DS cohort (2.5 ±.9 days versus 2.1±.7 days, p < .001). There were no
significant differences between the groups in relation to 30-day postoperative rates of leak (0.3% versus 0.6%, p > 0.99),
bleed (0% versus 0.3%, p > 0.99), reoperation (1.2% versus .6%, p > .99), or readmission (3% versus 1.2%, p = .45).
There were no mortalities. After matching for age, sex, and BMI, the authors found no significant differences between
BPD/DS and SG with regard to 30-day postoperative rates of leak, bleed, reoperation, readmission, or mortality.
Risstad et al. (2017) conducted a randomized clinical trial with 60 patients with body mass index 50-60 kg/m2 to
investigate bile acid profiles up to 5 years after RYGB and BPD/DS. Total bile acid concentrations increased substantially
over 5 years after both RYGB and BPD/DS, with greater increases in total and primary bile acids after BPD/DS. Higher
levels of total bile acids at 5 years were associated with lower body mass index, greater weight loss, and lower total
cholesterol.
In a systematic review and meta-analysis, Osland et al. (2016) evaluated the early postoperative complication rate (i.e.,
within 30-days) in 6 RCTs involving a total of 695 patients (LVSG n = 347, LRYGB n = 348). A statistically significant
reduction in relative odds of early major complications favoring the LVSG procedure was noted (p = 0.05). Five RCTs
representing 633 patients (LVSG n = 317, LRYGB n = 316) reported early minor complications. A non-statically significant
reduction in relative odds of 29 % favoring the LVSG procedure was observed for early minor complications (p = 0.4).
However, other outcomes directly related to complications which included reoperation rates, readmission rate, and 30-day
mortality rate showed comparable effect size for both surgical procedures. The authors concluded that this meta-analysis
and systematic review of RCTs suggests that fewer early major and minor complications are associated with LVSG
compared with LRYGB procedure. However, this does not translate into higher readmission rate, reoperation rate, or 30-
day mortality for either procedure.
Xie et al. (2016) prospectively evaluated Apnea-Hypopnea Index (AHI) and Functional Outcomes of Sleep Questionnaires
Scores (FOSQ) pre- and post-operatively in patients undergoing bariatric surgery. A total of 167 subjects were studied.
The median age was 46 (14-75) years and BMI 49 (36-69) kg/m2. Ninety-two (55.0%) patients were diagnosed with OSA
preoperatively. Fifty (54.0%) required positive airway pressure (PAP) therapy. The mean reduction in BMI post bariatric
surgery was 12.2 ± 4.52 kg/m2 at 6.56± 2.70 months. Eighty (87.9%) reported improved sleep quality reflected in
improved scores in all domains of the FOSQ (p < 0.001, paired t-test). Improvement in FOSQ scores remained significant
(p < 0.05) in those with and without OSA. Thirty-nine (90.7%) patients discontinued PAP due to resolution of daytime
sleepiness. In conclusion, the authors identified that weight loss following bariatric surgery has a positive impact on sleep
in patients with and without OSAS. The findings are however limited by lack of comparison group without bariatric surgery.
Giordano (2015) conducted retrospective comparative study of consecutive super-obese patients. Patients either
underwent RYGB (n = 102) or LAGB (n = 79). Early complications and weight loss outcomes were comparable between
the two groups in the short term. However, weight loss and excess weight loss percent at 6 and 12 months of follow-up
was significantly higher in patients who underwent RYGB than LAGB.
Arterburn et al. (2015) evaluated the association between bariatric surgery and long-term survival in a retrospective cohort
study of obese patients treated at the Veterans Administration (VA) health system. A cohort of surgical patients (n = 2500;
mean age, 52 years; mean body mass index [BMI] of 47), undergoing any bariatric surgery procedure, were compared
with control patients (n = 7462). At the end of 14 years, there were a total of 263 deaths in the surgical cohort group (n =
2500) and 1277 deaths in the matched controls (n = 7462). Based on Kaplan-Meier estimates, mortality rates were 2.4%
at 1 year, 6.4% at 5 years, and 13.8% at 10 years for surgical cohort patients. In the matched controls, mortality rates
Bariatric Surgery
Page 13 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
were 1.7% at 1 year, 10.4% at 5 years, and 23.9% at 10 years. Bariatric surgery was associated with reduced mortality
compared controls after 1 to 5 years (hazard ratio [HR], 0.45; 95% CI, 0.36 to 0.56) and after 5 years (HR, 0.47; 95%CI,
0.39 to 0.58). Across different subgroups based on diabetes diagnosis, sex, and period of surgery, there were no
significant differences between surgery and survival at the mid- and long-term evaluations. Limitations include lack of
randomization and retrospective design, lack of disease specificity due to inaccurate identification of comorbid conditions
with ICD-9 classification, and a small number of cases missing preoperative BMI data which may have affected the
results.
Magallares et al. (2015) conducted a meta-analysis of 21 studies evaluating the mental and physical health-related quality
of life (HR-QOL) measures with the Short Form-36 (SF-36) before and after bariatric surgery. Study authors reported that
obese patients scored less in the mental health component of SF-36 prior to bariatric surgery (n = 2680) compared with
after surgery (n = 2251). Similar results were observed in the physical health component of SF-36. Study authors
concluded that obese patients experienced strong improvement in mental and physical QOL measures following surgery.
The findings are limited by lack of comparison group.
A retrospective cohort study was conducted by Yska et al. (2015) within the Clinical Practice Research Datalink involving
2978 patients with a record of bariatric surgery, with a BMI of > 35. They identified 569 patients with T2D and matched
them to 1881 patients with T2D without bariatric surgery. Data on the use of medication and laboratory results were
evaluated. Among patients undergoing bariatric surgery, the authors found a prevalence of 19.1% for T2D. Per 1000
person-years, 94.5 T2D remissions were found in patients who underwent bariatric surgery compared with 4.9 remissions
in matched control patients. Patients with T2D who underwent bariatric surgery had an 18-fold increased chance for
remission (adjusted relative rate [RR], 17.8; 95% CI, 11.2-28.4) compared with matched control patients. The authors
conclude that bariatric surgery strongly increases the chance for remission of T2D with gastric bypass and sleeve
gastrectomy having a greater effect than gastric banding. Limitations included discrepancy between the patient’s actual
use of medication and what was recorded along with incomplete recording of clinical and laboratory testing.
A 2014 Cochrane Systematic Review of RCTs by Colquitt et al. found that surgery results in greater improvement in
weight loss outcomes and weight associated comorbidities compared with non-surgical interventions, regardless of the
type of procedures used. They noted the overall quality of evidence in this analysis to be moderate. When compared with
each other, certain procedures resulted in greater weight loss and improvements in comorbidities than others. Outcomes
were similar between RYGB and SG, and both of these procedures had better outcomes than AGB. However, in one
RCT, the LRGYB procedure resulted in greater duration of hospitalization in two RCTs (4/3.1 versus 2/1.5 days) and a
greater number of late major complications (26.1% versus 11.6%). For people with very high BMI, biliopancreatic
diversion with duodenal switch resulted in greater weight loss than RYGB. Duodenojejunal bypass with sleeve
gastrectomy and LRYGB had similar outcomes; however, this was based on one small trial. Isolated SG led to better
weight-loss outcomes than AGB after three years follow-up. This was based on one trial only. Weight-related outcomes
were similar between laparoscopic gastric imbrication and LSG in one trial. Across all studies adverse event rates and
reoperation rates were generally poorly reported. The authors also found that most trials followed participants for only one
or two years, therefore the long-term effects of surgery remain unclear. In addition, open RYGB, LRYGB and LSG led to
losses of weight and/or BMI but there was no consistent picture as to which procedure was better or worse in the seven
included trials. (The following publications previously cited in this policy, are included in this systematic review: Dixon
2008, Mingrone 2012, Schauer 2012).
A randomized, nonblinded, single-center trial, Schauer et al. (2012) evaluated the efficacy of intensive medical therapy
alone versus medical therapy plus RYGB or SG in 150 obese patients with uncontrolled T2D. The mean age of the
patients was 49 ±8 years, and 66% were women. The average glycated hemoglobin level was 9.2 ±1.5%. The primary
end point was the proportion of patients with a glycated hemoglobin level of 6.0% or less 12 months after treatment. In
obese patients with uncontrolled T2D, 12 months of medical therapy plus bariatric surgery achieved glycemic control in
significantly more patients than medical therapy alone. The authors conclude that further studies will be necessary to
assess the durability of these results.
In a systematic review and meta-analysis, Kadeli et al. (2012) evaluated whether preoperative weight loss before gastric
bypass correlates to weight loss up to 1-year post-surgery. Of the 186 studies screened, 12 were identified. A meta-
analysis was performed to further classify studies (A class, B class, regression, and rejected). The authors conclude that
losing weight leads to better outcomes because a patient entering surgery with a lower weight than someone entering
surgery without weight loss had more weight loss in total. (Publication by Still 2007, which was previously cited in this
policy, is included in this systematic review).
Bariatric Surgery
Page 14 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Biliopancreatic Diversion/Biliopancreatic Diversion with Duodenal Switch (BPD/DS)
Strain et al. (2017) reported nine-year outcomes of BPD/DS. Initially 284 patients received a BPD/DS; 275 patients (69.8
% women) age 42.7 years, BMI 53.4 kg/m2 qualified for baseline analysis. Two hundred seventy-five patients were
available in year 1; 275 patients in year 3; 273 patients in year 5; 259 patients in year 7; and 228 patients in year 9.
Gender distribution was not different. BMI was 30.1 at 1 year and 32.0 at 9 years. Body fat was reduced to 26 % after 2
years. Nutritional problems developed in 29.8% of patients over the course of observation. There were significant positive
changes in quality of life between baseline and year 1 for most patients. Data showed that after surgery, the resolution of
comorbidities continued for the 9-year follow-up period. Weight loss during the first year was well maintained, resolving
comorbidities, and improving quality of life. According to the authors, rates of surgical complications resemble other
bariatric procedures; however long-term nutritional deficiencies are of concern. The findings are limited by lack of
comparison group.
Gastric Bypass (Roux-en-Y; Gastrojejunal Anastomosis)
Ikramuddin et al. (2018) conducted an observational follow-up of a multi-center randomized clinical trial involving 120
participants with T2D who had a hemoglobin A1c (HbA1c) level of 8.0% or higher and a BMI between 30.0 and 39.9.
Lifestyle-intensive medical management intervention was based on the Diabetes Prevention Program and Look AHEAD
trials for 2 years, with and without (60 participants each) RYGB followed by observation to year 5. Ninety-eight (82%)
patients completed 5 years of follow-up. At 5 years, 13 participants (23%) in the gastric bypass group and 2 (4%) in the
lifestyle-intensive medical management group had achieved the composite triple end point (difference, 19%; 95% CI, 4%-
34%; p = 0.01). In the 5th year, 31 patients (55%) in the gastric bypass group vs 8 (14%) in the lifestyle-medical
management group achieved an HbA1c level of less than 7.0% (difference, 41%; 95% CI, 19%-63%; p = 0.002).
Participants undergoing RYGB had more serious adverse events than did the lifestyle-medical management intervention,
66 events versus 38 events, most frequently gastrointestinal events, and surgical complications such as strictures, small
bowel obstructions, and leaks. The authors concluded that in this patient population there remained a significantly better
composite triple end point in the surgical group at 5 years. However, because the effect size diminished over 5 years,
further follow-up is needed to understand the durability of the improvement. One limitation included a poorly controlled
glycemic group of patients thus unsure if study results would be the same with a group of better controlled glycemic
patients. Additional limitations included incomplete follow up creating opportunity for bias and testing of a single type of
bariatric surgery therefore unable to apply conclusions to other bariatric surgical approaches.
In a matched observational cohort study, Liakopoulos et al. (2017) evaluated 6132 patients with a baseline BMI of 42
kg/m2 and T2D who underwent RYGB compared to patients who had not undergone RYGB. Over a 6-year follow-up
period, beneficial changes in BMI, hemoglobin A1C, blood lipids and blood pressure were seen compared with controls.
The authors concluded that improvements in risk factors might contribute to the reduction of mortality risk after RYGB in
obese individuals with type 2 diabetes, but the main effect seems to be mediated through a decrease in BMI, which could
serve as a proxy for several mechanisms.
In a retrospective analysis, Jirapinyo et al. (2017) evaluated the Bariatric Quality of Life (BQoL) scores for 56 patients who
underwent RYGB. The enrolled patients were divided into two groups: stable weight and weight regain with a review of
the BQoL Index scores for each. The authors demonstrated and found in addition to a return to comorbid illness, weight
regain was associated with worsening QoL thus showing the importance of close follow-up, early recognition, and
intervention. Limitations included lack of established definition of weight regain in the current literature, the imbalance of
weight regain and weight stable patients, and the retrospective nature of the study.
In a systematic review and meta-analysis, Yan et al. (2016) compared RYGB surgery versus medical treatment for type 2
diabetes mellitus T2D in obese patients. Six RCTs with a total of 410 patients with obesity and T2D were included, and
follow-up ranged from 12 to 60 months. The pooled analysis of T2D remission rates revealed a significantly higher
remission rate after RYGB surgery than after medical treatment alone. The meta-analysis showed a significant lower BMI
in individuals who underwent RYGB than those who received medical therapy alone. Based on the results, the authors
conclude that RYGB surgery is superior to medical treatment for short- to medium-term remission of T2D, improvement of
metabolic condition, and cardiovascular risk factors. The authors recommend well-designed studies with consistent
definition of adverse events, as well as a larger number of RCTs with long-term follow-up (> 60 months) to evaluate the
safety and long-term benefits of RYGB surgery on obese patients with T2D.
Cooper et al. (2015) assessed weight loss and occurrence of weight regain among patients (n = 300) at 1 year follow-up
who underwent RYGB at a single institution. The mean weight regain for all patients was 23.4 % of maximum weight loss.
Using categorical analysis, mean weight regain in the < 25, 25-30, 30-35, and > 35 % weight loss cohorts was 29.1, 21.9,
20.9, and 23.8 %, respectively. Excessive weight regain, defined as 25 % of total lost weight, occurred in 37 % of
patients. Despite the percentage of weight loss over the first year, all cohort patient groups regained on average between
Bariatric Surgery
Page 15 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
21 and 29 % of lost weight. Excessive weight gain was experienced by over one third of patients. Greater initial absolute
weight loss leads to more successful long-term weight outcomes.
Robotic-Assisted Gastric Bypass Surgery
Beckmann et al. (2020) conducted a retrospective analysis on 108 laparoscopic RYGB surgeries and 114 robotic RYGB
surgeries which were performed between 2016 and 2019. Analysis found operation time for the robotic RYGB was
significantly shorter, had less complications and fewer revisions were required when robotic surgery was used. The
authors concluded robotic RYGB surgery is safe and effective. Findings are limited by lack of randomization.
Gray et al. (2017) conducted a retrospective review of adult patients undergoing laparoscopic revisional bariatric surgery
(LRBS) or robotic revisional bariatric surgery (RRBS). A total of 84 patients who underwent LRBS (n = 66) or RRBS (n =
18) were included. The index operation was AGB in 39/84 (46%), sleeve gastrectomy (VSG) in 23/84 (27%), RYGB in
13/84 (16%), and vertical banded gastroplasty (VBG) in 9/84 (11%). For patients undergoing conversion from AGB (n =
39), there was no difference in operative time, length of stay, or complications by surgical approach. For patients
undergoing conversion from a stapled procedure (n = 45), the robotic approach was associated with a shorter length of
stay (5.8 ±3.3 vs 3.7±1.7 days, p = 0.04) with equivalent operative time and post-operative complications. There were
three leaks in the LRBS group and none in the RRBS group (p = 0.36). Major complications occurred in 3/39 (8%) of
patients undergoing conversion from AGB and 2/45 (4%) of patients undergoing conversion from a stapled procedure (p =
0.53) with no difference by surgical approach. RRBS is associated with a shorter length of stay than LRBS in complex
procedures and has at least an equivalent safety profile. Long-term follow-up data is anticipated.
Ayloo et al. (2016) retrospectively reviewed their experience with robotic approaches to RYGB using prospectively
maintained data. Procedures were categorized into three groups: laparoscopic, hybrid robotic (HR), and total robotic (TR).
The study included 192 consecutive patients who underwent laparoscopic, HR, or TR surgery. Mean patient age,
preoperative BMI, and preoperative weight were 40.4 ±9.3 years (range 22-64), 46. 2 ±5.9 kg/m
2
(range 35-64), and 130.3
±22.1 kg (range 76.7-193.4) respectively. Ninety-two patients (47.9%) had undergone previous abdominal surgery. Mean
operative time, estimated blood loss, and length of stay were 223.4 ±39.2 min (range 130-338), 21.9 ±18.8 mL (range 5-
10), and 2.6 ±1.1 days (range 2-15), respectively. There were 248 concomitant procedures such as upper endoscopy,
cholecystectomy, etc., 7 revisional surgeries, and 2 conversions to open surgery. Intraoperative complications included
one liver laceration and one bowel injury. There were two cases each of bowel obstruction, transfusions, and deep vein
thrombosis/pulmonary embolus, but no deaths or anastomotic leaks. Although there were variables such as different
concomitant procedures, the authors conclude that early experience with a total robotic approach for RYGB appears to be
safe, with similar outcomes to the laparoscopic approach. The findings are however limited by lack of randomization.
Ahmad et al. (2016) conducted a retrospective review to compare the operative and early peri-operative outcomes
between laparoscopic and robotic-assisted RYGB. There were no statistically significant differences in complication rates,
estimated blood loss, or length of stay between the two groups. There was a significant difference between the total
operative times (135.30 ±37.60 min for the laparoscopic procedure versus 154.84 ±38.44 min for the robotic procedure, p
< 0.05). There were no adverse intraoperative events, conversions to open procedures, leaks, strictures, returns to the
operating room within 30 days, or mortalities in either group. The authors concluded that both techniques are comparable
in terms of safety, efficacy, and operative and early perioperative outcomes. The findings are however limited by lack of
randomization.
In a systematic review and meta-analysis, Bindal et al. (2015) evaluated the role of robotics in bariatric surgical
procedures compared with laparoscopic approaches. Several studies showed a lower complication rate with the robotic
platform including leaks, hemorrhage, and stricture. Another advantage noted by the authors for the use of the robotic
system is improved ergonomics and lesser operator fatigue. The authors observed that the use of robotics may provide
specific advantages in some situations and overcome limitations of laparoscopic surgery. With the advent of newer
technologies in robotics the authors conclude that it will provide an empowering tool to the surgeons, which can potentially
change the way surgery is practiced. (Publication by Sanchez 2005, which was previously cited in this policy, is included
in this systematic review).
Economopoulos et al. (2015) conducted a systematic review and meta-analysis to evaluate the available literature on
patients treated with robotic RYGB and compared the clinical outcomes of patients treated with robotic RYGB with those
treated with the standard laparoscopic RYGB. Fourteen comparative and 11 non-comparative studies were included in
this study, reporting data on 5145 patients. Based on their review they found robotic-assisted RYGB was associated with
significantly less frequent anastomotic stricture events, reoperations, and a decreased length of hospital stay compared
with the standard laparoscopic procedures; however, these findings should be interpreted with caution given the low
number and poor quality of the studies currently available in the literature.
Bariatric Surgery
Page 16 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Laparoscopic Adjustable Gastric Banding (LAGB)
In a longitudinal case series, Mistry et al. (2018) reported changes in glycemic control, blood pressure and lipids 5 years
following LAGB combined with medical care in patients with T2D. A total of 200 patients (age 47 ±9.7 years; body mass
index [BMI] 52.8 ±9.2 kg m
-2
; glycosylated hemoglobin (HbA1c) 7.9 ±1.9% [62.8 mmol mol
-1
]; women, n = 123 [61.5%];
insulin treatment, n = 71 [35.5%]) were included. The mean follow-up was 62.0 ±13.0 months (range 18-84 months).
There were significant reductions in body weight (-24.4 ±12.3% [38 ±22.7 kg]), HbA1c (-1.4 ±2.0%), systolic blood
pressure [BP] (-11.7 ±23.5 mmHg), total cholesterol and triglyceride levels. The proportion of patients requiring insulin
reduced from 36.2% to 12.3%. The overall band complication rate was 21% (21 patients). The authors concluded that
LAGB, when combined with multidisciplinary medical care, significantly improved metabolic outcomes in patients with T2D
independent of diabetes duration, and baseline BMI over 5 years. Diabetes duration and baseline BMI did not predict
changes in glycemic control, BP or lipids following LAGB. The findings are limited by lack of comparison group.
Froylich et al. (2018) conducted a retrospective case series of LAGB in 74 patients. The mean age at LAGB placement
was 50.5 ±9.6 years, and the mean BMI was 45.5 ±4.8 kg/m2. Preoperative comorbidities were diabetes mellitus (13.5%),
hypertension (32%), hyperlipidemia (12.1%), obstructive sleep apnea (5.4%), joints disease (10.8%), mood disorders
(5.4%), and gastro-esophageal reflux disease (GERD) symptoms (8.1%). The mean follow-up was 162.96 ±13.9 months;
44 patients (59.4%) had their band removed, and 22 (30%) had another bariatric surgery. The follow-up BMI was 35.7
±6.9 (p < 0.001), and the % TWL was 21.0 ±0.13. There was no improvement in any of the comorbidities. GERD
symptoms worsened at long-term follow-up (p < 0.001). Undergoing another bariatric procedure was associated with a
higher weight loss (OR 12.8; CI 95% 1.62-23.9; p = 0.02). LAGB required removal in the majority of patients and showed
poor resolution of comorbidities with worsening of GERD-related symptoms. In the authors’ opinion, patients who go on to
have another bariatric procedure have more durable weight loss outcomes.
In a retrospective case series, Khoraki et al. (2018) reported long-term outcomes from a cohort of 208 patients who
underwent LAGB. Complete follow-up was available for 90% at one year (186/207), 80% at five years (136/171), and 71%
at ten years (10/14). Percentage of EWL at one, five, and ten years was 29.9, 30, and 16.9, respectively. LAGB failure
occurred in 118 (57%) and 48 patients (23.1%) required a reoperation. Higher baseline BMI was the only independently
associated factor (OR 1.1; 95%CI 1.0-1.1; p = 0.016).
Giet et al. (2018) conducted a retrospective study of 2246 patients who underwent LAGB. Patients were followed for a
minimum of 2 years, and up to 9 years post-procedure. Operative mortality was zero and there were no in-hospital re-
operations. Mean preoperative weight and BMI were 111.2 ±22.1 kg and 39.9 ±6.7 kg/m
2,
respectively. Mean excess %
BMI loss at 1-, 2-, 5- and 8-years of follow-up was 43.1±25.4, 47.9 ±31.9, 52.4 ±41.7 and 57.1%±28.6, respectively.
There was no significant difference in mean excess % BMI loss between those < 50 or 50 years old (p value =0.23) or
between patients with an initial BMI of < or≥ 50 kg/m
2
(p value = 0.65). Complications over nine years occurred in 130
(5.8%) patients and included: 39 (1.7%) slippage or pouch dilatation, 2 (0.04%) erosions and 76 (3.4%) complications
related to the access port or LAGB tubing. The overall re-operation rate for LAGB complications was 4.2% over 9 years
with a LAGB explanation rate of 1.5%. Thirty-nine LAGBs were converted to a sleeve or gastric bypass procedure, 11 of
these due to complications.
Vinzens et al. (2017) evaluated the long-term results of 405 patients (age 41 ±10 years), with a BMI of 44.3 ±6 kg/m2,
who were treated with LAGB. Mean follow-up was 13±3 years, with a follow-up rate of 85% (range 8-18 years),
corresponding to 343 patients. One hundred patients exceeded 15-year follow-up. In 216 patients (63%), sleeve
gastrectomy, gastric bypass, or biliopancreatic diversion with duodenal switch was performed as revisional surgery.
Twenty-seven patients (8%) refused revisional surgery after band removal. Finally, 100 patients (29%) still had the band
in place at the final follow-up, with a mean BMI of 35 ±7 kg/m2, corresponding to an excess BMI loss of 48 ±27%.
According to the Bariatric Analysis and Reporting Outcome System (BAROS), the failure rate was 25%, and 50% had
what was considered to be a good to excellent outcome. The authors concluded that more than 10 years after LAGB, 71%
of patients lost their bands and only 15% of the 343 followed patients with the band in place had a good to excellent
result. The findings are limited by lack of comparison group.
Angrisani et al. (2013) retrospectively evaluated the efficacy and safety of LAGB in moderately obese subjects with or
without obesity-related co-morbidities. Thirty-four patients with BMI between 30 and 35 kg/m2 and mean percentage
excess weight 48.7 ±9 %) who underwent LAGB were included. Good response was defined as BMI < 30 kg/m
2
or
percentage estimated weight loss > 50. Poor response was defined as BMI > 30 kg/m
2
or percentage estimated weight
loss less than 50 after a minimum of 1 year. Mean weight, BMI and percentage estimated weight loss were recorded at 1,
3, 5 and 7 years and were 77.4 ±7.6, 69.9±10.8, 70.9 ±9.3 and 73.3 ±12.0 kg; 28.8 ±2.9, 26.4 ±3.2, 26.5 ±3.4 and 27.4
±5.0 kg/m2; and 36 ±23, 46.1 ±33.8, 58.6 ±31.5 and 45 ±57, respectively (p < 0.01). Co-morbidities were diagnosed in
17/34 (50 %) patients at baseline and underwent remission or improvement in all cases after 1 year. The authors
Bariatric Surgery
Page 17 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
concluded that LAGB is a safe and effective procedure in patients with a BMI < 35 kg/m2. Small sample size and lack of
comparison group were limitations to this study.
Sleeve Gastrectomy (Vertical Gastrectomy)
Clapp et al. (2018) conducted a meta-analysis to evaluate long-term (7 or more years) outcomes of LSG. Nine studies
met the inclusion criteria, with a total of 2280 patients included initially. Only 652 patients had completed 7 years of
follow-up. At ≥ 7 years, the long-term weight recidivism rate was estimated to be 27.8% (I
2
= .60%; 95% CI: 22.8%-32.7%)
with a range of 14% to 37%. The overall revision rate was estimated to be 19.9% (I
2
= 93.8%; 95% CI: 11.3%-28.5%).
This was broken down into 13.1% (I
2
= 93.8%; 95% CI: 5.6%-20.6%) due to weight regain (5 studies) and 2.9% (I
2
=
60.8%; 95% CI: 1%-4.9%) due to gastroesophageal reflux disease (5 studies). Based on available data up to the
beginning of 2017, in the authors’ opinion bariatric surgeons should be aware of the long-term outcomes of the sleeve
gastrectomy, especially regarding revisions and weight regain. (Publication by Noel 2017, which was previously cited in
this policy, is included in this systematic review).
Felsenreich et al. (2017) evaluated long-term outcomes and complications following SG. 53 patients did not have
symptomatic reflux or hiatal hernia preoperatively and of the 43 patients available for follow-up, six patients (14.0%) were
converted to RYGB due to intractable reflux over a period of 130 months. Ten out of the remaining non-converted patients
(n = 26) also suffered from symptomatic reflux. Gastroscopies revealed de novo hiatal hernias in 45% of the patients and
Barrett's metaplasia in 15%. SG patients suffering from symptomatic reflux scored significantly higher in the RSI (p = 0.04)
and significantly lower in the GIQLI (p = 0.02) questionnaire. This study shows a high incidence of Barrett's esophagus
and hiatal hernias at more than 10 years after SG. Its results therefore suggest maintaining pre-existing large hiatal
hernia, GERD, and Barrett's esophagus as relative contraindications to SG. The limitations of this study include its small
sample size as well as the fact that it was based on early experience with SG-make drawing any general conclusions
about this procedure inconclusive.
Flølo et al. (2017) presented 5-year outcomes after VSG, including complications and revisions, weight change, obesity-
related diseases, and health-related quality of life (HRQOL). Of 168 operated patients (mean age, 40.3 ±10.5 years; 71%
females), 92% completed 2-year and 82% 5-year follow-up. Re-intervention for complications occurred in four patients,
whereas revision surgery was performed in six patients for weight regain and in one patient for GERD. BMI decreased
from 46.2 ±6.4 kg/m
2
at baseline to 30.5 ±5.8 kg/m
2
at 2 years and 32.9 ±6.1 kg/m
2
at 5 years. Remission of T2DM and
hypertension occurred in 79 and 62% at 2 years, and 63 and 60% at 5 years, respectively. The percentage of patients
treated for GERD increased from 12% preoperatively to 29% at 2 years and 35% at 5 years. Preventing weight regain and
GERD are important considerations with this procedure. The findings are limited by lack of comparison group.
Nocca et al. (2017) reported 5-year outcomes from a cohort of 1050 patients who underwent SG (mean preoperative BMI
was 44.58 kg/m
2
) either as the primary or revisional surgical procedure. The overall preoperative rate was 6.8%, and the
most common late complication was GERD (39.1%). After 3, 4 and 5 years of LSG, the average of %EBL was,
respectively, 75.95% (±29.16) (382 patients), 73.23% (±31.08) (222 patients) and 69.26% (±30.86) (144 patients). The
success rate at 5 years was 65.97% (95 patients). The improvement or remission of comorbidities was found,
respectively, in 88.4 and 57.2% of diabetic patients; 76.9 and 19.2% for hypertensive patients and 98 and 85% for
patients with sleep apnea syndrome. The authors conclude that five-year results are very convincing for SG, although
GERD is the main long-term complication. The findings are limited by lack of comparison group.
El Chaar et al. (2017) evaluated the incidence, indications, and outcomes of revisional surgery following LSG in adult
patients. Of the 630 LSGs performed, 481 patients were included in the analysis (mean age and BMI = 46.2 and 44.3,
respectively; 79.5 % female; 82.3 % white). A total of 12/481 patients underwent conversion to a different bariatric
procedure due to inadequate weight loss, GERD, or both. The 6/12 patients with GERD-related symptoms and failed
medical management underwent conversion to RYBG following preoperative wireless Bravo pH monitoring (Given
Imaging) to confirm the diagnosis objectively. The other 6/12 patients with inadequate weight loss received either RYBG
or BPD/DS based on personal choice. Overall, 9/12 patients underwent conversion to RYBG, and 3/12 underwent
conversion to BPD/DS. Median time from the initial surgery to conversion was 27 months (range 17-41). Median operating
room time was 168 min (range 130-268). Median length of stay was 48 h (range 24-72). The follow-up rate at 3 months
was 100 % (12/12 patients). The authors conclude that conversion to RYBG or BPD/DS may be done safely and
effectively in patients present following LSG with refractory GERD or inadequate weight loss. Longer term outcomes are
needed. The findings are limited by lack of comparison group.
Brethauer et al. (2009) performed a systematic review (n = 36 studies) of the evidence on SG as a primary or staged
procedure. Studies included a single nonrandomized matched cohort analysis, RCTs (n = 2 studies) and uncontrolled
case series (n = 33 studies). Of these studies, 13 differentiated that the SG was used as a staged procedure or as a
management strategy for a high-risk patient population. Those patients who underwent SG as a planned staged
Bariatric Surgery
Page 18 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
procedure went on to receive RYGB or BPD/DS within 2 years of SG after improvement of their co-morbidities and
surgical risk status. The mean BMI in all 36 studies was 51.2 kg/m2. The mean baseline BMI was 46.9 kg/m2 for the high-
risk patients (range 49.1-69.0) and 60.4 kg/m
2
for the primary SG patients (range 37.2-54.5). The follow-up period ranged
from 360 months. The mean %EWL after SG reported in 24 studies was 3385%, with an overall mean %EWL of 55.4%.
The mean postoperative BMI was reported in 26 studies and decreased from a baseline mean of 51.2 kg/m
2
to 37.1 kg/m
2
postoperatively. Improvement or remission of T2D was found in more than 70% of patients. Significant improvements
were also seen in hypertension and hyperlipidemia, as well as in sleep apnea and joint pain. The mean complication rate
for a primary procedure was 6.2%, while the mean complication rate for high risk/staged procedure was 9.4%. The overall
mortality rate for all studies was 0.19% which included 0.24% for high risk/staged procedure. Despite the high surgical risk
of this patient population, report complication rates were acceptably low. The authors conclude that SG is an effective
weight loss procedure that can be performed safely as a first stage or primary procedure. Limitations include lack of long-
term follow-up for the high-risk group mainly due to patients who refused a second-stage operation or had sufficient
weight loss and co-morbidity reduction with SG alone.
Revision Surgery
Chierici et al. (2022) conducted a systematic review and meta-analysis to identify which revisional bariatric surgery
performs best after a failed primary restrictive surgery. A literature search was conducted using Embase, PubMed,
Cochrane Library, and Scopus databases which returned 39 retrospective and prospective comparative studies. Inclusion
criteria included patients undergoing revisional bariatric surgery after a failed primary restrictive surgery of LAGB, VBG, or
SG. The authors confirmed SG continues to have a low rate of immediate postoperative complications. The authors found
duodenal switch (DS) and biliopancreatic diversion (BPD) were superior when it came to %EWL and %TWL, but not free
from the risk of weight regain. Secondary SG ensures the lowest rate of early and late complications when compared to
single-anastomosis duodeno-ileal bypass (SADI) and one-anastomosis gastric bypass (OAGB), but it also provides the
worst benefits for either 1 and 3 years %EWL and % TWL thus should not be considered when planning revisional
surgery unless there are exceptional circumstances that warrants its use. RYGB is the most frequently performed
revisional surgery following a primary bariatric procedure, however this approach has not always been justified in terms of
weight loss when compared to SG. In addition, RYGB is more frequently associated with early and late complications
when compared to SG, OAGB, and SADI. Finally, the authors found the most balanced procedures were OAGB and
SADI; these two procedures were determined to have 21.16% and 14.66% more EWL, respectively, after 3 years.
Limitations included retrospective design, surgical intervention allocation bias, heterogeneity, and lack of evaluation of
important outcomes like GERD or malabsorption which could affect the patient’s quality of life. (Publication by Qiu 2018,
which was previously cited in this policy, is included in this systematic review).
Koh et al. (2020) performed at systematic review and meta-analysis to examine the impact revisional bariatric surgery has
on obesity related metabolic outcomes. The analysis included review of 33 articles which contained 1593 patients. The
outcomes examined included improvement of diabetes, hypertension (HTN), hyperlipidemia, and OSA. The surgeries
used for revision included SG, RYGB, pouch revision, duodenal switch, and mini-gastric bypass. The authors found 92%
of the patients improved their diabetes, 81% achieved improvement in HTN and 86% had improvement of OSA. The
authors concluded revisional bariatric surgery improved patient outcomes and should be considered in patients with
persistent metabolic disease after primary bariatric surgery. Limitations included lack of randomized control trials, lack of
long-term outcomes, and significant heterogeneity.
Janik et al. (2019) assessed the safety of revisional surgery to LSG compared to LRYGB after failed LAGB. Converted
LSG cases were matched (1:1) with converted LRYGB patients by age (± year), body mass index (± kg/m), sex, and
comorbidities including diabetes, hypertension, hyperlipidemia, venous stasis, and sleep apnea. A total of 2708 patients
(1354 matched pairs) were included in the study. The mean operative time in conv-LRYGB was significantly longer in
comparison to conv-LSG patients (151 ±58 vs 113 ±45 minutes, p < 0.001). No mortality was observed in either group.
Patients after conv-LRYGB had a clinically increased anastomotic leakage rate (2.07% vs 1.18%, p = 0.070) and
significantly increased bleed rate (2.66% vs 0.44%, p < 0.001). Thirty-day readmission rate was significantly higher in
conv-LRYGB patients (7.46% vs 3.69%, p < 0.001), as was 30-day reoperation rate (3.25% vs 1.26%, p < 0.001). The
length of hospital stay was longer in conv-LRYGB. The authors concluded that a single-stage conversion of failed LAGB
leads to greater morbidity and higher complication rates when converted to LRYGB versus LSG in the first 30 days
postoperatively. These differences are particularly notable with regard to bleed events, 30-day reoperation, 30-day
readmission, operative time, and hospital stay.
Dardamanis et al. (2018) conducted a retrospective comparative study of primary versus revisional LRYGB for insufficient
weight loss after VBG or adjustable gastric banding. Three hundred forty-two LRYGB operations were performed, 245
were primary, and 97 revisional. Median follow-up was 30 months (range 0-108 months). Mean BMI (kg/m2) before
bypass was 45.2 for primary LRYGB (pLRYGB) and 41.1 for revisional laparoscopic RYGB (rLRYGB). Median operative
time and length of stay were longer for rLRYGB 157.5 versus 235 min (p < 0.001) and 6 versus 6.5 days (p = 0.05).
Bariatric Surgery
Page 19 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Conversion to laparotomy was performed in eight patients, 0.4% of primary and 7.2% of revisional. Morbidity rate was
6.5% in pLRYGB versus 10% in rLRYGB (NS). There was one death in the primary group. Percentage of excess BMI loss
was significantly lower in the revisional group at 12, 18, and 24 months of follow-up. The authors concluded that revisional
and primary gastric bypass have no statistical differences in terms of morbidity. The % of excess BMI loss is lower after
revisional gastric bypass during the first 2 years of follow-up. The trend of weight loss or weight regain was similar in both
groups.
Altieri et al. (2018) reported the rate of revisions or conversions (RC) in patients who originally underwent RYGB, LSG, or
LAGB. Patients were followed for at least 4 years. There were 40,994 bariatric procedures with 16,444 LAGB, 22,769
RYGB, and 1781 LSG. Rate of RC was 26.0% for LAGB, 9.8% for SG, and 4.9% for RYGB. Multiple RCs were more
common for LAGB (5.7% for LAGB, 0.5% for RYGB, and 0.2% for LSG). Band revision/replacements required further
procedures compared with patients who underwent conversion to RYGB/SG (939 compared with 48 procedures). The
majority of RCs were not performed at the initial institution (68.2% of LAGB patients, 75.9% for RYGB, 63.7% of SG). Risk
factors for multiple procedures included surgery type, as LAGB was more likely to have multiple RCs. The authors
concluded that reoperation was common for LAGB, but less common for RYGB (4.9%) and SG (9.8%). The RC rate is
almost twice after SG than after RYGB. LAGB had the highest rate (5.7%) of multiple reoperations. Conversion was the
procedure of choice after a failed LAGB.
Wijngaarden et al. (2017) identified that non-responders of LAGB showed inferior weight loss results after revisional
LRYGB compared with responders of LAGB, and primary LRYGB at all moments of follow-up (12, 24, 36 months). This is
based on an observational study of 96 non-responders, and 120 responders. In addition, the failure rate was significantly
higher after revisional LRYGB compared with primary LRYGB (10.9% no responders, 8.5% responders, and 2.5%
primary, p = 0.001).
In a retrospective review of primary LRYGB (pLRYGB) versus revisional LRYGB (rLRYGB) after failed LSG, Malinka et al.
(2017) evaluated 3-year outcomes. There were no significant differences in patient demographics or median BMI (kg/m2)
for pLRYGB or rLRYGB (42.8 ±12.1 vs. 42.3 ±11.5, respectively; p = 0.748). Coexisting comorbidities were rated similarly
in both groups. At 3 years, the percentage of excess weight loss (74.4 ±23.3 vs 52.0 ±26, respectively; p = 0.007) was
higher for pLRYGB than rLRYGB, while similar improvements of coexisting comorbidities could be observed. The authors
concluded that rLRYGB is a feasible and practical surgical approach that allows effective weight loss at 3 years of follow-
up and alleviates refractory reflux symptoms. Although weight loss is lower compared to pLRYGB, resolution or
improvement of coexisting comorbidities appears similar. According to the authors, rLRYGB appears to be a reliable
procedure to address failure after LSG.
Pinto-Bastos et al. (2017) conducted a systematic review of preoperative surgery following the failure of primary bariatric
surgery. The etiology of reasons for undergoing a second surgery includes medical (e.g., fistula, ulcer disease) and
behavioral aspects. Eating and lifestyle behaviors, difficulty in embracing the required lifestyle changes, and
reappearance of depressive and anxious symptoms have been associated with failure of weight loss or weight regain after
primary surgeries. The authors recommend that particular attention be paid to surgical candidates with a history of
difficulties in engaging in healthy eating patterns.
In a retrospective review, Fulton et al. (2017) evaluated outcomes of revisional bariatric surgery in 2769 patients. The
mean preoperative BMI was 44.7 ±9.5 in revision patients compared with 45.7 ±7.6 in primary bariatric surgery patients.
Most revision patients had a prior VBG (48%) or a LAGB (24%). Bands were removed in 36% of all LAGB patients
presenting to clinic. Of the 134 procedures performed in the revision clinic, 83 were bariatric weight loss surgeries, and 51
were band removals. Revision clinic patients experienced a significant decrease in BMI (from 44.7 ±9.5 to 33.8 ±7.5, p <
0.001); their BMI at 12-month follow-up was similar to that of primary clinic patients (34.5 ±7.0, p = 0.7). The authors
identified that complications were significantly more frequent in revision patients than primary patients (41% v. 15%, p <
0.001).
Sharples et al. (2017) conducted a systematic review and meta-analysis of outcomes after revisional bariatric surgery.
2617 patients in 36 studies underwent either adjustable gastric band to Roux-en-Y gastric bypass (B-RYGB) or band to
sleeve gastrectomy (B-SG). There was no difference between the B-RYGB and B-SG groups in morbidity, leak rate or
return to surgery. %EWL following the revisional procedure for all patients combined at 6, 12 and 24 months was 44.5,
55.7 and 59.7%, respectively. There was no statistical difference in %EWL between B-RYGB and B-SG at any time point.
The rates of remission of diabetes, hypertension and obstructive sleep apnea were 46.5, 35.9 and 80.8%, respectively.
Available observational evidence does suggest that revisional bariatric surgery is associated with outcomes similar to
those experienced after primary surgery. Further, high-quality research, particularly RCTs, is required to assess long-term
weight loss, comorbidity, and quality of life outcomes.
Bariatric Surgery
Page 20 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Tran et al., (2016) conducted a systematic review of 24 studies and 866 patients to evaluate outcomes and complications
of different surgical methods of revision that were done after failed primary RYGB. All patients in the studies reported
significant early initial weight loss after revisional surgery. However, of the five surgical revision options considered,
biliopancreatic diversion/duodenal switch, distal RYGB, and gastric banding resulted in sustained weight loss, with what is
considered by the authors as an acceptable complication rate.
Quezada et al. (2016) conducted a retrospective analysis of SG conversion to RYGB (n = 50) due to the observation of
increased complications of SG as the number of procedures increase. Revisions were done due to weight regain, GERD,
or gastric stenosis. At follow-up (over a 3-year period), the authors reported median excess weight loss was 60.7 lbs., all
gastric stenosis symptoms had resolved, and over 90% of GERD patients reported either a resolution or improvement in
symptoms. Despite their findings, long term follow-up on this patient population is needed.
Buttelmann et al. (2015) compared outcomes for patients undergoing diet/exercise intervention with patients undergoing
surgical intervention through restorative obesity surgery-endolumenal (ROSE), band over bypass, and endoscopic gastro
gastric fistula closure. A retrospective analysis of 60 patients was performed on those who underwent gastric bypass and
failed to lose weight. Records were reevaluated at 3, 6, and 12 months after intervention for primary outcomes of, weight
loss and comorbidity resolution. The authors concluded ROSE, band over bypass, and endoscopic fistula closure results
in greater weight loss and trend toward greater comorbidity resolution compared with diet and exercise. This study is
limited by small sample size, lack of randomization, and short-term follow-up.
David et al. (2015) reported their experience in laparoscopic conversion of failed VGB to RYGB or BPD (n = 39), noting
that the reoperation rate for VGB in long-term studies is approximately 50%. Most (89%) of the conversions were
completed laparoscopically. The mean operative time was 195 and 200 min for RYGB and BPD, respectively. There was
no mortality. Complications occurred in 11 patients (28%), 5 in RYGB (19%) and 6 in BPD (42%). At the 3-year follow-up,
the mean body mass index decreased from 47 ±8 kg/m2 to 26 ±4 kg/m2 for BPD, and from 43 kg/m2 to 34 kg/m2 (p =
0.05) for RYGB. Weight (kg) decreased from 110 to 84 and to 92, and from 123 to 81 and 68, at 1 and 3 years for RYGB
and BPD, respectively. The weight loss for RYGB and BPD was equal at 1 year but tended to be better for BPD at 3 years
postoperatively. Laparoscopic conversion of failed VBG to RYGB or BPD was feasible, but it was followed by prohibitively
high complication rates in BPD patients. The authors concluded that the risk: benefit ratio of these procedures in this
series is questionable.
Brethauer et al. (2014) was part of a task force that reviewed current evidence which identified procedure-specific
indications and outcomes for reoperative procedures. 175 articles were included in the systematic review and analysis
and the majority of the published studies were single center retrospective reviews. The evidence supporting reoperative
surgery for acute and chronic complications is described along with additional guiding principles. GERD was identified as
a long-term complication that may occur following a sleeve gastroplasty. The authors indicate treatment begins with
medical management and behavioral modifications; if medical control of GERD fails, surgical revision may be required.
Pediatric and Adolescent Bariatric Surgery
Hoeltzel et al. (2021) evaluated adolescent bariatric surgeries from the Metabolic and Bariatric Surgery Accreditation and
Quality Improvement Program (MBSAQIP) database from 2015 to 2018. Participants included patients 19 years old and
younger with a BMI ≥ 30 kg/m
2
and underwent laparoscopic RYGB or SG. Primary outcomes included mortality and
overall complications; secondary outcomes included rates of readmission and reoperation. A total of 5068 individuals met
inclusion criteria for the study with 78.5% being females and 70.4% being white. Patients between the ages of 10 to 14
years comprised 1.5% of participants, 15 to 17 years 18.5%, and 18 to 19 years 79.9%. The mean BMI was 47.3 kg/m
2
and the most prevalent comorbidities were HTN, OSA, GERD, and diabetes. The 30-day analysis following surgery
demonstrated intraoperative or postop complications in only 1.2% of patients and the death of two patients which was
likely due to internal hernia. The authors concluded that bariatric surgery for adolescents was a safe and effective
procedure with low complication rate and a recommendation of future robust studies to evaluate the long-term outcomes
in this age group of patients.
Alqahtani et al. (2021) analyzed the long-term results and adverse events associated with LSG in children and
adolescents with severe obesity. 2,504 children and adolescents that underwent LSG between 2008 and 2021 were
enrolled in the program. Weight loss was reported in terms of mean weight change, percentage of weight lost, %EWL,
change in BMI, and BMI for age percentile along with assessment of comorbidity conditions. The mean standard deviation
(SD) %EWL for one to three years was 82.3%, for 4 to 6 years was 76.3% and 7 to 10 years was 71.1%; 10-year results
demonstrated that 30% of total weight was lost permanently. Prior to surgery 263 patients were diagnosed with T2D, 227
with dyslipidemia, and 377 had hypertension. After more than 7 years of follow-up, complete remission was observed in
188 patients for T2D, 130 patients for dyslipidemia, and 219 patients for HTN. Only 1% of the patients were readmitted
within the first 90 days after the operation; two patients had a staple line leak and 22 were readmitted with nausea and
Bariatric Surgery
Page 21 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
vomiting. The data showed no significant change in growth velocity, including among participants younger than age 14
years. The authors concluded long-term follow-up after LSG in children and adolescents demonstrates positive weight
loss and comorbidity resolution. The findings are however limited by lack of comparison group.
Lainas et al. (2020) conducted a study to assess whether bariatric surgery was successful for adolescents under the age
of 18. The authors evaluated 84 adolescent patients (57 females, 27 males) that underwent LSG. Surgical postop care
included blood work and diet restrictions with a discharge when oral diet was well tolerated. Patients follow-up included 4
outpatient visits the first year then annually; complete metabolic screening was done at 3 months, again at one year and
annually thereafter. The quality of life was evaluated prior to surgery using the French version of the Short Form 36
questionnaire which assessed general health, physical function, social function, emotional and mental status, and bodily
pain. The scoring ranged from 0-100 with higher scores indicating better wellbeing. All patients were contacted one-year
post-surgery to answer the same questions. Comorbidities assessed included HTN, T2D, OSA, dyslipidemia, arthralgia,
and GERD. According to the authors, the study showed LSG is a safe and effective procedure for patients under the age
of 18, resulting in significant weight loss, comorbidity remission, and improvement in quality of life. In addition, it was felt
that adherence to the medical team was an essential component for successful treatment in this group of patients.
Limitations included small sample size, retrospective design, substantial loss to follow-up thus affecting long-term
outcomes and lack of comparison group.
A Hayes (2019, updated 2022) comparativeness effective review for bariatric surgeries for treatment of obesity in
adolescents analyzed nineteen studies which compared AGB, VSG and RYGB. The authors concluded that while the
body of evidence is moderate in size with a low quality overall, these surgical procedures are superior to medical
management for promoting weight loss and improving obesity-related comorbidities in adolescents. AGB was inferior to
the others, but all three types are associated with low to moderate risk of postop complications and show similar efficacy.
Inge et al. (2018) compared glycemic control in cohorts of severely obese adolescents with T2D undergoing medical and
surgical interventions. Participants in the Teen-LABS group (n = 242) underwent a primary bariatric procedure, while
those in the Youth TODAY consortia (n = 699) were randomized to receive medication alone, or an intensive lifestyle
intervention. After selection of 30 participants from Teen-LABS with diabetes (mean [SD] age at baseline, 16.9 [1.3] years;
21 [70%] female; 18 [66%] white), 63 matched controls from TODAY were selected (mean [SD] age at baseline, 15.3 [1.3]
years; 28 [44%] female; 45 [71%] white) and the two groups were compared. During 2 years, mean hemoglobin A1c
concentration decreased from 6.8% (95% CI, 6.4%-7.3%) to 5.5% (95% CI, 4.7% -6.3%) in Teen-LABS and increased
from 6.4% (95% CI, 6.1%-6.7%) to 7.8% (95% CI, 7.2%-8.3%) in TODAY. Compared with baseline, the BMI decreased by
29% (95% CI, 24%-34%) in Teen-LABS and increased by 3.7% (95% CI, 0.8%-6.7%) in TODAY. Twenty-three percent of
Teen-LABS participants required a subsequent operation during the 2-year follow-up. Compared with medical therapy,
surgical treatment of severely obese adolescents with type 2 diabetes was associated with better glycemic control,
reduced weight, and improvement of other comorbidities. According to the authors, these data support the need for a well-
designed, prospective controlled study to define the role of surgery for adolescents with T2D, including health and surgical
outcomes.
Ryder et al. (2018) evaluated factors associated with long-term weight-loss maintenance following bariatric surgery in
adolescents (n = 50) with severe obesity who underwent RYGB. Follow-up visits at 1 year and at a visit between 5- and
12-years following surgery (follow-up of Adolescent Bariatric Surgery at 5 Plus years (FABS-5+) visit. A non-surgical
comparison group (n = 30; mean ±s.d. age and BMI = 15.3 ±1.7 years and BMI = 52 ±8 kg m
-2
) was recruited to compare
weight trajectories over time. The BMI of the surgical group declined from baseline to 1 year (-38.5 ±6.9%), which, despite
some regain, was largely maintained until FABS-5+ (-29.6 ±13.9% change). The BMI of the comparison group increased
from baseline to the FABS-5+ visit (+10.3 ±20.6%). When the surgical group was split into maintainers and re-gainers, no
differences in weight-related and eating behaviors, health responsibility, physical activity/inactivity, or dietary habits were
observed between groups. However, at FABS-5+, maintainers had greater overall QOL scores than re-gainers (87.5
±10.5 vs 65.4 ±20.2, p < 0.001) and in each QOL sub-domain (p < 0.01 all).
In a retrospective review of 79 adolescents who underwent LSG, Elhag et al. (2018) assessed preoperative levels and
postoperative changes in 4 anthropometric, 15 nutritional and 10 cardiometabolic parameters. At a mean of 24.2 months
post-LSG, significantly reduced mean weight and BMI by 51.82 ±28.1 kg and 17 ±6.24 kg/m
2
, respectively were observed.
The highest prevalence of post-LSG deficiencies pertained to vitamin D, albumin, and ferritin (89.3, 38, and 33.3%,
respectively). Low hemoglobin levels (29.3%) were reported only in females. Trace elements were not deficient.
Significant reductions in percentage of adolescents with elevated low-density lipoprotein (from 66.1 to 38.9%), alanine
aminotransferase (from 45.3 to 10.9%), and aspartate aminotransferase (from 24.1 to 8.6%) levels were reported. Finally,
100% remission of prediabetes cases, and 80% remission of T2Dcases were observed. The slight worsening of
preexisting nutritional deficiencies warrants careful preoperative surveillance and appropriate postoperative nutritional
supplementation.
Bariatric Surgery
Page 22 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Benedix et al. (2017) compared the perioperative course, weight loss and resolution of comorbidities after primary LSG for
morbid obesity between adolescents (n = 362) and adults (n = 15,428). Pre-procedure BMI was similar between these
populations, but the adult cohort had a higher incidence of co-morbidities. Late adolescents experienced the highest
weight loss; resolution rate of comorbidities was lower in adults. Resolution rate of hypertension was significantly higher in
adolescents. In the authors’ opinion, the results at 12 and 24 months demonstrate that LSG is a safe therapeutic option
that can be performed in adolescents without mortality.
Beamish et al. (2017) studied bone health and body composition in 72 adolescents who underwent RYGB. Inclusion
criteria included the following: age 13-18years and BMI > 35kg/m2. Patients underwent dual-energy X-ray absorptiometry
and serum bone marker analyses preoperatively and annually for 2years. Differences in body fat and lean mass
proportions were observed according to sex following RYGB. Mean BMI reduction at 2years was 15.1kg/m2. Body
composition changes included a reduction in fat mass (51.8% to 39.6%, p<0.001) and relative increase in lean mass
(47.0% to 58.1%, p<0.001). In contrast to previous studies in adults, adolescent boys lost a greater percentage of their
body fat than girls (-17.3% vs. -9.5%, p<0.001). Individual bone mineral density Z-scores (BMD-Z) at baseline were within
or above the normal range. The mean (SD) BMD-Z was 2.02 (1.2) at baseline, decreasing to 0.52 (1.19) at 2years.
Higher concentrations of serum CTX (p<0.001) and osteocalcin (p<0.001) were observed in boys throughout the study
period. Levels rose in the first year, before decreasing modestly in the second. Levels of serum markers of bone synthesis
and resorption were higher in boys, whose skeletal maturity occurs later than girls. Bone turnover increased, and BMD
decreased to levels approaching a norm for age. Long-term outcome will determine the clinical relevance.
In a systematic review and meta-analysis, Qi et al. (2017) evaluated the effects of bariatric surgery on glycemic and lipid
metabolism, surgical complications, and quality of life in adolescents with obesity. A total of 49 studies with 3007 patients
were included. RYGB (n = 1216), LAGB (n = 1028), and LSG (n = 665) were the most common bariatric surgeries
performed. At the longest follow-up (range, 12-120 months), bariatric surgery led to an overall 16.43 kg/m2 (95%
confidence interval [CI]: 14.84-18.01) and 31% (95% CI: 28%-34%) reduction in BMI. There were significant
improvements in glycemic and lipid profiles including glycosylated hemoglobin A1C, fasting blood insulin, fasting blood
glucose, total cholesterol, triglyceride, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol,
postoperatively at 12 months. The remission rate of dyslipidemia was 55% (95% CI: 34%-76%), 70% (95% CI: 55%-82%),
and 95% (95% CI: 80%-100%) at 1, 3, and > 5 years after surgery. RYGB produced better improvements than other
surgical procedures. The authors concluded that bariatric surgery in adolescents may achieve significant weight loss, and
glycemic and lipid control. (Publications by Manco 2017, Serrano 2016, Inge 2016, Olbers 2017, Shah 2017, Hervieux
2017, and O’Brien 2010, which were previously cited in this policy, are included in this systematic review).
The Teen-Longitudinal Assessment of Bariatric Surgery (LABS) Study was a prospective, multicenter, observational
study, which enrolled 242 adolescents (≤ 19 years of age) who were undergoing bariatric surgery from March 2007
through February 2012 at 5 U.S. adolescent bariatric surgery centers. The patients underwent RYGB (n = 161), SG (n =
67), or LAGB (n = 14). Ryder et al. (2016) evaluated 2-year outcomes to determine the impact of bariatric surgery on
functional mobility and musculoskeletal pain. Participants completed a 400-m walk test prior to bariatric surgery (n = 206)
and at 6 months (n = 195), 12 months (n = 176), and 24 months (n = 149) after surgery. Time to completion, resting heart
rate (HR), immediate posttest HR, and HR difference (resting HR minus posttest HR) were measured and
musculoskeletal pain concerns, during and after the test, were documented. Data were adjusted for age, sex,
race/ethnicity, baseline BMI, and surgical center (posttest HR and HR difference were further adjusted for changes in time
to completion). Compared with the baseline, the post-surgery data showed an improvement in all measurements at all
times measured. The authors conclude that bariatric surgery in adolescents with extreme obesity is associated with
significant improvement in functional mobility and in the reduction of walking-related musculoskeletal pain up to 2 years
after surgery. Findings are however limited by lack of comparison group.
In a systematic review and meta-analysis, Paulus et al. (2015) evaluated the efficacy, safety, and psychosocial health
benefits of various bariatric surgical techniques (RYGB, LAGB, LSG) as a treatment for morbid obesity in adolescents. A
total of 37 peer-reviewed articles were included, although the studies were mainly observational and varied in quality.
Authors of 9 studies were contacted for additional information. All three procedures lead to significant weight loss in
morbidly obese adolescents, and weight loss is most pronounced after RYGB. For LSG studies, long-term follow-up were
not yet available. While adverse events were relatively mild and long-term complication rates were acceptable, they were
more frequent and more serious after RYGB than after LAGB. In the currently available follow-up after LSG, the rate of
adverse events appeared to be similar to that after LAGB. Although a healthy nutritional status in adolescents is important
to prevent developmental and growth deficiencies, standard postoperative vitamin supplementation regimens and the
occurrence of deficiencies were not reported in most studies (not at all in LSG studies). However, more and more severe
deficiencies occurred after RYGB than after LAGB. Reduction of comorbidity, which is pivotal for health gain, was
impressive in all techniques, and QOL consistently showed improvement, although follow-up up to 24 months may not be
Bariatric Surgery
Page 23 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
enough to capture negative long-term effects in life after bariatric surgery. A limitation of this review is the lack of high-
quality, prospective randomized controlled trials, which increases the risk of bias and therefore introduces heterogeneity.
Zitsman et al. (2015) studied a population of morbidly obese teenagers (n = 137) who underwent LAGB to evaluate its
safety and effectiveness. The mean weight gain between enrollment and LAGB was 4.7 kg. Mean preoperative weight,
body BMI, and excess BMI were 136.1 kg, 48.3 kg/m2, and 23.6 kg/m2, respectively. Mean BMI at 6, 12, 18, 24, and 36
months was 43.8, 41.6, 41.5, 40.5, and 39.3. Excess BMI loss was 28.4%, 35.9%, and 41.1% at 1, 2, and 3 years postop.
Co-morbid conditions improved or resolved with weight loss after LAGB. Thirty patients (22%) underwent one or more
additional operations for complications. Twenty-seven patients (20%) converted to other weight loss procedures or had
their bands removed. The authors concluded that morbidly obese adolescents can lose weight successfully and
experience health improvement following LAGB, but the role of LAGB in the younger population requires long-term
evaluation.
Bariatric Artery Embolization (BAE)
There is insufficient evidence for bariatric artery embolization and its outcomes for weight loss; additional robust RCTs are
warranted for safety and efficacy along with long-term follow up.
Reddy et al. (2020) conducted a single-center, sham controlled, masked RCT to evaluate the efficacy of transcatheter
bariatric embolization (TBE) for weight reduction in obesity. Participants were randomized to either sham procedure (n =
20) or TBE targeting the left gastric artery using embolic beads (n = 20). The primary efficacy endpoint was the difference
in TBWL between the two groups at 6 months. All patients entered a lifestyle counseling program and follow-up was
completed by physicians that were masked to allocated therapy. At 6 months, the TBWL for TBE in the intention to treat
(ITT) population was 7.4 kg compared to 3.0 kg for sham procedure. The change in BMI at 6 months for ITT was -2.6 in
TBE versus -1.1 in sham. The TBE ITT population did maintain the weight loss at 12 months. Patients within the sham
group were unblinded at 6 months and permitted to crossover to TBE and then only initial group was followed for 12
months. Limitations included small sample size, single center, no control group after 6 months, and possibility that the
efficacy of TBE was related to subject participation in weight management counseling as it is unknown if TBE alone would
have an impact on obesity without lifestyle counseling. Additionally, four subjects withdrew consent after randomization
and another three prior to the 6-month visit. Furthermore, the clinical significance of the effect, its long-term sustainability,
and safety are unclear.
Hafezi-Nejad et al. (2019) conducted a systematic review and meta-analysis of case series investigating the safety and
efficacy of left gastric artery (LGA) embolization as a bariatric procedure. Meta-regression was performed to assess
associations of age, sex, body mass index, and ghrelin and leptin levels with weight change after LGA embolization were
selected. Six case series published between January 2014 and April 2019, comprising 47 patients investigating the safety
and/or efficacy of LGA embolization for weight loss were included in the meta-analysis. The results showed a mean
weight loss of 8.68 kg (19.14 lbs.) after 12 months of follow-up, approximately 8% of baseline total body weight which is
superior to weight loss from diet and exercise, and comparable to other more invasive interventions. Transient superficial
mucosal ulcers were common after LGA embolization, and one case of major complications (severe pancreatitis, splenic
infarct, and gastric perforation) was identified. There were considerable variations in patient age, sex distribution, and
baseline characteristics among the studies. Significant variation was observed in the duration of follow-up, which ranged
from 3 months to 2024 months. Limitations of this study include variations in the indications for LGA embolization, study
designs, embolization techniques, follow-up plans, dietary assessments, patient comorbidities, and availability of control
subjects, the authors concluded that LGA embolization is an investigative method and not yet proven to be effective
management for obesity. Larger studies are needed to expand these findings and determine other correlates of weight
loss after LGA embolization. (Publications by Bai 2018, Syed 2016, and Weiss 2017, which were previously cited in this
policy, are included in this systematic review).
Weiss et al. (2019) evaluated the safety and efficacy of bariatric artery embolization up to twelve months following surgery
in 20 severely obese patients (five of which are identified below in the Weiss et al. (2017) case series). The primary
endpoint was weight loss with additional end points assessed. Bariatric embolization was performed successfully in all
participants. Participants experienced mean excess weight loss of 8.2% at one month, 11.5% at 3 months, 12.8% at six
months and 11.5% at twelve months. The mean total weight loss was 7.6kg at twelve months. As a result of loss to follow-
up, 18 participants remained at three months, 16 at six months, and 15 at twelve months. No major adverse events (AE)
were identified and only eleven minor AE occurred in eight participants. The authors found bariatric embolization is well
tolerated and promotes clinically relevant weight loss in adults with severe obesity. Limitations included lack of
comparison group, small sample size, insufficient data due to lack of continuous follow up for several participants,
required weight management compliance before the embolization procedure on the first five participants only and a large
portion of participants were African American thus overrepresenting that population.
Bariatric Surgery
Page 24 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Gastric Electrical Stimulator (GES)
While gastric electrical stimulation may provide benefit for obesity, additional well designed RCTs with long-term follow-up
are warranted to demonstrate safety and efficacy.
In this 2020 first-in-human (early feasibility) multicenter, phase 1, open prospective cohort study, (Paulus et al., 2020) the
authors assessed the safety of the Exilis
gastric electrical stimulation. They also sought to investigate whether the
settings can be adjusted for comfortable chronic use in Class II or III obese patients. Meal intake and gastric emptying and
motility were also evaluated. In this study, 20 obese patients were implanted with the Exilis system and amplitude was
individually set during 4 amplitude titration visits. Subjects underwent two blinded baseline test days (GES ON vs. OFF),
after which long-term, monthly follow-up continued for up to 52 weeks. The results suggested that this device is safe and
caused no patient discomfort. At baseline food intake and satiety were not significantly different when the device was on
or off, and significant weight loss occurred at week 26, with EWL of 14% at 52 weeks. The authors conclude that the data
were comparable with studies of subjects on diet and/or exercise alone, but disappointing when compared to minimally
invasive procedures, such as gastric banding or endoscopic gastroplication. Furthermore, the authors did not observe
changes in plasma glucose and insulin levels which other bariatric procedures are known to improve. The authors
concluded that considerably more basic research is required before clinical use. Limitations included small sample size,
lack of control group, and lack of long-term outcomes.
In a in a 12-month prospective multicenter study, Morales-Conde et al. (2018) monitored all participants (n = 47) up to 24
months after laparoscopic implantation of a closed-loop GES system (CLGES). Weight loss, safety, quality of life (QOL),
and cardiac risk factors were analyzed. Weight regain was limited in the 35 (74%) participants remaining enrolled at 24
months. Mean %TBWL changed by only 1.5% between 12 and 24 months, reported at 14.8% (95% CI 12.3 to 17.3) and
13.3% (95% CI 10.7 to 15.8), respectively. The only serious device-/procedure-related AEs were two elective system
replacements due to lead failure in the first 12 months, while improvements in QOL and cardiovascular risk factors were
stable thru 24 months. The authors conclude that during the 24-month follow-up, CLGES was shown to limit weight regain
with strong safety outcomes, including no serious AEs in the second year. They hypothesize that CLGES and objective
sensor-based behavior data combined to produce behavior change, and in their opinion supports GES as a safe obesity
treatment with potential for long-term health benefits. Larger well-designed randomized controlled trials are needed to
further evaluate the safety and efficacy of GES therapy in the treatment of obesity.
In a post-implant analysis, Alarcón Del Agua I, et al. (2017) evaluated possible preoperative predictors for obtaining
clinically meaningful weight loss with GES. Ninety-seven obese participants in a prospective multicenter study conducted
in nine European centers were implanted laparoscopically with the abiliti
®
CLGES system. The mean 12-month %EWL
with CLGES was 35.1 ±19.7%, with a success rate of 52% and a failure rate of 19%. Significant predictors of success
were BMI < 40 kg/m
2
and age ≥ 50 years, increasing probability of success by 22 and 29%, respectively. A low F1-
cognitive-restraint score was a significant predictor of failure (p = 0.004). The best predictive model for success included
F1-cognitive-restraint, F2-disinhibition, BMI < 40, and age ≥ 50 (p = 0.002). The authors concluded that age, preoperative
BMI, and F1-cognitive-restraint and F2-disinhibition scores from a preoperative questionnaire are predictive of weight loss
outcomes with closed-loop GES and may be used for patient selection.
In a systematic review, Cha et al. (2014) evaluated the current state regarding implantable gastric stimulators. Thirty-one
studies consisting of a total of 33 different trials were included in the systematic review for data analysis. Weight loss was
achieved in most studies, especially during the first 12 months, but only very few studies had a follow-up period longer
than 1 year. Among those that had a longer follow-up period, many were from the Transcend(
®
) (Implantable Gastric
Stimulation) device group and maintained significant weight loss. Other significant results included changes in
appetite/satiety, gastric emptying rate, blood pressure and neurohormone levels or biochemical markers such as ghrelin
or HbA1c, respectively. The authors conclude that although gastric electrical stimulation holds great promise, stronger
evidence is required through more studies with a standardized way of carrying out trials and reporting outcomes, to
determine the long-term effect of gastric electrical stimulation on obesity. (Publications by Shikora 2009, Sarr 2012, and
Camilleri 2008, which were previously cited in this policy, are included in this systematic review).
Intragastric Balloon (IGB)
There is mixed evidence regarding the long-term efficacy and safety for intragastric balloons and their use with obesity;
additional well designed RCTs and long-term data are warranted.
Based on a clinical evidence assessment by ECRI (2022), the evidence for the Spatz3
®
IGB is inconclusive. Assessment
of two RCTs, three nonrandomized comparison studies, two case series, and two chart reviews assessing weight loss
and adverse events for Spatz3
®
in adults with obesity revealed short-term clinically significant weight loss but whether
these results were long-term remains to be seen. Limitations included small sample sizes, retrospective design of studies,
Bariatric Surgery
Page 25 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
lack of randomization, masking, and controls along with single-center focus. Large robust studies with long-term results
are warranted and several ongoing clinical trials may address this in the future.
Zou et al. (2021) performed a systematic review and meta-analysis to evaluate the efficacy of the intragastric balloon
(IGB) as an obesity management tool for metabolic dysfunction-associated fatty liver disease (MAFLD). Thirteen
observational studies and one RCT met the inclusion criteria (624 participants in total). The results showed that over time,
IGB therapy significantly improved the serum markers homeostasis model assessment of insulin resistance (HOMA-IR),
alanine aminotransferase (ALT), aspartate aminotransferase (AST), and gamma-glutamyl transpeptidase (GGT) levels
from baseline to follow-up. The authors concluded that IGB has the potential to become a multidisciplinary management
tool of MAFLD, however IGB is a temporary measure, and if the patient cannot maintain an active lifestyle after the first
balloon is removed, relapse of MAFLD is expected. Limitations include lack of comparison group; further RCT’s are
needed.
Hayes (2018, updated 2022) low-quality evidence suggests that IGB have mixed results with regard to weight loss over
the short term when used as an adjunct to diet and exercise. These devices are consistently associated with high AE and
all studies analyzed lacked long term follow up on maintaining weight loss and safety concerns.
A 2021 ECRI clinical evidence assessment on the Orbera
®
Intragastric Balloon System concluded that the evidence is
inconclusive with mixed results, and shows the use of Orbera results in short-term, clinically significant weight loss in most
patients; however, most patients regain weight, and by 1 year, the sustained weight loss has unclear clinical significance.
Additional randomized studies are needed to determine whether Orbera use can reduce bariatric surgery risks for patients
who are not surgery candidates and/or use the device to lose weight to become eligible for surgery. Additional studies that
directly compare Orbera with other IGBs would also be useful.
In a multicenter, open-label industry-sponsored RCT, Abu Dayyeh et al. (2021, included in ECRI 2022 report above)
investigated the safety and efficacy of the Spatz IGB in adults with obesity. 288 patients were randomly assigned to
receive either the IGB plus dietary and exercise counselling or dietary and exercise counselling alone for 32 weeks.
Inclusion criteria were patients aged 22-65 years, BMI of 30 kg/m
2
or greater for past two years, history of unsuccessful
non-surgical weight loss methods and willingness to participate in the required dietary restrictions. The IGB was implanted
via esophagogastroduodenoscopy (EGD) under conscious or monitored anesthesia sedation; depending on the patient’s
height an initial volume of 400 ml, 450 ml, 500 ml, or 550 ml was utilized. During the 32 weeks, all patients followed a
10001200 kcal/day diet and exercise plan. After 32 weeks, the IGB was removed and patients were followed for another
24 weeks. Primary outcomes consisted of %TBWL and clinical responder rate, which was achieved by a decrease of at
least 5% total bodyweight loss at 32 weeks. Mean %TBWL at 32 weeks was 15·0% (95% CI 13·9-16·1) in the IGB group
versus 3·3% (2·0-4·6) in the control group (p < 0·0001). The authors found the adjustable IGB combined with lifestyle
modification enabled significant weight loss over a period of 6 months with an observed acceptable safety profile.
Limitations included no masking or sham intervention, and an approximately 20% loss to follow-up at 32 weeks. Future
studies should assess the long-term safety of the device.
ECRI (2020) Health Technology Assessment focused on the safety and efficacy of the Elipse
and Obalon
®
, two
ingestible IGBs. The evidence was inconclusive citing RCTs would be beneficial to determine whether any differences
exist in weight loss and serious AE risks. Available clinical guideline recommendations on IGB are mixed and none pertain
to ingestible IGB. Thus, major evidence gaps remain and additional comparative studies of ingestible and conventional
IGB are needed.
Moore et al. (2019, included in the ECRI report above) performed a retrospective analysis of patients that underwent the
Obalon Balloon System (OBS), a swallowable, gas-filled intragastric balloon system for weight loss. A web-based registry
was accessed for the data on 1,343 patients with a starting BMI 25 kg/m
2
. Nonserious and serious adverse events were
reported in 14.2% and 0.15% of patients, respectively. Weight loss in the indicated use (BMI 30-40 kg/m2) was 9.7 ±6.1
kg and 10.0 ±6.1% TBWL. Weight loss in other BMI categories was 8.2 ±5.6 kg or 10.3 ±7.0% total body weight loss for
BMI 25 to 29.9 kg/m2; and 11.6 ±7.8 kg or percent total body weight loss 9.3 ±6.0 for BMI > 40 kg/m2. The authors
concluded that the OBS safe and effective at stimulating weight loss and provides practitioners with another tool to treat
obese patients who have failed other weight loss programs. Limitations included lack of comparison group, the possible
bias of a manufacturer-sponsored study, variation with loss and behavior modification data collection, and lack of data
collection for co-morbidities and metabolic data resulting in inability of data analysis for these areas.
Coffin et al. (2017, included in the Hayes 2021 report above) published findings from their multicenter randomized
controlled trial, in which they compared 6 months of IGB or standard medical care (low-calorie diet, with bimonthly
dietician evaluations) as bridge therapies to laparoscopic gastric bypass in super-obese patients (> 45 kg/m2). The
surgery was performed at 6 months, shortly after removal of the IGB, and assessments were undertaken through 12
Bariatric Surgery
Page 26 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
months. While the BMIs between groups were comparable at baseline, IGBs significantly reduced BMI by 6 months
compared with standard care, with median BMI of 47.9 kg/m2 for IGB patients and 50.7 kg/m2 for control patients (p <
0.001). However, while the implanted IGB was effective on the short term, having the IGB before surgery did not impact
postsurgical outcomes after 12 months (approximately 6 months post-surgery), the groups’ BMIs were not significantly
different at this time point (median BMI:IGB, 38.1 kg/m2 versus standard care, 37.6 kg/m2; p = 0.56). The authors
concluded that IGB insertion before LGBP induced weight loss but did not improve the perioperative outcomes or affect
postoperative weight loss. Limitations of the study included short duration of the IGB intervention, poor recruitment rate, a
higher-than-expected use of ICU facilities, and the poor weight loss in the IGB group.
Nunes et al. (2017) conducted a retrospective review of 2002 patients who underwent an IGB procedure to determine its
effectiveness with different degrees of obesity. A total of 946 patients were lost to follow-up. Overall, 40 (3.78%) had
device removal due to intolerance, and 1016 patients completed the 6-month treatment. The mean weight loss was
18.9%, excess weight loss 60.1% and a BMI reduction of 6.76 points. Six months after removal of the balloon 842 patients
had continued follow-up (82.8%). At this time, weight loss was 19.84%, excess weight loss was 59.49%, and BMI
reduction of 7.06 points. In all groups there was statistical difference between the times T0 and T1 and between T1 and
T2 (p < 0.001). There was no statistical difference between T2 and T3, in any group. The authors concluded that IGB
provided sustained weight loss in patients who remained in dietary follow-up for 1 year. The study is limited by lack of
comparison group and high lost-to-follow up rate. Longer term outcomes with well-designed randomized clinical controlled
trials are needed to further evaluate the IGB.
Saber et al. (2017) conducted a systematic review and meta-analysis to evaluate the efficacy and safety of intragastric
balloon (IGB) treatment. A total of 20 RCTs involving 1,195 participants were identified. Weight loss results before and
after 3 months were analyzed separately. The weight loss results of patients with and without IGB treatment were
compared. A significant effect size was calculated that favored fluid filled IGBs over air-filled IGBs. Flatulence, abdominal
fullness, abdominal pain, abdominal discomfort, and gastric ulcer were significantly more prevalent among IGB patients
than among non-IGB control patients. No mortality was reported from IGB treatment. In the authors’ opinion, IGB
treatment, in addition to lifestyle modification, is an effective short-term modality for weight loss. However, there is not
sufficient evidence confirming its safety or long-term efficacy.
The REDUCE pivotal trial (Ponce et al., 2015, included in the Hayes 2021 report above, and Jung 2020 systematic
review) was a prospective, randomized controlled pivotal trial of a dual intragastric balloon to evaluate the safety and
effectiveness of a dual balloon system plus diet and exercise in the treatment of obesity compared to diet and exercise
alone. Participants (n = 326) with body mass index (BMI) 30-40 kg/m
2
were randomized to endoscopic dual balloon
system (DBS) treatment plus diet and exercise (DUO, n = 187) or sham endoscopy plus diet and exercise alone (DIET, n
= 139). Co-primary endpoints were a between-group comparison of %EWL and DUO subject responder rate, both at 24
weeks. Thereafter DUO patients had the DBS retrieved followed by 24 additional weeks of counseling; DIET patients were
offered DBS treatment. Mean BMI was 35.4. Both primary endpoints were met. DUO weight loss was over twice that of
DIET. DUO patients had significantly greater %EWL at 24 weeks (25.1% intent-to-treat (ITT), 27.9% completed cases
(CC, n = 167) compared with DIET patients (11.3% ITT, p = .004, 12.3% CC, n = 126). DUO patients significantly
exceeded a 35% response rate (49.1% ITT, p < .001, 54.5% CC) for weight loss dichotomized at 25%EWL.
Accommodative symptoms abated rapidly with support and medication. Balloon deflation occurred in 6% without
migrations. Early retrieval for non-ulcer intolerance occurred in 9%. Gastric ulcers were observed; a minor device change
led to significantly reduced ulcer size and frequency (10%). The authors concluded that the dual balloon system was
significantly more effective than diet and exercise in causing weight loss with a low adverse event profile. Additional RCT
with longer follow-up are needed.
Laparoscopic Greater Curvature Plication (LGCP)
While laparoscopic greater curvature plication may appear to be safe for weight loss, additional robust RCTs with
comparison groups and long-term data are needed.
In a 2023 single center retrospective analysis Park and Kim presented the weight loss and revision surgery rate outcomes
of 75 patients following laparoscopic gastric greater curvature plication (LGGCP) surgery. The results showed that 13 out
of 75 patients underwent revision surgery. The main reason for revision was weight regain, however chronic intermittent
GERD, dyspepsia and chronic relapsing melena were also reasons. The mean body weight and BMI at initial LGGCP
surgery were 207 lbs. (± 24) and 35.6 (± 3.9 kg/m2) respectively. Mean nadir body weight after LGGCP was 149 lbs.
13), and BMI was 25.8 (± 2.8 kg/m2). At revision, mean body weight was 196 lbs. (± 25) and BMI was 33.9 (± 4.2 kg/m2).
The results showed that after 5 years, there was weight gain close to pre-surgery levels. The authors concluded that
LGGCP as a primary surgery, results in high rate of weight gain and the need for revisional surgery.
Bariatric Surgery
Page 27 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Doležalova-Kormanova et al. (2017) reported outcomes in a cohort of LGCP patients at 5-year follow-up. Patients with
complete weight data through 5-year follow-up was 86.9%, (212/244). The ANOVA database indicated a significant BMI
reduction out to 2 years (p < 0.001), a plateau at 3 and 4 years, and a moderate but significant BMI increase at 5 years (p
< 0.01). Excess BMI loss at 1, 2, 3, 4, and 5 years was as follows: 50.7 ±9.1%, 61.5 ±8.1%, 60.2 ±7.0%, 58.5 ±7.0%, and
56.8 ±6.3%. At 5 years, 79.2% (168/212) of patients were successful; 20.8% (44/212) experienced a suboptimal weight
outcome; mean weight regain, 9.2%. Cluster analysis identified four distinct LGCP patient profiles. Diabetes improvement
rate was 65.5%. There were 12 reoperations (4.9%): 4 emergency (1.6%) and 8 (3.3%) elective. There was no mortality.
The authors concluded that based on their original cohort and a 56.8% Excess BMI loss and low rate of complications,
LGCP proved to be safe and effective. The findings are limited by lack of comparison group. Additional long-term
outcomes are needed to evaluate LGCP in comparison to other bariatric procedures.
In an 18-month prospective, observational, open-label study, Bužga et al. (2017) reported outcomes of 127 patients; 84
underwent LSG and 43, LGCP. LSG and LGCP were then compared during long-term follow-ups in terms of glycemic
control, hormone and lipid secretion, and changes in body composition. Significant weight-loss and an improved body
composition resulted from either procedure vs. baseline (i.e., pre-surgery), with levels of fasting glucose and glycated
hemoglobin also showing statistically significant reductions (at 3 and 18 months for either surgery). Intergroup
comparisons for glycemic parameters yielded no statistically significant differences. However, a dramatic reduction in
ghrelin was detected following LSG, falling from pre-surgery levels of 140.7 to 69.6 ng/L by 6 months (p < 0.001).
Subsequently, ghrelin levels increased, reaching 107.8 ng/L by month 12. Conversely, after LGCP, a statistically
significant increase in ghrelin was seen, rising from 130.0 ng/L before surgery to 169.0 ng/L by month 12, followed by a
slow decline. The authors concluded that although the data showed good metabolic outcomes following LGCP, this
method was less effective than LSG, possibly due to its preservation of the entire stomach, including secretory regions.
Grubnik et al. (2016) compared two-year outcomes in a European prospective randomized controlled trial comparing
LGCP versus LSG. A total of 54 patients with morbid obesity were allocated either to LGCP group (n = 25) or LSG group
(n = 27). Main exclusion criteria were ASA > III, age > 75 and BMI > 65 kg/m
2
. There were 40 women and 12 men, and
the mean age was 42.6 ±6.8 years (range 35-62). Data on the operation time, complications, hospital stay, BMI loss,
%EWL, loss of appetite and improvement in comorbidities were collected during the follow-up examinations. One year
after surgery, the mean %EWL was 59.5 ±15.4 % in LSG group and 45.8 ±17 % in LGCP group (p > 0.05). After 2 years,
mean %EWL was 78.9 ±20 % in the LSG group and 42.4 ±18 % in the LGCP group (p < 0.01). After 3 years, mean
%EWL was 72.8 ±22 in the LSG group and only 20.5 ±23.9 in the LGCP group (p < 0.01). Loss of feeling of hunger after 2
years was 25 % in LGCP group and 76.9 % in the LSG group (p < 0.05). The comorbidities including diabetes, sleep
apnea and hypertension were markedly improved in both groups after surgery. The authors concluded that the short-term
outcomes demonstrated equal effectiveness of the both procedures, but 2-year follow-up showed that LGCP is not as
effective as LSG as a restrictive procedure for weight loss.
Tang et al. (2015) conducted a meta-analysis to compare LGCP with LSG in terms of efficacy and safety. Eligible studies
included one randomized controlled trial and three non-randomized controlled trials involving 299 patients. The meta-
analysis demonstrated a significantly greater %EWL after LSG than LGCP at the follow-up time points of 3 months (Z =
2.26, p = 0.02), 6 months (Z = 4.49, p < 0.00001), and 12 months (Z = 6.99, p < 0.00001). The difference in the resolution
of diabetes mellitus between these two approaches did not reach statistical significance (p = 0.66). According to the
pooled data, LGCP was associated with more adverse events than was LSG (p = 0.01). The operation time (p = 0.54) and
postoperative hospital stay (p = 0.44) were comparable between the two groups. LGCP is inferior to LSG not only in
terms of providing effective weight loss but also in terms of safety.
Mini-Gastric Bypass (MGB)/Laparoscopic Mini-Gastric Bypass (LMGB)/One-
Anastomosis Gastric Bypass (OAGB)
Currently there is insufficient evidence regarding the long-term effectiveness and safety of mini-gastric bypass for obesity
and weight loss; additional well designed RCTs are needed along with long-term effects, and safety and efficacy results.
In a 2023 systematic review and meta-analysis, Li et al. assessed the efficacy and safety outcomes of one anastomosis
gastric bypass (OAGB) compared to the Roux-en-Y (RNY) procedure in eight randomized controlled trials that comprised
a total of 931 patients. The mean preoperative BMI ranged from 42.6 to 53.5 kg/m
2
. Due to inconsistent outcome
measures being described in each study, the authors performed a meta-analysis using the post operative outcome
measures of BMI, percent of excess weight loss (%EWL), or excess body mass index loss (EBMIL). The results showed
that 6 months after surgery, BMI and %EWL did not show a statistically significant difference. Twelve months post-
surgery, 4 articles showed OAGB resulted in better weight loss than RYGB for %EWL, and two articles showed OAGB
had superior BMI reduction. Two articles reported 5-year outcomes and showed Five years no statistically significant
differences in %EBIML and BMI. Two articles reported intraoperative complications for which there were no statistically
significant differences between the two procedures. Three articles were included in the early postoperative complications
Bariatric Surgery
Page 28 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
OAGB showed fewer complications than RNY, 3 versus 8 serious complications, respectively. There was inconsistent
reporting of obesity related illnesses across the studies, but articles that did report them, included diabetes, hypertension,
hyperlipidemia, and gastroesophageal reflux disease (GERD), and all showed a high rate of remission. The authors
concluded that OAGB is not inferior to RNY in terms of weight loss and remission of comorbidities during the first 2 years
post operatively but may have a higher incidence of malnutrition. Additional large sample and long-term randomized
controlled trials are needed to verify these findings. (Eskandaros 2021, and Musella 2017 previously cited in this policy
were included in this review)
Parmar et al. (2020) evaluated the role of One Anastomosis/Mini Gastric Bypass (OAGB-MGB) as a revisional/secondary
procedure in patients who needed revisional bariatric surgery (RBS). A total of 17 studies were included in this systematic
review with a total of 1075 patients. The mean age was 43 years and 75% were female. The follow-up ranged from 6 to
60 months with a mean of 29 months. The following identifies the breakout of primary procedures performed: LAGB - 569
patients, SG 397 patients, VBG 105 patients, and lap gastric plication - 5 patients. The most common reason for RBS
was poor response in 81%, followed by gastric band failure in almost 36% of patients. The mean BMI prior to RBS was
41.6 kg/m
2
. Following the OAGB-MGB procedure, the mean %EWL was 50.8% at 6 months, 65.2% at one year, 68.5% at
24 months and 71.6% at 5 years. The author’s conclusion suggests that OAGB-MGB is a safe and an effective choice for
revisional surgery, however randomized studies and large prospective studies with long term follow-up are needed to
validate these findings. Limitations included lack of comparison group or RCTs in analysis along with race and ethnicity
differences which may have impacted the patient’s eating habits, education, compliance, and expectations.
In a comparative effectiveness review from Hayes (2019) for primary bariatric surgery, the mini gastric bypass-one
anastomosis gastric bypass (MGB-OAGB) was compared to RYGB and LSG separately. Data from two systematic
reviews and 4 RCTs suggest an overall increase in percentage of weight loss with the MGB-OAGB procedure when
compared to RYGB and LSG. The evidence also suggested MGB-OAGB may have a positive impact on resolution of T2D
and HTN. However, additional long-term follow-up is warranted for further research on long-term follow-up, complications,
adverse effects, and impact on nutrition.
Carbajo et al. (2018) conducted a prospective, single-center case series to analyze weight evolution in 100 patients from
the first pre-surgery appointment through a 2-year follow-up after one anastomosis gastric bypass. No surgical
complications were observed in the patients studied. The patients’ mean pre-surgery BMI was 42.61±6.66 kg/m
2
. Greatest
weight loss was observed at 12 months post-surgery (68.56 ±13.10 kg). Relative weight loss showed significant positive
correlation, with greatest weight loss at 12 months and %excess BMI loss > 50% achieved from the 3-month follow-up in
92.46% of patients.
The authors reported that in this series, 48% of patients had normal weight (BMI > 18.5 < 25 kg/m
2
) at
24 months post-surgery. A limitation of this study is the lack of comparison group, short-term follow-up of the sample
selected; patient evolution should be completed with medium- and long-term data.
In a prospective, case series of 150 morbidly obese patients who underwent laparoscopic OAGB, lipid profiles were
evaluated preoperatively and at different intervals during a 2-year follow-up. The authors (Carbajo et al., 2017) reported a
mean weight loss of 48.85 kg ±15.64 and mean %EWL of 71.87 ±13.41. kg. Total cholesterol and low-density lipoprotein
(LDL) levels significantly decreased, and high-density lipoprotein (HDL) levels significantly increased which the authors
believe translate into theoretical relevant cardiovascular risk benefits. The findings are limited by lack of comparison
group. Long-term randomized studies are needed to fully evaluate the impact of this procedure.
Lessing et al. (2017) conducted a retrospective analysis of all patients (n = 407) who underwent OAGB, reporting an
average excess weight loss 1 year following surgery as 88.9 ±27.3 and 72.8 ±43.5% in patients that underwent primary
and revision OAGB, respectively. Study limitations include lack of comparison group and single center data.
Wang et al. (2017) conducted a systematic review and meta-analysis to compare the safety and efficacy between
laparoscopic mini-gastric bypass (MGB) and laparoscopic SG. Thirteen studies met the inclusion criteria of comparative
studies between MGB and SG; patients were adults, with age ranging from 20 to 70 years old; at least one of the following
endpoints was included: operation time, mortality, overall early complications, specific early complications, overall late
complications, specific late complications, hospital stay, revision rate, remission rate of comorbidities, 1-year %EWL or 5-
year %EWL. The authors observed that patients receiving mini-gastric bypass had more advantageous indexes than
patients receiving sleeve gastrectomy, such as higher 1-year EWL% (excess weight loss), higher 5-year EWL%, higher
T2D remission rate, higher hypertension remission rate, higher OSA remission rate, lower osteoarthritis remission rate,
lower leakage rate, lower overall late complications rate, higher ulcer rate, lower GERD rate, shorter hospital stay and
lower revision rate. No significant statistical difference was observed on overall early complications rate, bleed rate,
vomiting rate, anemia rate, and operation time between MGB and SG. In their opinion, due to the biased data, small
sample size and short follow-up time, the results of this review may be unreliable. RCTs with larger samples sizes are
Bariatric Surgery
Page 29 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
needed to compare the effectiveness and safety between MGB and SG. (Publications by Kansou 2016 and Plamper
2017, which were previously cited in this policy, are included in this systematic review).
Piazza et al. (2015) reported their experience with laparoscopic mini-gastric bypass (LMGB) as a revisional procedure for
failed primary LAGB. From June 2007 to November 2012, 48 patients, who had undergone LAGB, underwent revisional
surgery to LMGB. The revisions to a MGB were completed laparoscopically in all cases except in four, when the MGB
was deferred because of gastric tube damage. Mean age was 38 years (range 20-59), and BMI was 43.4 ±4.2 kg/m
2
; 82
% of patients were females. Revision was performed after a mean of 28.6 months. The mean hospital stay was 3.25 days.
Within 60 days of the MGB, mortality and morbidity were nil. They observed a significant difference in mean BMI after 6
months' follow-up (p < 0.001). Diabetes remission was observed in 88 % of patients, apnea remission in 66 %, and
hypertension remission in 66 % after LMGB (p < 0.001). Moreover, four patients with GERD reported symptom resolution.
All LAGB patients had positive outcomes after the conversion to MGB, with a mean gain of 1.7 points in the bariatric
analysis and reporting outcome system questionnaire. The authors suggest that based on their results, LMGB is a safe,
feasible, effective and easy-to-perform revisional procedure for failed LAGB. The findings are however limited by lack of
comparison group.
Single-Anastomosis Duodenal-Ileal Switch (SADI-S/SADI/SADS)
There is insufficient evidence regarding the safety and efficacy of the single-anastomosis duodenal switch (SADS) for
obesity; additional robust RCTs with comparison groups along with long-term results are needed. Several clinical trials are
in progress for the single-anastomosis duodenal switch; information can be found at https://www.clinicaltrials.gov.
In a 2021 retrospective cohort study, Iranmanesh et al. compared short- and medium-term outcomes between the
standard double-anastomosis duodenal switch (DADS), and single-anastomosis duodenal switch (SADS). Data of 107
patients was collected in the Ontario Bariatric Registry from a Canadian bariatric center of excellence between 2010 and
2019, with the primary outcome measurement weight loss at 1- and 2-years post-surgery. Short-term secondary
outcomes included operative times, intra- and early postoperative complications, hospital LOS, and 30-day readmissions.
Medium-term secondary outcomes included late postoperative complications as well as nutritional deficiencies and
persistent diarrhea at 1- and 2-years post-surgery. Of the 107 patients, 25 received SADS surgery and 82 received
DADS. Follow up data was available for 59 patients at one year, and 47 after 2 years. The results showed similar %TWL
at 1 year (23.6 versus 26.2) and 2 years (24.8 versus 30.2,) after surgery. Short- and medium-term outcomes were similar
between groups. This study is limited by a small number of patients receiving the SADS procedure and large rate of lost-
to-follow-up. Additional high-quality studies with longer follow up are necessary to validate these retrospective findings.
Pereira et al. (2021) conducted a prospective, observational cohort study of 112 patients receiving SADS or BPD/DS.
Primary endpoints were BMI and TWL, and secondary endpoints included remission of obesity related disorders (T2D,
hypertension and dyslipidemia), nutritional deficiencies and post-operative complications. 83 patients received SADS and
29 BPD/DS. There were no statistically significant differences between groups’ demographic characteristics or clinical
features, except for baseline weight and BMI, which were significantly higher in the BPD/DS group. Follow up times for
SADS and BPD/DS ranged from an average of 40 months to 23 months, respectively. The results showed no significant
differences in BMI and percent excess BMI loss (%EBMIL) between the groups, although the percentages of total weight
loss observed from 12, 24, and 36months were significantly higher after BPD/DS. Obesity related comorbidities resolved
numerically better in the BPD/DS group than the SADS group, but it was not statistically significant. Nutritional status was
not consistently significant between the two procedures, and no differences were observed in surgical complications.
Operative time and hospital stay was shorter for the SADS group. The authors concluded SADS is a simpler technique
and shows similar results to BPD/DS. They acknowledged several limitations, including that there was a considerable
numerical imbalance between the two groups, and the number of patients with a follow-up was small. Large-scale,
randomized controlled clinical trials with long-term data are needed to confirm these results.
In a Medtronic funded study, Cottam et al. (2020) evaluated weight loss and one-year nutritional outcomes of the SADS
procedure. 120 patients at six different sites were enrolled; participant inclusion criteria included BMI of 35-40 kg/m2 with
one obesity related comorbidity or a BMI of 40-60 kg/m2 with no related comorbidity. Weight loss, comorbidities, quality of
life, and AEs were followed post-procedure for 12 months. The authors found SADS to be an effective weight loss
operation and the ability to reduce comorbid conditions, particularly diabetes. Limitations included lack of comparative
cohort, patient loss to follow up and lack of long-term results for efficacy.
In a retrospective cohort study, Surve et al. (2017) compared biliopancreatic diversion with duodenal switch (BPD-DS)
with single anastomosis duodenal switch (SIPS-stomach intestinal pylorus sparing surgery) at a single institution with two-
year follow-up. One-hundred eighty-two patients received either a BPD-DS (n = 62) or SIPS (n = 120) procedure. BPD-DS
and SIPS had weight loss at 3 months that were not statistically significantly different but %EWL was more with BPD-DS
than SIPS at 6, 9, 12, 18, and 24 months. Patient lost a mean BMI of 23.3 (follow-up: 69%) and 20.3 kg/m2 (follow-up:
Bariatric Surgery
Page 30 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
71%) at 2 years from the BPD-DS and SIPS surgery, respectively. However, patients who had undergone SIPS procedure
had significantly shorter operative time, shorter length of stay, fewer perioperative and postoperative complications than
BPD-DS (p < 0.001). There was no statistical difference between 2 groups for postoperative nutritional data such as
vitamins D, B1, B12, serum calcium, fasting blood glucose, glycosylated hemoglobin (HbA1C), insulin, serum albumin,
serum total protein, and lipid panel. The authors noted that as the BPD-DS procedures were done prior to SIPS, learning
curve and experience may account for the post-operative complications. RCTs with larger patient populations and longer
follow-up periods are needed to evaluate the SIPS procedure.
Cottam et al. (2016) conducted a retrospective matched cohort analysis to compare RYGB with SADS with 18-month
follow-up. One-hundred eight patients received either a RYGB (n = 54) or SADS (n = 54). Regression analysis was used
to compare weight loss outcomes as measured by BMI and weight loss percentages. The results failed to show
statistically significant differences between the two procedures on weight loss at 18 months (39.6 vs 41 % weight loss,
respectively). However, there were significantly more nausea complaints (26 vs 5), diagnostic endoscopies (EGD) (21 vs
3) and ulcers (6 vs 0) with the RYGB than the SADS. The 2-year outcomes for this same patient cohort had similar results
(Cottam et al., 2017). RCTs with larger patient populations and longer follow-up periods are needed to validate these
findings.
Stomach Aspiration Therapy
Currently there is insufficient evidence regarding the safety and efficacy of stomach aspiration therefore additional robust
RCTs with comparison groups are needed along with long-term results.
A 2021 ECRI clinical evidence assessment on AspireAssist Gastric Aspiration Port (Aspire Bariatrics, Inc.) noted that
evidence is somewhat favorable for AspireAssist when adding to lifestyle modification. It was noted to improve weight loss
at 1 year which was maintained at up to 4 years, however, these findings are based on low-quality evidence from 2
systematic reviews and 1 single-arm extension of an RCT. It is unknown if AspireAssist therapy contributes to abnormal
eating behaviors as only one single-arm extension of RCT reported too few events. Evidence limitations included risk of
bias in most studies included in the systematic reviews due to small study size, lack of control group, or both. Additional
larger RCTs are needed to confirm findings, especially in the long term, as well as to compare AspireAssist with other
minimally invasive treatments.
Jirapinyo et al. (2020) conducted a systematic review and meta-analysis of 5 studies with a total of 590 patients to assess
the outcomes of aspiration therapy (AT) (AspireAssist
®
) on obesity related comorbidities at one year follow up.
Comorbidities included hypertension, hyperlipidemia, T2D, and NAFLD. Secondary outcomes were the amount of weight
loss up to four years post operatively, and pooled serious adverse events (SAEs). The results showed after one year
hypertension, hyperlipidemia, HbA1C, and NAFLD significantly improved. Weight loss at one year was 17% TWL (296
patients), 2 years 18.3% (174 patients), 3 years 18.6% (88 patients), and 4 years 18.6% (27 patients). The pooled SAEs
rate was 4.1% and included buried bumper, peritonitis severe abdominal pain, abdominal pain secondary to pre-pyloric
ulcer and device malfunction requiring A tube replacement. Two studies reported a rate of persistent fistula following A-
tube removal. The authors concluded that at 1 year AT resulted in significant improvement in metabolic function
parameters and 4 years, patients maintained their significant weight loss of 18.6% of their baseline weight, meeting the
definition of successful weight loss maintenance, and may improve access to treatment in obese patients with
concomitant comorbidities. The authors acknowledge the limitations of this study. The number of studies is small (to
account for this, conference abstracts that met the a priori inclusion criteria were included in the analysis), and most of
them were retrospective and observational in nature. Larger, high-quality studies with longer follow-up are required to
validate these findings. (Publications by Sullivan 2013, Thompson 2017, and Nyström 2018, which were previously cited
in this policy, are included in this systematic review).
In the post study of the PATHWAY Trial, Thompson et al. (2019) provide 4-year outcomes of the AT patients from the
initial trial. 58 participants were enrolled in the follow up study; of these 55 had achieved at least 10% TWL at the end of
the first year. Of the 58 patients who enrolled in the follow-up study, 15, 21, and 7 patients elected to have the A tube
removed between years 1 and 2, 2 and 3, and 3 and 4, respectively, thus withdrawing from the study but no loss to follow-
up. The 43 patients who withdrew from the study between years 2 and 4, 25 (58.1%) achieved at least 10% TWL. The
mean %EWL of AT participants at years 1, 2, 3, and 4 was 37.1 ±27.6 (n/n = 81/110), 40.8 ±25.3 (n/n = 42/55), 44.7 ±29.7
(n/n = 22/55), and 50.8 ±31.9 (n = 15/55), respectively. The clinical success rate for patients participating in the follow-up
study was 40/58 (69%) at 4 years from A-tube placement. The authors concluded the AT is a safe and effective
intervention for people with class II and III obesity and can achieve weight loss along with improvement of quality of life.
Limitations of this study are the relatively small number of participants by the fourth year, participant commitment and the
absence of weight loss data after A-tube removal. Additionally, the findings are limited by the design that only allowed
continued follow-up of participants maintained at least 10% TWL from baseline at each year end and lack of comparison
group for the long-term.
Bariatric Surgery
Page 31 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Norén and Forssel (2016) reported 1 and 2-year outcomes from their prospective observational study of 25 obese
subjects to evaluate weight reduction and safety of AT with AspireAssist
. Twenty of the original 25 subjects completed
the initial 1-year treatment. These 20 subjects lost mean 54% of their excess weight. At 2 years, 15 subjects had lost
mean 61% of their excess weight. This weight loss surpassed our expectation and is nearly at the level of gastric bypass
procedure and other major abdominal surgery for obesity. The subjects reported improved quality of life during treatment.
There was neither mortality nor any event more severe than grade III-a according to Clavien-Dindo grading system.
Limitation of this study is the combination of AT and cognitive behavioral therapy (CBT) without any control group. Long
term patency is still unknown.
Forssell and Norén (2015) conducted an observational study of 25 obese patients (BMI 39.8 ±0.9 kg/m2) who after
following a very low-calorie diet for 4 weeks had the AspireAssist gastrostomy tube placed. A low-profile valve was
installed 14 days later, and aspiration of gastric contents was performed approximately 20 minutes after meals three times
per day. Cognitive behavioral therapy was also started. At 6 months, mean weight lost was 16.5 ±7.8 kg in the 22 subjects
who completed 26 weeks of therapy (p = 0.001). The mean percentage excess weight lost was 40.8 ±19.8 % (p = 0.001).
Two subjects were hospitalized for complications: one subject for pain after gastrostomy tube placement, which was
treated with analgesics, and another because of an aseptic intra-abdominal fluid collection 1 day after gastrostomy tube
placement. No clinically significant changes in serum potassium or other electrolytes occurred. The authors concluded
that the results suggest the potential of the AspireAssist as an attractive therapeutic device for obese patients. Further
research with randomized controlled trials is needed to validate these findings.
Transoral Endoscopic Surgery [Including Transpyloric Shuttle
®
(TPS) Device]
The evidence for transoral endoscopic surgery for bariatric surgery is limited; additional studies including RCTs, long-term
data including the safety and efficacy of the procedure are warranted.
In a 2023 retrospective study, Gudur et al. analyzed over 600, 000 patients in the Metabolic and Bariatric Surgery
Accreditation and Quality Improvement Program database and compared short term (30 days) adverse events (AEs),
readmissions, reoperations, and reinterventions in patients that underwent endoscopic sleeve gastroplasty (ESG)
compared to sleeve gastrectomy (SG). A total of 6054 patients underwent ESG, and 597463 SG. The results showed that
there was no significant difference in major AEs, but patients undergoing ESG had more readmissions, reoperations, and
reinterventions. An additional analysis showed that chronic steroid use, renal insufficiency, and anticoagulation therapy
contributed the most to the AEs in both groups. Race did not impact AEs after ESG, with an increased risk of AEs
identified for black patients after SG. This retrospective study is limited by a very short follow up period. The authors
concluded that further prospective long-term evaluations of ESG versus SG with regards to safety and efficacy are
needed.
In a brief from ECRI (2019), the evidence for the Transpyloric Shuttle
®
(TPS) device is inconclusive. The evidence is
limited indicating longer-term follow-up data is warranted. The RCT reviewed appeared to have a low risk of bias but
results from a single trial were not conclusive and need independent confirmation in another controlled trial. The case
series had a very high risk of bias due to small sample size, lack of a control group and randomization, and blinding. Both
the RCT and case series report relatively short follow-up.
In a prospective, multicenter, single-arm, feasibility trial, Sandler et al. (2018) evaluate 32 obese subjects with a trans-oral
endoscopic gastrointestinal bypass device. The device is a cuff attached to the distal esophagus by transmural anchors
and connected to a 120-cm sleeve diverting undigested nutrients to the jejunum. Baseline data collected included
bodyweight, vital signs, AEs, medications, HbA1c, fasting glucose, and lipids in addition to follow-up visits. The device
status was endoscopically assessed every 6 months. At 12 months, the 32 subjects had lost an average of 44.8% of
excess body weight, 17.6% of total body weight, 20.8 kg, and 7.5 BMI points. The authors concluded this study
demonstrated the feasibility, safety, and efficacy of a fully trans-oral gastrointestinal bypass implant and that this
endoscopic device may provide a valuable addition to the available treatment for the management of morbid obesity.
However, this study is limited by lack of comparison group, small sample size and short-term follow-up.
Marinos et al. (2014) conducted a prospective, open-label, nonrandomized, single-center investigational clinical trial
performed to evaluate the safety and efficacy of the transpyloric shuttle (TPS) device. The study enrolled twenty patients
meeting the criteria in 2 cohorts with treatment periods of 3 and 6 months. Patients were required to be ≥ 18 and ≤ 55
years of age with a BMI between 30 and 50 kg/m2. Before device placement, patients were provided with nutritional
guidelines for a low-calorie diet and no additional dietary counseling was given after the initial consultation. Patients were
placed under general anesthesia and the devices were deployed and retrieved with no complications. All 20 patients
enrolled in the study had lost weight at the time of device removal. Both the 3- and 6-month patients had statistically
significant improvements to the overall IWQOL-Lite score that exceeded the 7.7- to 12-point threshold to define a clinical
change. All but two patients completed the planned treatment period; both patients had the device removed due to
Bariatric Surgery
Page 32 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
complaints of epigastric pain. Limitations of the study were small participant size and short treatment duration. The
authors concluded the TPS is a promising technology that has potential to benefit obese patients seeking to lose weight.
Eid et al. (2014) conducted a prospective, single-center, randomized, single-blinded study from July 2009 through
February 2011, to investigate the safety and effectiveness of endoscopic gastric plication with the StomaphyX device vs a
sham procedure for revisional surgery in RYGB patients to reduce regained weight. Enrollment was closed prematurely
because preliminary results indicated failure to achieve the primary efficacy end point in at least 50% of StomaphyX-
treated patients. One-year follow-up was completed by 45 patients treated with StomaphyX and 29 patients in the sham
treatment group. Primary efficacy outcome was achieved by 22.2% (10) with StomaphyX vs 3.4% (1) with the sham
procedure (p < 0.01). Patients undergoing StomaphyX treatment experienced significantly greater reduction in weight and
BMI at 3, 6, and 12 months (p 0.05). There was one causally related adverse event with StomaphyX, that required
laparoscopic exploration and repair.
A case series by Mullady et al. (2009) evaluated 20 patients who underwent restorative obesity surgery, endoluminal
(ROSE) procedure due to weight regain post gastric bypass, with a confirmed dilated pouch and gastrojejunal
anastomosis (GJA) on endoscopy. Seventeen of 20 (85%) patients had an average reduction in stoma diameter of 16 mm
(65% reduction) and an average reduction in pouch length of 2.5 cm (36% reduction). The mean weight loss in successful
cases was 8.8 kg at 3 months. The authors concluded that the ROSE procedure is effective in reducing not only the size
of the gastrojejunal anastomosis but also the gastric pouch and may provide an endoscopic alternative for weight regain
in gastric bypass patients. This study is limited by lack of comparison group, small sample size and short-term follow-up.
Endoscopic Sleeve Gastroplasty (OverStitch)
There is insufficient quality evidence regarding the safety and efficacy of endoscopic sleeve gastroplasty for obesity.
Future studies including RCTs are needed to assess the safety and efficacy of this procedure along with long-term results.
Current evidence in an evolving technology report from Hayes (2022) identified four comparative studies and two
systematic reviews which revealed minimal support for endoscopic sleeve gastroplasty (ESG) with the OverStitch device.
Even though the OverStitch device is associated with clinically significant weight loss and fewer AEs, studies did not
suggest the weight loss was more beneficial than an LSG.
Abu Dayyeh et al. (2022) conducted a randomized clinical trial to explore the safety and efficacy of endoscopic sleeve
gastroplasty (ESG) with lifestyle modifications compared to lifestyle modification alone for the treatment of Class 1 and 2
obesity. Inclusion criteria was aged 21-65 with a BMI of 30 to less than 40 with a history of failure with non-surgical weight
loss methods, and who agreed to comply with lifelong dietary restrictions required by this procedure. The primary outcome
on efficacy was %EWL at 52 weeks. Secondary efficacy outcomes included proportion of patients with 25% or more EWL,
% of total weight loss, and the proportion of patients with 5% or more and 10% or more of total weight loss. The effect of
ESG on obesity related comorbidities and safety were also assessed. Seventy-seven participants were randomized to the
ESG plus moderate-intensity lifestyle modifications (ESG group), and 110 to the moderate-intensity lifestyle modifications
alone (control group). During the first year, 12 follow up visits were completed at weeks 1 and 4, and then every 4 weeks
until the 52-week visit. The results showed ESG with lifestyle modifications, compared with lifestyle modifications alone,
resulted in significant improvements in terms of weight loss, and metabolic comorbidities with no GERD incidence as seen
with other bariatric surgeries. Adverse events included gastrointestinal symptoms such as pain, heartburn, nausea and
vomiting which is not unexpected when acclimating post procedure. Three participants had a Clavien-Dindo grade 3
device or procedure related adverse event requiring intervention and included abscess, GI bleeding and one case of
malnutrition requiring reversal of the ESG. The authors concluded that as a minimally invasive alternative to surgical
sleeve gastrectomy, ESG is a safe and effective option for individuals that prefer a non-surgical option. This study is
limited by the impact of the COVID-19 pandemic on study follow-up and participant retention, as well as a small number of
participants.
Singh (2020) conducted a systematic review and found eight studies addressing the OverStitch
device which included a
total of 1,859 patients. Studies were all observational and included single center and multicenter experiences. Primary
outcomes measured were %TWL), %EWL, and SAE. The authors found the pooled mean %TWL at 6, 12, and 24 months
was 14.86, 16.43, and 20.01. Similarly, %EWL at 6, 12, and 24 months was 55.75, 61.84, and 60.40. The incidence of
SAE was 2.26%, and no mortality was reported. Gastrointestinal bleeding was the most common documented SAE and
was usually managed conservatively with packed red blood cell transfusion. Based on the analysis, the authors concluded
that ESG is a promising technique with effective weight loss outcomes. Limitations included lack of controlled studies, lack
of standardization definition for SAE and lack of long-term follow up data. (Publication by Lopez-Hava 2017, which was
previously cited in this policy, was included in this systematic review).
Bariatric Surgery
Page 33 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Hedjoudje et al. (2020) conducted a systematic review and meta-analysis from eight studies which included 1,772 patients
that underwent ESG. Primary outcome measurements included relative weight loss, decrease in BMI and relative
estimated weight loss. Serious adverse events were reported in all studies with an occurrence of 2.2% and included 18
patients with pain or nausea that required hospitalization, 9 patients that experience upper GI bleeding, 8 patients with
perigastric leak or collection, one patient experienced pneumoperitoneum and one patient had a pulmonary embolism.
The authors found the data suggested ESG gave way to significant sustained weight loss and safety. Patients had a BMI
decrease of 5.6 kg/m
2
, mean TBWL was 15.1% and relative EWL of 57.7%. These results appear to be sustained through
18-24 months of follow-up. Limitations included lack of control group, large loss to follow-up, lack of reporting for mild
adverse events and lack of long-term outcomes; future studies are warranted.
Neto et al. (2020) evaluated 233 patients that underwent ESG between April 2017 and December 2018. The ESG
procedure was performed using the OverStitch
device. The authors found average weight loss was approximately 17%
at six months and 19% at 12 months. The short-term results suggest that ESG is safe and effective, however additional
studies are warranted.
Vagus Nerve Blocking
Currently there is insufficient quality evidence supporting the long-tern effectiveness of vagus nerve blocking for obesity
treatment; additional robust studies including randomization are warranted.
Apovian et al. (2017) reported the two-year outcomes from the ReCharge study among participants initially randomized to
an active intervention. At 24 months, 123 (76%) vBloc participants remained in the trial. Participants who presented at 24
months (n = 103) had a mean EWL of 21% (8% TWL); 58% of participants had ≥ 5% TWL and 34% had ≥ 10% TWL.
Among the subset of participants with abnormal preoperative values, significant improvements were observed in mean
LDL (-16 mg/dL) and HDL cholesterol (+4 mg/dL), triglycerides (-46 mg/dL), HbA1c (-0.3%), and systolic (-11 mmHg) and
diastolic blood pressures (-10 mmHg). QOL measures were significantly improved. Heartburn/dyspepsia and implant site
pain were the most frequently reported AEs. The primary related serious AE rate was 4.3%. The findings are limited by
lack of comparison group.
Morton et al. (2016) reported 12-month outcomes from the ReCharge study. Fifty-three participants were randomized to
vBloc and 31 to sham. Qualifying obesity-related comorbidities included dyslipidemia (73%), hypertension (58%), sleep
apnea (33%), and T2D (8%). The vBloc group achieved a %EWL of 33% (11% %TWL) compared to 19% EWL (6% TWL)
with sham at 12 months (treatment difference 14 percentage points, 95% CI, 7-22; p < 0.0001). Common AEs of vBloc
through 12 months were heartburn/dyspepsia and implant site pain; the majority of events were reported as mild or
moderate. The authors concluded that vBloc therapy resulted in significantly greater weight loss than the sham control
among participants with moderate obesity and comorbidities, and with a well-tolerated safety profile. Longer-term
outcomes are needed to demonstrate the continued durability of this procedure.
Shikora et al. (2016) reported two-year outcomes from the VBLOC DM2 study, a prospective, case series of 28 subjects
with T2D and BMI between 30 and 40 kg/m
2
who underwent a VBLOC procedure. At 24 months, the mean percentage of
EWL was 22% (95% CI, 15 to 28, p < 0.0001) or 7.0% TWL (95% CI, 5.0 to 9.0, p < 0.0001). Hemoglobin A1c decreased
by 0.6 percentage points (95% CI, 0.2 to 1.0, p = 0.0026) on average from 7.8% at baseline. Fasting plasma glucose
declined by 15 mg/dL (95% CI, 0 to 29, p = 0.0564) on average from 151 mg/dL at baseline. Among subjects who were
hypertensive at baseline, systolic blood pressure declined 10 mmHg (95% CI, 2 to 19, p = 0.02), diastolic blood pressure
declined by 6 mmHg (95% CI, 0 to 12, p = 0.0423), and mean arterial pressure declined 7 mmHg (95% CI, 2 to 13, p =
0.014). Waist circumference was significantly reduced by 7 cm (95% CI, 4 to 10, p < 0.0001) from a baseline of 120 cm.
The most common AEs were mild or moderate heartburn, implant site pain, and constipation. The authors concluded that
improvements in obesity and glycemic control were largely sustained after 2 years of treatment with VBLOC therapy with
a well-tolerated risk profile. The findings are limited by lack of comparison group. Randomized controlled studies with
larger patient populations are needed to validate these findings.
The ReCharge pivotal study, sponsored by the manufacturer, (Ikramuddin et al., 2014), was a prospective, randomized,
double-blind, sham-controlled, multi-center trial to evaluate the safety and effectiveness of the Maestro system in treating
obesity. The trial enrolled subjects who had a BMI 40-45 kg/m
2
or a BMI 35-39.9 kg/m
2
with at least one obesity-related
co-morbid condition, and who had failed a more conservative weight reduction alternative. A total of 239 subjects were
enrolled at 10 investigational sites; 162 subjects were randomized to the device group, and 77 were randomized to the
sham control group. Subjects randomized to the sham control group underwent a surgical procedure consisting of
anesthesia, implantation of a nonfunctional neuroregulator, and the same number of incisions an investigator would use
during the laparoscopic placement of the leads. The study authors noted that the trial met its primary safety endpoint and
helped more than half of patients lose at least 20% of their excess weight. The use of vagal nerve block therapy
compared with a sham control device did not meet either of the prespecified coprimary efficacy objectives which were to
Bariatric Surgery
Page 34 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
determine whether the vagal nerve block was superior in mean percentage excess weight loss to sham by a 10-point
margin with at least 55% of patients in the vagal block group achieving a 20% loss and 45% achieving a 25% loss.
Gastrointestinal Liner (EndoBarrier
®
)
Currently there is insufficient evidence regarding the effectiveness and safety of gastrointestinal liners for obesity and
weight loss; additional well designed RCTs are needed along with long-term effects, and safety and efficacy results.
Several clinical trials are in progress for the Endobarrier
®
device; information can be found at https://www.clinicaltrials.gov.
Ruban et al. (2022) conducted an RCT to study the clinical efficacy and safety of the duodenal-jejunal bypass liner
(DJBL). Participants aged 18 to 65 years, with a BMI of 30 to 50 kg/m2 and confirmed diagnosis of T2D for at least 1 year
with inadequate glycemic control and on glucose-lowering medications were included in the trial. 170 patients were
originally selected but due to several participants dropping out, 55 and 58 patients (DJBL and control arms, respectively)
were included in the primary analysis at one year and 58 and 51 patients were included at year two. All participants
received dietary and physical activity counselling. The primary outcome was to achieve an HbA1c reduction of 20% at 12
months post intervention. Secondary outcomes included lowered blood pressure, and a reduction in total body weight loss
and the number of medications taken. The authors found that while the addition of the DJBL resulted in superior weight
loss and improvement in cardiovascular risk factors, it did not make a significant impact on the patients’ HbA1c. The
findings are limited by the open-label design of the study and large loss to follow up that could have introduced biases.
Quezada et al. (2018) conducted a single-arm, open-label, case series to evaluate the safety and efficacy of
endoscopically placed DJBL over a 3-year period. Of 80 patients enrolled in the study, (age: 35 ±10 years; 69% female;
weight: 109 ±17 kg; BMI: 42 ±5.4 kg/m
2
), 72 AEs were observed in 55 patients (68%). Nine subjects required a prolonged
hospital stay and three subjects required major interventions. At 52 weeks (71 patients), 104 weeks (40 patients), and 156
weeks (11 patients), the mean %EWL were 44 ±16, 40 ±22, and 39 ±20, respectively (p < 0.001). This study shows
significant and sustained weight loss after 3 years of treatment with the new DJBL. However, the high frequency and
severity of AEs preclude the use of this prototype for periods longer than 1 year.
Forner et al. (2017) evaluated the outcomes of 114 obese patients treated with a DJBL. Mean total body weight change
from baseline was 12.0 kg (SD 8.5 kg, p < 0.001). Over an average of 51 weeks, the mean %TWL was 10.5% (SD 7.3%).
Mean HbA1c was not significantly improved, but of 10 patients on insulin, 4 ceased insulin and 4 reduced insulin dosages.
There was a significant decrease in hemoglobin and total cholesterol and a significant increase in serum alkaline
phosphatase. Seventy-four percent of patients experienced at least one AE, some of them serious including 6 device
obstructions, 5 gastrointestinal hemorrhages, 2 liver abscesses, and 1 acute pancreatitis. Seventy-four percent of patients
experienced weight gain after removal with a mean 4.5 ±6.1 kg (p < 0.0001) within the first 6 months after explanation.
The authors conclude that the DJBL provides significant but highly variable weight loss, and variable glycemic control.
Most patients experienced an adverse event and most regained significant weight after device removal. In addition, the
authors observed that major adverse events can occur, including the potentially life-threatening complications of hepatic
abscess and gastrointestinal hemorrhage. The findings are limited by lack of comparison group. Further studies are
needed to determine the long-term safety and efficacy of this procedure.
In a retrospective review, Betzel et al. (2017) evaluated the efficacy and safety profile of the DJBL. Inclusion criteria for
treatment with a DJBL were age 18-70 years, BMI 28-45 kg/m2, and T2D with a HbA1c > 48 mmol/mol. Primary outcomes
were changes in HbA1c and body weight. Secondary outcomes included changes in blood pressure, lipids, and anti-
diabetic medication. Predictive factors for success of treatment with the DJBL were determined. The authors reported that
185 out of 198 patients successfully underwent a DJBL implantation procedure, with an intended implantation time of 12
months. In these 185 patients, body weight decreased by 12.8 ±8.0 kg (total body weight loss of 11.9 ±6.9%, p < 0.001),
HbA1c decreased from 67 to 61 mmol/mol (p < 0.001) despite a reduction in anti-diabetic medication, and blood pressure
and serum lipid levels all decreased. In total, 57 (31%) DJBLs were explanted early after a median duration of 33 weeks.
AE occurred in 17% of patients. C-peptide ≥ 1.0 nmol/L and body weight ≥ 107 kg at screening were independent
predictive factors for success. The authors concluded that treatment with the DJBL in patients with T2D and obesity
resulted in improvement in glucose control, a reduction in anti-diabetic medication, and significant weight loss. The largest
changes are observed within the first 3-6 months. Initial C-peptide levels and body weight may help to select patients with
the greatest chance of success. The findings are limited by lack of comparison group.
Vilarrasa et al. (2017) evaluated the efficacy and safety of Endobarrier
®
in grade 1 obese patients with T2D and poor
metabolic control and the role of gastro-intestinal hormone changes on the metabolic outcomes. Twenty-one patients
aged 54.1±9.5 years, diabetes duration 14.8 ±8.5 years, BMI 33.4 ±1.9 kg/m2, and HbA1c 9.1 ±1.3 %, under insulin
therapy, were implanted with Endobarrier
®
. Fasting concentrations of PYY, ghrelin and glucagon, and AUC for GLP-1
after a standard meal test were determined prior to and at months 1 and 12 after implantation. They found that the
Bariatric Surgery
Page 35 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Endobarrier
®
in in this subset of patients is associated with significant weight decrease and moderate reduction in HbA1c
at month 12. Longer term outcome data is needed, and the findings are limited by lack of comparison group.
In a systematic review and meta-analysis, Rohde et al. (2016) evaluated the efficacy and safety of the DJBS. Five RCTs
(235 subjects) and 10 observational studies (211 subjects) were included. The risk of bias was evaluated as high in all
studies. The mean BMI ranged from 30 to 49.2 kg/m2 and 10-100% of the subjects had T2D. Meta-analysis showed that
the DJBS was associated with significant mean differences in body weight and excess weight loss of -5.1 kg [95%
confidence interval (CI) -7.3, -3.0; four trials; n = 151; I(2)  = 37%] and 12.6% (95% CI 9.0, 16.2; four trials; n = 166; I(2)
 = 24%), respectively, compared with diet modification. The mean differences in glycated hemoglobin (-0.9%; 95% CI -1.8,
0.0) and fasting plasma glucose (-3.7 mM; 95% CI -8.2, 0.8) among subjects with T2D did not reach statistical
significance. Adverse events consisted mainly of abdominal pain, nausea, and vomiting. No deaths occurred. Future high-
quality long-term RCTs are needed to further assess efficacy and safety of the DJBS for obesity.
Clinical Practice Guidelines
American Diabetes Association (ADA)
The American Diabetes Association (ADA) Standards of Medical Care in Diabetes 2022 states that metabolic surgery
should be recommended as an option to treat type 2 diabetes in appropriate surgical candidates with a BMI of 40 kg/m2
(BMI 37.5 kg/m2 in Asian Americans), regardless of the level of glycemic control or complexity of glucose-lowering
regimens, and in adults with a BMI of 35.039.9 kg/m2 (32.537.4 kg/m2 in Asian Americans) when hyperglycemia is
inadequately controlled despite lifestyle and optimal medical therapy. Metabolic surgery may be considered as an option
for adults with type 2 diabetes and a BMI of 30.034.9 kg/m2 (27.532.4 kg/m2 in Asian Americans) if hyperglycemia is
inadequately controlled despite nonsurgical methods. They strongly recommend that long-term lifestyle support and
routine monitoring of micronutrient and nutritional status be provided to patients after surgery, according to guidelines for
postoperative management of metabolic surgery by national and international professional societies. The ADA’s 2017
Standards of Medical Care in Diabetes noted that the ADA now refers to bariatric surgery as metabolic surgery.
The joint statement by international diabetes organizations on metabolic surgery in the treatment algorithm for type 2
diabetes (American Diabetes Association, International Diabetes Foundation, Diabetes UK, Chinese Diabetes Society,
and Diabetes India) made the following recommendations:
Metabolic surgery is recommended as an option to treat T2D in patients with the following conditions:
o Class III obesity (BMI ≥ 40 kg/m
2
), regardless of the level of glycemic control or complexity of glucose-lowering
regimens.
o Class II obesity (BMI 35.0 39.9 kg/m
2
) with inadequately controlled hyperglycemia despite lifestyle and optimal
medical therapy.
Metabolic surgery should also be considered and an option to treat T2D in patients with class I obesity and
inadequately controlled hyperglycemia despite optimal medical treatment by either oral or injectable medications.
All BMI thresholds used in these recommendations should be reconsidered depending on the ancestry of the patient.
For example, for patients of Asian descent, the BMI values above should be reduced by 2.5 kg/m
2
.
The organizations note that additional studies are needed to further demonstrate long-term benefits (Rubino et al., 2016).
American College of Gastroenterology (ACG)
In an ACG Clinical Guideline for the Diagnosis and Management of Gastroesophageal Reflux Disease (Katz, et al. 2022),
the following recommendations are made:
For refractory GERD, recommend optimization of PPI therapy as the first step in management of refractory GERD
(Moderate quality of evidence/strong strength of evidence)
For GERD management, recommend maintenance PPI therapy indefinitely or antireflux surgery for patients with LA
grade C or D esophagitis (Moderate quality of evidence/strong strength of evidence)
American Gastroenterological Association (AGA)
In 2021, the AGA conducted a technical review on intragastric balloons (IGB) for the management of morbid obesity
(Muniraj et al., 2021).
The review suggests that IGB therapy with lifestyle modification is an effective weight-loss intervention and seems to
result in improvements in metabolic parameters and medical comorbidities. Several evidence gaps were addressed in this
review and include long-term efficacy of IGB therapy compared with SOC beyond 1 year, variables such as the filling
medium (fluid vs gas) the potential efficacy of an ongoing dietary intervention, pharmacotherapy, or the need for
sequential balloon placement for sustained weight loss, and the role of exercise in weight-loss sustainability. Although the
Bariatric Surgery
Page 36 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
risk of serious adverse events appears to be relatively low, early removal due to device intolerance seems to be relatively
common. The AGA makes the following recommendations:
In individuals with obesity seeking a weight-loss intervention who have failed a trial of conventional weight-loss
strategies, suggest the use of IGB therapy with lifestyle modification over lifestyle modification alone. (Conditional
recommendation, moderate certainty)
In individuals with obesity undergoing IGB therapy, recommend moderate- to high-intensity concomitant lifestyle
modification interventions to maintain and augment weight loss. (Strong recommendation, moderate certainty)
In individuals undergoing IGB therapy, recommend prophylaxis with proton pump inhibitors. (Strong recommendation,
moderate certainty)
In individuals undergoing IGB therapy, suggest using the intraoperative anesthetic regimens associated with the
lowest incidence of nausea along with perioperative antiemetics; suggest a scheduled antiemetic regimen for 2 weeks
after IGB placement. (Conditional recommendation, low certainty)
In individuals undergoing IGB therapy, suggest against perioperative laboratory screening for nutritional deficiencies.
(Conditional recommendation, low certainty)
Suggest daily supplementation with 12 adult dose multivitamins after IGB placement. (Conditional recommendation,
very low certainty)
After IGB removal, suggest subsequent weight loss or maintenance interventions that include dietary interventions,
pharmacotherapy, repeat IGB or bariatric surgery; the choice of weight loss or maintenance method after IGB is
determined based on patient’s context and comorbidities following a shared decision-making approach. (Conditional
recommendation, low certainty)
American Society for Gastrointestinal Endoscopy (ASGE)
The ASGE Technology Committee conducted a systematic review and meta-analysis to evaluate whether endoscopic
technologies have met appropriate thresholds outlined by ASGE by the Preservation and Incorporation of Valuable
endoscopic Innovations (PIVI) document (Abu Dayyeh et al., 2015a). The study authors evaluated Orbera intragastric
balloon (IGB) (Apollo Endosurgery) and the EndoBarrier duodenal-jejunal bypass sleeve (DJBS) (GI Dynamics). Results
of the meta-analysis (17 studies, n = 1683) indicate that the Orbera IGB satisfies the PIVI thresholds for therapy for
primary and non-primary bridge obesity. The percentage of EWL (%EWL) associated with the Orbera IGB at 12 months
was 25.44% (95% CI, 21.45 to 29.41%) with a mean difference over controls of 26.9% (%EWL) (95% CI, 15.66% to
38.24%; p 0.01) in a total of 3 RCTs. The pooled %TWL after use of Orbera IGW was 13% at 6 months (95% CI,
12.37% to 13.95%) and 11.27% (95% CI, 8.17% to 14.36%), both which exceed the PIVI threshold of 5% TBWL for
nonprimary bridge obesity therapy.
In its position statement on EBTs in clinical practice, the ASGE states that EBTs that have been approved by the FDA and
meet thresholds of efficacy and safety as defined in the ASGE/ASMBS Preservation and Incorporation of Valuable
Endoscopic Innovations should be included in the obesity treatment algorithm as adjunctive therapies to a lifestyle
intervention program as outlined in the 2013 American Heart Association(AHA)/American College of Cardiology(ACC)/The
Obesity Society (TOS) guidelines for the management of overweight and obesity in adults. ASGE advises that
endoscopists performing EBT have a mechanism to enroll patients in long-term follow-up care for weight loss
maintenance (Sullivan et al., 2015).
American Association of Clinical Endocrinologists (AACE)/Obesity Society/American
Society for Metabolic and Bariatric Surgery (ASMBS)
In a clinical practice guideline for the perioperative nutritional, metabolic, and nonsurgical support of the bariatric surgery
patient, the AACE, the Obesity Society, and the ASMBS (Mechanick, et al., 2019) cite the following:
Patients with a BMI 40 kg/m2 without coexisting medical problems and for whom bariatric surgery would not be
associated with excessive risk should be eligible.
Patients with a BMI 35 kg/m2 and one or more severe obesity-related complications remediable by weight loss,
including type 2 diabetes (T2D), high risk for T2D (insulin resistance, prediabetes, and/or metabolic syndrome), poorly
controlled hypertension, nonalcoholic fatty liver disease/nonalcoholic steatohepatitis, obstructive sleep apnea,
osteoarthritis of the knee or hip, and urinary stress incontinence, should be considered for a bariatric procedure.
Patients with the following comorbidities and BMI ≥ 35 kg/m2 may also be considered for a bariatric procedure,
though the strength of evidence is more variable: obesity-hypoventilation syndrome and Pickwickian syndrome after a
careful evaluation of operative risk; idiopathic intracranial hypertension; gastroesophageal reflux disease; severe
venous stasis disease; impaired mobility due to obesity; and considerably impaired quality of life.
Patients with BMI of 3034.9 kg/m2 and T2D with inadequate glycemic control despite optimal lifestyle and medical
therapy should be considered for a bariatric procedure; current evidence is insufficient to support recommending a
bariatric procedure in the absence of obesity.
Bariatric Surgery
Page 37 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
The body mass index criterion for bariatric procedures should be adjusted for ethnicity (e.g., 18.5 to 22.9 kg/m
2
is
normal range, 23 to 24.9 kg/m
2
overweight, and ≥ 25 kg/m
2
obesity for Asians).
Interventions should first include a multidisciplinary approach, including dietary change, physical activity, behavioral
modification with frequent follow up; and then if appropriate, pharmacologic therapy and/or surgical revision.
Selection of a bariatric procedure should be based on the individualized goals of therapy (e.g., weight loss and/or
metabolic [glycemic] control), available local-regional expertise (surgeon and institution), patient preferences, and
personalized risk stratification.
In addition, they recommend that all patients seeking bariatric surgery have a comprehensive preoperative evaluation.
This assessment is to include an obesity-focused history, physical examination, and pertinent laboratory and diagnostic
testing. A detailed weight history should be documented, including a description of the onset and duration of obesity, the
severity, and recent trends in weight. Causative factors to note include a family history of obesity, use of weight-gaining
medications, and dietary and physical activity patterns.
A brief summary of personal weight loss attempts, commercial plans, and physician-supervised programs should be
reviewed and documented, along with the greatest duration of weight loss and maintenance. This information is useful in
substantiating that the patient has made reasonable attempts to control weight before considering obesity surgery. The
guidelines state that preoperative weight loss should be considered for patients in whom reduced liver volume can
improve the technical aspects of surgery.
American Association of Clinical Endocrinologists (AACE)/American College of
Endocrinology (ACE)
The AACE and the ACE developed comprehensive clinical practice guidelines for the medical care of patients with obesity
(Garvey, et al., 2016) based on diligent review of clinical evidence with “transparent incorporation of subjective factors.”
The final recommendations recognize that obesity is a complex, adiposity-based chronic disease, where management
targets both weight-related complications and adiposity to improve overall health and quality of life. The detailed evidence-
based recommendations allow for nuanced clinical decision-making that addresses real-world medical care of patients
with obesity, including screening, diagnosis, evaluation, selection of therapy, treatment goals, and individualization of
care. The goal is to facilitate high-quality care of patients with obesity and provide a rational, scientific approach to
management that optimizes health outcomes and safety. Included in their clinical guideline are the following
recommendations pertaining to BMI:
Patients with a BMI of ≥ 40 kg/m2 without coexisting medical problems and for whom the procedure would not be
associated with excessive risk should be eligible for bariatric surgery.
Patients with a BMI of ≥ 35 kg/m2 and 1 or more severe obesity-related complications, including T2DM, hypertension,
obstructive sleep apnea, obesity-hypoventilation syndrome, Pickwickian syndrome, nonalcoholic fatty liver disease or
nonalcoholic steatohepatitis, pseudotumor cerebri, gastroesophageal reflux disease, asthma, venous stasis disease,
severe urinary incontinence, debilitating arthritis, or considerably impaired quality of life may also be considered for a
bariatric surgery procedure.
Patients with a BMI of 30-34.9 kg/m2 with diabetes or metabolic syndrome may also be considered for a bariatric
procedure, although current evidence is limited by the number of patients studied and lack of long-term data
demonstrating net benefit.
Independent of BMI criteria, there is insufficient evidence for recommending a bariatric surgical procedure specifically
for glycemic control alone, lipid lowering alone, or CVD risk reduction alone.
American Heart Association/American College of Cardiology (AHA/ACC)/Obesity
Society
The AHA/ACC and the Obesity Society published an updated 2013 Practice Guideline and Management of Overweight
and Obesity in Adults (Jensen et al., 2014). The updated guidelines reflect such consensus and offer update regarding
treatment for patients who are overweight or obese. While the focus remains on sustained weight loss and decreased
waist circumference, the authors also recommend use of bariatric surgery for patients with a BMI > 40, or BMI > 35 with
comorbidities.
In a scientific statement on severe obesity in children and adolescents the American Heart Association (Kelly et al., 2013),
summarized that RYGB has been associated with improvement or resolution of numerous comorbid conditions, including
OSAS, T2DM, features of metabolic syndrome, pseudotumor cerebri, and psychosocial functioning. Controlled,
prospective adult studies demonstrate a marked effect of bariatric surgery on mortality, comorbidity reversal, and
prevention of comorbidity over ensuing decades; these beneficial effects of bariatric surgery help to inform clinical
decision making for severely obese adolescents when no other treatments have demonstrated long-term effectiveness.
Bariatric Surgery
Page 38 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
American Society for Metabolic & Bariatric Surgery (ASMBS)
Presurgical Evaluations
The ASMBS published recommendations for the presurgical psychosocial evaluation of bariatric surgery patients (Sogg et
al., 2016). They recommend that bariatric behavioral health clinicians with specialized knowledge and experience be
involved in the evaluation and care of patients both before and after surgery. Given the importance of long-term follow up
after weight loss surgery (WLS), the preoperative psychosocial assessment provides a valuable opportunity for patients to
establish a trusted connection to a behavioral health provider as an additional resource and integral participant in their
postoperative care. The need to ensure that postoperative psychosocial care is available has been noted in established
practice guidelines and evidence suggests that such care is associated with better outcomes after surgery.
In a 2016 position statement on preoperative supervised weight loss requirements, the ASMBS noted that there is no data
from any randomized controlled trial, large prospective study, or meta-analysis to support the practice of mandated
preoperative weight loss. Further, there is no Level I data in the surgical literature, or consensus in the medical literature
(based on over 40 published RCTs) that has clearly identified any one dietary regimen, duration or type of weight loss
program that is optimal for patients with clinically severe obesity. Finally, they recommend that patients seeking surgical
treatment for clinically severe obesity should be evaluated based on their initial BMI and co-morbid conditions.
Nutritional Impact of Bariatric Surgery
In an updated guideline on the integrated health nutritional guidelines for surgical weight loss, the ASMBS (Parrott et al.,
2017) states that optimizing postoperative patient outcomes and nutritional status begins preoperatively. Patients should
be educated before and after WLS on the expected nutrient deficiencies associated with alterations in physiology.
Although surgery can exacerbate preexisting nutrient deficiencies, preoperative screening for vitamin deficiencies has not
been the norm in the majority of WLS practices. Screening is important because it is common for patients who present for
WLS to have at least 1 vitamin or mineral deficiency preoperatively.
Data continue to suggest that the prevalence of micronutrient deficiencies is increasing, while monitoring of patients at
follow-up is decreasing. The ASMBS recommends that their guideline be considered a reasonable approach to patient
nutritional care based on the most recent research, scientific evidence, resources, and information available. It is the
responsibility of the registered dietitian nutritionist and WLS program to determine individual variations as they relate to
patient nutritional care.
Indications for Surgery
In a joint update, the ASMBS and the International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO)
released revised guidelines on indications for metabolic and bariatric surgery (MBS) (Eisenberg et al., 2023). Updates to
the guidelines include:
MBS is recommended for individuals with a BMI ≥ 35 kg/m
2
, regardless of presence, absence, or severity of
comorbidities
MBS should be considered for individuals with metabolic disease and BMI of 30-34.9 kg/m
2
BMI thresholds should be adjusted in the Asian population such that a BMI ≥ 25 kg/m
2
suggests clinical obesity, and
individuals with BMI ≥ 27.5 kg/m
2
should be offered MBS
Long-term results of MBS consistently demonstrate safety and efficacy
Appropriately selected children and adolescents should be considered for MBS
Specific Bariatric Procedures
The ASMBS (2016, updated 2019) has approved, and supports the use of the following bariatric procedures and
associated devices:
Roux-en-Y Gastric Bypass
BPD/Duodenal Switch
Intragastric Balloon
Sleeve Gastrectomy
Adjustable Gastric Banding
Single Anastomosis Duodeno-ileostomy with Sleeve
Bariatric Reoperative Procedures
A 2017 ASMBS updated position statement on sleeve gastrectomy (SG) as a bariatric procedure (Ali et al., 2017)
summarized that:
Bariatric Surgery
Page 39 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Substantial long-term outcome data published in the peer-reviewed literature including studies comparing outcomes of
various surgical procedures, confirm that sleeve gastrectomy (SG) provides significant and durable weight loss,
improvements in medical co-morbidities, improved quality of life, and low complication and mortality rates for obesity
treatment.
SG is now the most commonly performed procedure in the United States (~53.8% of all bariatric procedures),
followed by Roux-en-Y gastric bypass (RYGB; 23.1% of all procedures) (Chaar et al., 2018).
In terms of initial early weight loss and improvement of most weight-related co-morbid conditions, SG and RYGB
appear similar.
SG is an acceptable option for a primary bariatric procedure or as a first-stage procedure in high-risk patients as part
of a planned, staged approach.
The effect of SG on GERD is less clear because GERD improvement is less predictable, and GERD may worsen or
develop de novo. Preoperative counseling specific to GERD-related outcomes is recommended for all patients
undergoing SG.
Based on safety and efficacy data, there is a trend toward SG as the procedure of choice for adolescents, although
both RYGB and SG are routinely performed in teen weight loss surgery programs.
As with any bariatric procedure, long-term weight regain can occur after SG and may require one or more of a variety
of re-interventions.
In an updated statement (Kallies and Rogers, 2020) on the single-anastomosis duodenal switch (SADS), the ASMBS has
concluded that single-anastomosis duodenoileal bypass with sleeve gastrectomy (SADI-S) provides for similar outcomes
to those for the classic biliopancreatic diversion with duodenal switch (BPD-DS) procedure and therefore should be
recognized. The society conclusion is that the current available peer-reviewed literature does not suggest outcomes will
differ substantially from those seen with classic DS procedure. While the ASMBS endorses SADI-S as an appropriate
bariatric surgical procedure, the society indicates publication of long-term safety and efficacy outcomes is still needed and
is strongly encouraged; concerns remain about intestinal adaptation, nutritional issues, and long-term weight loss/regain
following this procedure.
The ASMBS Clinical Issues Committee position statement on intragastric balloon therapy endorsed by SAGES (Ali, et al.,
2016) includes the following summary and recommendations:
Level 1 data regarding the clinical utility, efficacy, and safety of intragastric balloon therapy for obesity are derived
from randomized clinical studies.
Implantation of intragastric balloons can result in notable weight loss during treatment.
Although utilization of intragastric balloons results in notable weight loss, separating the effect of the balloon alone
from those of supervised diet and lifestyle changes may be challenging. Of note, recent FDA pivotal trials
demonstrated a benefit to balloon use compared with diet alone in their study populations. In general, any obesity
treatment, including intragastric balloon therapy, would benefit from a multidisciplinary team that is skilled and
experienced in providing in-person medical, nutritional, psychological, and exercise counseling.
The safety profiles for intragastric balloons indicate a safe intervention, with serious complications being rare. Early
postoperative tolerance challenges can be significant but can be controlled with pharmacotherapy in the majority of
patients, thereby minimizing voluntary balloon removals. These early symptoms should be discussed with the patient
before the procedure.
Although therapy with prolonged balloon in situ time and the use of sequential treatments with multiple balloons have
been studied, awareness and adherence to absolute and relative contraindications of use and timely removal optimize
device safety.
Based on current evidence, balloon therapy is FDA approved as an endoscopic, temporary (maximum 6 months) tool
for the management of obesity. Further review will evaluate the impact of diet, lifestyle changes, and
pharmacotherapy during and after balloon removal.
The ability to perform appropriate follow-up is essential when intragastric balloons are used for weight loss to enhance
their safety and avoid complications related to spontaneous deflation and bowel obstruction.
The ASMBS (Moore and Rosenthal, 2018) released an addendum to their intragastric balloon therapy position statement
in response to the FDA’s warnings on complications not identified during initial clinical trials, and worldwide mortalities
associated with intragastric balloons. They recommend that:
As with all procedures, it is important that patients give informed consent and are aware of potential adverse events.
Laypeople may need to be counseled to correct a misperception that endolumenal treatments are nonsurgical and
thus risk-free.
When less powerful treatments are chosen, behavioral modification increases in importance and there is risk of weight
regain after the device is retrieved. The ASMBS routinely advocates for multidisciplinary care and support of the
weight loss patient, and this recommendation is even more crucial for intragastric balloon recipients.
Bariatric Surgery
Page 40 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
The ASMBS, in their 2015 position statement on vagal blocking therapy for obesity (Papasavas et al., 2016), conclude
that the quantity of the data available at this time (6 published studies; approximately 600 implanted devices) and the
length of follow-up indicate adequate safety and efficacy in the short term. More prospective studies with longer follow-up
are required to establish the clinically significant efficacy and patient tolerance of this device.
In a 2015 position statement on intragastric balloon therapy endorsed by SAGES, the ASMBS acknowledges that
although utilization of intragastric balloons results in notable weight loss, separating the effect of the balloon alone from
those of supervised diet and lifestyle changes may be challenging (Ali et al., 2016).
Bariatric Surgery in Adolescents
The updated ASMBS pediatric metabolic and bariatric surgery guidelines (Pratt et al., 2018) state that the disease of
obesity has become recognized as a metabolic disease controlled by genetic factors, with clear evidence that the
physiologic control of weight is through neuroendocrine pathways that regulate body mass by affecting satiety, hunger,
and metabolism. The recognition that weight is largely not under volitional control leads to a strong need to offer effective,
sustainable, proven therapies to children with obesity.
The summary of major changes in the guideline includes:
Patient selection criteria of a BMI ≥ 20% of the 95th percentile with a co-morbidity or a BMI ≥ 140% of the 95th
percentile should be used when determining weight cut offs for adolescents to undergo metabolic and bariatric
surgery (MBS). In their opinion, Tanner stage and linear growth should not be used to determine readiness for MBS.
Preoperative attempts at diet and exercise: there are no data that the number of weight loss attempts correlates with
success after MBS. Compliance with a multi- disciplinary preoperative program may improve out-comes after MBS but
prior attempts at weight loss should be removed as a barrier to definitive treatment for obesity.
Requiring adolescents with a BMI > 40 to have a co-morbidity (as in the old guidelines) puts children at a significant
disadvantage to attaining a healthy weight. Earlier surgical intervention (at a BMI < 45 kg/m2) can allow adolescents
to reach a normal weight and avoid lifelong medication therapy and end organ damage from co-morbidities.
Certain co-morbidities should be considered in adolescents, specifically the psychosocial burden of obesity, the
orthopedic diseases specific to children, GERD, and cardiac risk factors. Given the poor outcomes of medical
therapies for type 2 diabetes in children, these co-morbidities may be considered an indication for MBS in younger
adolescents or those with lower obesity percentiles.
Nonalcoholic fatty liver disease (NAFLD) and steatohepatitis (NASH): NAFLD may be present in at least 59% of
adolescent patients referred for MBS. Given complete resolution of NASH in approximately 85% of patients who
undergo VSG or RYGB, NAFLD should be considered a strong indication for MBS in adolescents with severe obesity.
OSA has been shown to cause significantly decreased health-related quality of life (HRQoL) with increased risk of
morbidity and mortality in adolescents. MBS in adolescents results in significant improvement or resolution of OSA.
Thus, OSA should be considered a strong indication for MBS.
Adolescents who suffer from severe obesity and have failed medical management of idiopathic intracranial
hypertension should be considered for MBS.
Adolescents with severe obesity have significant risk factors for cardiovascular disease (CVD), including,
hyperlipidemia, elevated inflammatory markers, hypertension, and insulin resistance. MBS significantly improves
these risk factors, and therefore would be expected to decrease morbidity and mortality from CVD long term.
Multidisciplinary teams should stabilize and treat preexisting eating disorders, assure stable social support, assess,
and assist with nutrition and activity knowledge, and consider the addition of medications when appropriate.
The Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP) guidelines should
be followed when building an adolescent MBS program. It is the responsibility of the adolescent MBS program to have
a transition plan in place for adolescents to transition to an adult MBS program for lifelong care.
The ASMBS Pediatric Committee (Michalsky et al., 2012) best practice guidelines state that the associated risk/benefit
analysis of bariatric surgery in adolescents should also include the consideration of the potential long-term health risks of
untreated or inadequately treated obesity for the individual candidate. In addition, patients with a greater BMI and more
serious medical illness are at increased risk of complications after bariatric surgery. Providing access to bariatric surgery
earlier in life when the disease burden and severity is lower might decrease the operative risk, morbidity, and mortality.
Additionally, earlier surgical intervention alters the natural course of many obesity-related co-morbidities that otherwise
would put the patient at risk of long-term complications and early mortality.
Impact of Obesity and Obesity Treatment on Fertility and Fertility Therapy
In a position statement endorsed by the American College of Obstetricians and Gynecologists (ACOG) and the Obesity
Society (Kominiarek et al., 2017), the ASMBS summarized that:
Bariatric Surgery
Page 41 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Bariatric surgery is effective in achieving significant and sustained weight loss in morbidly obese women and has
been shown in case-control studies to improve fertility.
Pregnancy is not recommended during the rapid weight-loss phase after bariatric surgery; therefore, counseling and
follow-up regarding contraception during this period is important.
The specific impact of either medical weight-loss treatments or bariatric surgery on the responsiveness to subsequent
treatments for infertility in both men and women is not clearly understood at this time.
Revisional Bariatric Surgery
In a systematic review of reoperative bariatric surgery, the ASMBS Revision Task Force (Brethauer et al., 2014) states
that the indications and outcomes for reoperative bariatric surgery are procedure-specific, but the current evidence does
support additional treatment for persistent obesity, co-morbid disease, and complications. Additional surgical therapy may
benefit patients who present with insufficient weight loss, continued co-morbid disease, or weight gain after the index
bariatric procedure. A thorough evaluation should be conducted by a multi-disciplinary program to determine the potential
causes for their poor responses.
As the risks of reoperative bariatric surgery are higher than with the primary procedure, evidence suggests the need for
careful patient selection. In addition, the specific type of reoperative procedure performed should be based on the
patient’s primary procedure, the patient’s anatomy, the patient’s weight and co-morbidities, and the experience of the
surgeon.
An ASMBS Task Force (Sudan et al., 2015) on reoperative surgery provided the updated definitions for reoperative
surgery as follows:
Any operation after the first bariatric operation which qualified toward center of excellence volume requirements is
considered a reoperation. Reoperations were further divided into corrective operations or conversions.
An operation is considered corrective when complications or incomplete treatment effect of a previous bariatric
operation was addressed but the initial operation was not changed.
Conversions involve changing an index bariatric operation (first operation) to a different type of bariatric operation,
and reversal restored original anatomy.
The Task Force also conducted a systematic review to evaluate morbidity, mortality, and weight loss outcomes after
reoperative bariatric surgery. Data on reoperations was compared to that from patients who had initial bariatric operations
but did not undergo reoperations. Reoperations were subdivided into corrective operations and conversions.
Out of 449,753 bariatric operations, 28,720 (6.3%) underwent reoperations of which 19,970 (69.5%) were corrective
and 8,750 (30.5%) were conversions.
The mean % EBWL after conversion to a different bariatric operation was 39.3% and was 35.9% after a corrective
operation. Although this % EBWL was lower than that after a primary operation (43.5%), it is still considered by the
Task Force to be substantial and excellent weight loss. However, not all reoperations will result in further weight loss
or resolution of comorbidity.
Restorative operations necessitated by intolerable side effects or complications of the index procedure such as
removal of the laparoscopic adjustable gastric band for band intolerance or dilated esophagus or reversing a
duodenal switch or a gastric bypass for severe malabsorption, may in fact result in weight gain and return of
comorbidities.
Elderly patients (> 60 years of age) comprised 11% of the primary and 12% of the reoperative group of patients. The
data suggests an overall improvement in the rates of morbidity and mortality after bariatric operations in recent years,
even for higher risk populations.
The Task Force concluded that although most patients do not require reoperative surgery, among those who do, the
complication rate is low, and outcomes are clinically comparable to primary procedures.
American Academy of Pediatrics (AAP)
In 2023, the AAP published the first edition of the clinical practice guideline for evaluation and management of children
and adolescents with overweight and obesity. This document recommends metabolic and bariatric surgery for pediatric
patients over the age of 12 for the following:
Class II obesity, BMI 35 or 120% of the 95
th
percentile for age and sex, whichever is lower with clinically significant
disease, including but not limited to:
o T2DM
o Idiopathic intracranial hypertension
o Non-alcoholic steatohepatitis
o Blount’s Disease
Bariatric Surgery
Page 42 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
o Slipped capital femoral epiphysis
o GERD
o OSA with an AHI > 5
o Cardiovascular disease risks
o Depressed health related QOL
Class III obesity, BMI ≥ 40 or 140% of the 95th percentile for age and sex, whichever is lower
Furthermore, the following is stated:
The determination of eligibility for metabolic and bariatric surgery should rely heavily on a multicomponent and
individualized approach between members of the metabolic and bariatric surgery team, the patient, and the patient’s
parents or guardians.
A referral should be to a comprehensive metabolic and bariatric surgery center with experience and expertise in
treatment of patients younger than 18 years.
Evaluation for metabolic and bariatric surgery should include a holistic view of the patient and family, including
individual needs (physical and psychosocial) and social risk factors.
American Society for Metabolic and Bariatric Surgery (ASMBS)/National Lipid
Association (NLA)/Obesity Medicine Association (OMA)
The ASMBS, NLA and OMA published a 2-part joint scientific statement on lipids and bariatric procedures. Part 1
concluded that bariatric procedures reduce body fat and have favorable effects on adipocyte and adipose tissue function,
which contributes to improvement in metabolic diseases such as dyslipidemia, high glucose levels, and high blood
pressure. Among the mechanisms by which bariatric procedures may improve dyslipidemia includes favorable alterations
in endocrine and inflammatory homeostasis. Bariatric procedures may also have favorable effects on bile acid metabolism
and the intestinal microbiome, which may also improve dyslipidemia (Bays et al., 2016a).
Part 2 of this joint scientific statement summarized that the principles that apply to bariatric procedures and lipid levels
include the following: (1) The greater the fat mass loss, the greater the improvement in lipid parameters such as
triglycerides and especially LDL cholesterol; (2) bariatric procedures allow for a decrease in the use of drug treatment for
dyslipidemia; and (3) after bariatric procedures, HDL cholesterol may transiently decrease for the first 36 months after
the procedure, which is usually followed by an increase in HDL cholesterol above the baseline value before the bariatric
procedure. Finally, the authors observed that data are scarce regarding the effects of bariatric procedures on some of the
lipid parameters such as non-HDL cholesterol, apolipoprotein B, and lipoprotein particle number and remnant lipoproteins
(Bays et al., 2016b).
Endocrine Society
In its updated guideline for the assessment, prevention, and treatment of pediatric obesity (Styne et al., 2017) the
Endocrine Society’s recommendations include the following:
Diagnose a child or adolescent > 2 years of age as overweight if the BMI is ≥ 85th percentile but < 95th percentile for
age and sex, as obese if the BMI is ≥ 95th percentile, and as extremely obese if the BMI is ≥ 120% of the 95th
percentile or ≥ 35 kg/m
2
.
Children or adolescents with a BMI of ≥ 85th percentile should be evaluated for potential comorbidities.
Insulin concentrations should not be utilized when evaluating children or adolescents for obesity.
Bariatric surgery is suggested only under the following conditions:
o The patient has attained Tanner 4 or 5 pubertal development and final or near-final adult height, the patient has a
BMI of > 40 kg/m2 or has a BMI of > 35 kg/m
2
and significant, extreme comorbidities.
o T2DM, moderate to extreme sleep apnea, pseudotumor cerebri, debilitating orthopedic problems, and
nonalcoholic steatohepatitis with advanced fibrosis.
o Extreme obesity and comorbidities persist despite compliance with a formal program of lifestyle modification, with
or without pharmacotherapy.
o BMI of > 40 kg/m2 with mild comorbidities (hypertension, dyslipidemia, moderate orthopedic problems, mild sleep
apnea, nonalcoholic steatohepatitis, and extreme psychological distress that is secondary to their obesity).
o Psychological evaluation confirms the stability and competence of the family unit [psychological distress due to
impaired quality of live (QOL) from obesity may be present, but the patient does not have an underlying untreated
psychiatric illness].
o The patient demonstrates the ability to adhere to the principles of healthy dietary and activity habits.
o There is access to an experienced surgeon in a pediatric bariatric surgery center of excellence that provides the
necessary infrastructure for patient care, including a team capable of long-term follow-up of the metabolic and
psychosocial needs of the patient and family.
Bariatric Surgery
Page 43 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Bariatric surgery should not be performed in preadolescent children, pregnant or breast-feeding adolescents (and
those planning to become pregnant within 2 years of surgery), and in any patient who has not mastered the principles
of healthy dietary and activity habits and/or has an unresolved substance abuse, eating disorder, or untreated
psychiatric disorder.
Society of American Gastrointestinal and Endoscopic Surgeons (SAGES)
A 2010 guideline by SAGES states that due to concerns for higher failure rates after fundoplication in the morbidly obese
patient (BMI > 35 kg/m
2
) and the inability of fundoplication to address the underlying problem (obesity) and its associated
co-morbidities, gastric bypass should be the procedure of choice when treating GERD in this patient group. The benefits
in patients with BMI > 30 is less clear and needs further study (Stefanidis et al., 2010).
In its 2008 Guidelines for Clinical Application of Laparoscopic Bariatric Surgery, endorsed by the ASMBS, SAGES
confirms that bariatric surgery is medically indicated for morbidly obese patients who fail to respond to dietary, behavioral,
nutritional, and medical therapies, with clear evidence of efficacy and safety. BMI and age-based candidacy guidelines
should not limit access for patients suffering with progressive or poorly controlled obesity-related comorbidities if the risk-
versus-benefit analysis favors surgery. Laparoscopic RGB, AGB, and BPD have all been proven effective. They do not
make a definitive recommendation for one procedure over another and note that at the present time, decisions are driven
by patient and surgeon preferences, as well as considerations regarding the degree and timing of necessary outcomes
versus tolerance of risk and lifestyle change.
Further, the 2008 guidelines state that there are no absolute contraindications to bariatric surgery. Relative
contraindications to surgery may include severe heart failure, unstable coronary artery disease, end-stage lung disease,
active cancer diagnosis/treatment, cirrhosis with portal hypertension, uncontrolled drug or alcohol dependency, and
severely impaired intellectual capacity. Crohn’s disease may be a relative contraindication to Roux-en-Y gastric bypass
and biliopancreatic diversion.
Multidisciplinary Care Task Group
Greenberg et al. (2005) found a high incidence of depression, negative body image, eating disorders, and low quality of
life (QoL) in patients with severe obesity and that perceived obesity-related health problems, motivation, and sense of
coherence (SoC) predicted better weight loss. Although their investigation showed there are no predictive relationships
between preoperative psychological evaluations and postoperative weight loss, the Behavioral and Psychological
subgroup of the Multidisciplinary Care Task Group recommended that all bariatric surgery candidates be evaluated by a
licensed mental health care provider experienced in the treatment of severely obese patients and working with a
multidisciplinary team. Although research supports the association of psychological problems such as depression and
personality disorder with less successful obesity surgery outcomes, rarely are the psychological problems cited as
contraindications for surgery (Greenberg et al., 2005).
National Institute for Health and Care Excellence (NICE)
The National Institute for Health and Care Excellence (NICE) 2014 guideline on obesity identification, assessment and
management offers bariatric surgery as a treatment option for people with obesity when they have: a BMI of 40 kg/m2 or
more, or between 35 kg/m2 and 40 kg/m2 and other significant disease (for example, type 2 diabetes or high blood
pressure) that could be improved if they lost weight; all appropriate non-surgical measures have been tried but the person
has not achieved or maintained adequate, clinically beneficial weight loss; have a multi-disciplinary team approach; the
person is generally fit for surgery and anesthesia; and the person commits to the need for long-term follow-up. In addition,
the NICE guideline notes that bariatric surgery is the option of choice (instead of lifestyle interventions or drug treatment)
for adults with a BMI of more than 50 kg/m2 when other interventions have not been effective. Further, surgical
intervention is not generally recommended in children or young people, however it may be considered only in exceptional
circumstances, and if they have achieved or nearly achieved physiological maturity.
A 2015 NICE interventional procedure guidance on managing type 2 diabetes states that current evidence on the safety
and efficacy of implantation of a duodenaljejunal bypass liner for managing type 2 diabetes is limited in quality and
quantity. Therefore, the procedure should only be used in the context of research. Further research should give details of
patient selection, including information about use of the procedure in patients with different levels of BMI. The research
should provide information on complications; reasons for early removal of the device; medication used for treating type 2
diabetes, both when the device is in place and after its removal; and control of type 2 diabetes after device removal. In
2018, the following statement was added to this guidance: The device used in this procedure (EndoBarrier) no longer has
a current CE mark. The CE mark is necessary for medical devices to be marketed in the European Union. A non-CE
marked device can only be used in the context of clinical investigations with MHRA and research ethical approval.
Bariatric Surgery
Page 44 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Interventional procedures guidance [IPG569] from NICE (2016) states that the current evidence on the safety of single-
anastomosis duodeno-ileal bypass with sleeve gastrectomy (SADI
S
) for treating morbid obesity shows that there are
well-recognized complications. The evidence on efficacy is limited in both quality and quantity. Therefore, this procedure
should only be used with special arrangements for clinical governance, consent and audit or research.
A 2020 NICE interventional procedure guidance on swallowable gastric balloon for weight loss states that the evidence on
efficacy is inadequate and this procedure should only be done in a research setting.
American Academy of Sleep Medicine (AASM)
The American Academy of Sleep Medicine commissioned a task force of experts in sleep medicine, otolaryngology, and
bariatric surgery to develop recommendations based on a systematic review of the literature (Kent, 2021). The following
are recommendations intended as a guide for clinicians who treat overweight adults with OSA:
Recommend clinicians discuss referral to a sleep surgeon with adults with OSA and BMI < 40 who are intolerant or
unaccepting of CPAP (STRONG).
Recommend clinicians discuss referral to a bariatric surgeon with adults with OSA and obesity (class II/III, BMI ≥35)
who are intolerant or unaccepting of PAP (STRONG).
Suggest clinicians discuss referral to a sleep surgeon with adults with OSA, BMI < 40 and persistent inadequate PAP
adherence due to pressure-related side effects (CONDITIONAL).
Suggest clinicians recommend PAP as an initial therapy for adults with OSA and a major upper airway anatomic
abnormality prior to consideration of referral for upper airway surgery (CONDITIONAL).
Department of Veterans Affairs (VA)/Department of Defense (DoD)
The 2020 guideline from the VA/DoD (Mayer et al., 2020) for the management of adult overweight or obesity makes the
following suggestions or recommendations:
In patients with a body mass index of ≥ 30 kg/m2 and type 2 diabetes mellitus, suggest offering the option of
metabolic/bariatric surgery, in conjunction with a comprehensive lifestyle intervention.
In adult patients with a body mass index ≥ 40 kg/m2 or those with body mass index ≥ 35 kg/m2 with obesity-
associated condition(s), suggest offering the option of metabolic/bariatric surgery, in conjunction with a
comprehensive lifestyle intervention, for long-term weight loss/maintenance and/or to improve obesity-associated
condition(s).
In patients with obesity (body mass index 30 kg/m2) who prioritize short-term (up to six months) weight loss,
suggest offering intragastric balloons in conjunction with a comprehensive lifestyle intervention.
There is insufficient evidence to recommend for or against metabolic/bariatric surgery to patients over age 65.
There is insufficient evidence to recommend for or against percutaneous gastrostomy devices for weight loss in
patients with obesity.
There is insufficient evidence to recommend for or against intragastric balloons for long-term weight loss to support
chronic weight management or maintenance.
Thoracic Society
In a clinical practice guideline from the Thoracic Society (Hudgel, 2018), the following recommendations are made for
patients who are overweight and suffer from OSA:
Reduced-calorie diet, and
Exercise or increased physical activity, and
Behavioral guidance.
In addition, it was stated that pharmacological therapy and bariatric surgery are appropriate for selected patients who
require further assistance with weight loss.
U.S. Food and Drug Administration (FDA)
This section is to be used for informational purposes only. FDA approval alone is not a basis for coverage.
Bariatric surgical procedures are not subject to FDA regulation. FDA approval information for several devices related to
bariatric surgery is described below.
The FDA approved the ORBERA
Intragastric Balloon System (Apollo Endosurgery, Inc.) on August 5, 2015. The
ORBERA System is indicated for use as an adjunct to weight reduction in obese adults with BMI ≥ 30 and ≤ 40 kg/m2. It is
to be used in conjunction with a long-term supervised diet and behavior modification program designed to increase the
Bariatric Surgery
Page 45 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
likelihood of significant long-term weight loss and weight loss maintenance. It is indicated for adults who have failed
conservative weight reduction strategies, such as supervised diet, exercise, and behavior modification program. ORBERA
has a maximum placement period of 6 months. For more information, refer to:
https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpma/pma.cfm?id=p140008
https://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfpma/pma.cfm?id=P140008S016
(Accessed August 24, 2023)
Gastric banding involves the use of an adjustable or nonadjustable gastric band, which is subject to FDA marketing
approval. In 2001, the BioEnterics
®
LAP-BAND System was approved by FDA for marketing under the premarket
approval process. According to the FDA labeling, this is approved for surgical treatment for severely obese adults for
whom more conservative treatments (e.g., diet, exercise, behavioral modification) have failed. The LAP-BAND System is
indicated for use in weight reduction for severely obese patients with a Body Mass Index (BMI) of at least 40 or a BMI of
at least 35 with one or more severe co-morbid conditions, or those who are 100 lbs. or more over their estimated ideal
weight according to the 1983 Metropolitan Life Insurance Tables (use the midpoint for medium frame). It is indicated for
use only in severely obese adult patients who have failed more conservative weight-reduction alternatives, such as
supervised diet, exercise, and behavior modification programs.
In February 2011, the FDA approved the Lap-Band Adjustable Gastric Banding System, by Allergan, for weight reduction
in obese patients, with a Body Mass Index (BMI) of at least 40 kg/m2 or less obese patients who have at least a body
mass index (BMI) of 30 kg/m2 and one or more additional obesity-related co-morbid condition, such as diabetes or
hypertension. Additional information is available at: http://www.accessdata.fda.gov/cdrh_docs/pdf/p000008s017a.pdf
.
(Accessed August 24, 2023)
Adjustable gastric bands are contraindicated in patients younger than 18 years of age.
Surgical stapling devices are used in all bariatric surgical procedures except gastric banding. These devices have been
approved by FDA for use in various general surgical procedures. One device is the Endo Gia Universal Auto Suture,
which inserts six parallel rows of staples into tissue. Other surgical staplers are manufactured by Ethicon Endo-Surgery.
Additional information, product code GDW and GAG, is available at:
http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfRL/listing.cfm
. (Accessed August 24, 2023)
The OverStitch
Endoscopic Suturing System was granted 510(k) marketing approval on June 27, 2018. According to the
FDA, it is intended for endoscopic placement of suture(s) and approximation of soft tissue within the gastrointestinal tract.
The device can utilize either a single- or dual-channel endoscope. Additional information is available at:
https://www.accessdata.fda.gov/cdrh_docs/pdf18/K181141.pdf
. (Accessed August 24, 2023)
The TransPyloric Shuttle/TransPyloric Shuttle Delivery Device was granted Premarket Approval on April 18, 2019, and is
indicated for weight reduction in adult patients with obesity with a BMI of 35.0-40.0 kg/m2 or a BMI of 30.0 to 34.9 kg/m2
with one or more obesity related comorbid conditions and intended to be used in conjunction with a diet and behavior
modification program. https://www.accessdata.fda.gov/cdrh_docs/pdf18/P180024a.pdf
. (Accessed August 24, 2023)
In August of 2018, the FDA granted GI Dynamics Inc., Boston, MA an Investigational Device Exemption for the
EndoBarrier
®
gastrointestinal liner. Additional information is available at: https://www.fda.gov/medical-devices/how-study-
and-market-your-device/investigational-device-exemption-ide. (Accessed August 24, 2023)
References
Abu Dayyeh BK, Bazerbachi F, Vargas EJ, et al.; MERIT Study Group; Galvao Neto M, Zundel N, Wilson EB. Endoscopic
sleeve gastroplasty for treatment of class 1 and 2 obesity (MERIT): a prospective, multicentre, randomised trial. Lancet.
2022 Aug 6;400(10350):441-451.
Abu Dayyeh BK, Edmundowicz SA, Jonnalagadda S, et al. Endoscopic bariatric therapies. Gastrointest Endosc. 2015a;
81(5).
Abu Dayyeh BK, Kumar N, Edmundowicz SA, et al. ASGE Bariatric Endoscopy Task Force systematic review and meta-
analysis assessing the ASGE PIVI thresholds for adopting endoscopic bariatric therapies. Gastrointest Endosc. 2015b;
82(3):425-438.
Abu Dayyeh BK, Maselli DB, Rapaka B, et al. Adjustable intragastric balloon for treatment of obesity: a multicentre, open-
label, randomised clinical trial. Lancet. 2021 Nov 27;398(10315):1965-1973.
Bariatric Surgery
Page 46 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Ahmad A, Carleton JD, Ahmad ZF, et al. Laparoscopic versus robotic-assisted Roux-en-Y gastric bypass: a retrospective,
single-center study of early perioperative outcomes at a community hospital. Surg Endosc. 2016 Sep;30(9):3792-6.
Alarcón Del Agua I, Socas-Macias M, Busetto L, et al. Post-implant analysis of epidemiologic and eating behavior data
related to weight loss effectiveness in obese patients treated with gastric electrical stimulation. Obes Surg. 2017
Jun;27(6):1573-1580.
Ali M, El Chaar M, Ghiassi S, et al. ASMBS guidelines/statements. American Society for Metabolic and Bariatric Surgery
updated position statement on sleeve gastrectomy as a bariatric procedure. Surg Obes Relat Dis. 2017 Oct;13(10):1652-
1657.
Ali MR, Moustarah F, Kim JJ; American Society for Metabolic and Bariatric Surgery Clinical Issues Committee. American
Society for Metabolic and Bariatric Surgery position statement on intragastric balloon therapy endorsed by the Society of
American Gastrointestinal and Endoscopic Surgeons. Surg Obes Relat Dis. 2016 Mar-Apr;12(3):462-467.
Alqahtani AR, Elahmedi M, Abdurabu HY, et al. Ten-year outcomes of children and adolescents who underwent sleeve
gastrectomy: weight loss, comorbidity resolution, adverse events, and growth velocity. J Am Coll Surg. 2021 Aug
24:S1072-7515(21)01929-3.
Altieri MS, Yang J, Nie L, et al. Rate of revisions or conversion after bariatric surgery over 10 years in the state of New
York. Surg Obes Relat Dis. 2018 Apr;14(4):500-507.
American Academy of Pediatrics (AAP). Clinical practice guideline for the evaluation and treatment of children and
adolescents with obesity. February 2023. Available at:
https://publications.aap.org/pediatrics/article/151/2/e2022060640/190443/Clinical-Practice-Guideline-for-the-Evaluation-
and?autologincheck=redirected. Accessed October 5, 2023.
American Diabetes Association (ADA). Obesity and Weight Management for the Treatment of Type 2 Diabetes: Standards
of medical care in diabetes 2022. Diabetes Care 2022 Jan 1;45(Suppl 1):S113-S124.
American Diabetes Association (ADA). Standards of medical care in diabetes 2017. Diabetes Care
2017;40(Suppl.1):S1S2.
American Society for Metabolic and Bariatric Surgery (ASMBS). Position statement. Preoperative supervised weight loss
requirements. April 2016.
American Society for Metabolic and Bariatric Surgery (ASMBS). Approved procedures and devices. March 2019.
Available at: https://asmbs.org/resources/endorsed-procedures-and-devices?/resources/approved-procedures
. Accessed
August 24, 2023.
Angrisani L, Cutolo PP, Formisano G, et al. Long-term outcomes of laparoscopic adjustable silicone gastric banding
(LAGB) in moderately obese patients with and without co-morbidities. Obes Surg. 2013 Jul;23(7):897-902.
Apovian CM, Shah SN, Wolfe BM, et al. Two-year outcomes of vagal verve blocking (vBloc) for the treatment of obesity in
the ReCharge Trial. Obes Surg. 2017 Jan;27(1):169-176.
Arterburn DE, Olsen MK, Smith VA, et al. Association between bariatric surgery and long-term survival. JAMA. 2015;
313(1):62-70.
Ayloo S, Roh Y, Choudhury N. Laparoscopic, hybrid, and totally robotic Roux-en-Y gastric bypass. J Robot Surg. 2016
Mar;10(1):41-7.
Bai ZB, Qin YL, Deng G, et al. Bariatric embolization of the left gastric arteries for the treatment of obesity: 9-month data
in 5 patients. Obes Surg. 2018 Apr;28(4):907-915.
Bays HE, Jones PH, Jacobson TA, et al. Lipids and bariatric procedures part 1 of 2: scientific statement from the National
Lipid Association, American Society for Metabolic and Bariatric Surgery, and Obesity Medicine Association: full report. J
Clin Lipidol. 2016a Jan-Feb;10(1):33-57.
Bays HE, Kothari SN, Azagury DE, et al. ASMBS guidelines/statements. Lipids and bariatric procedures part 2 of 2:
scientific statement from the American Society for Metabolic and Bariatric Surgery (ASMBS), the National Lipid
Association (NLA), and Obesity Medicine Association (OMA). Surg Obes Relat Dis. 2016b Mar-Apr;12(3):468-495.
Beamish AJ, Gronowitz E, Olbers T, et al. Body composition and bone health in adolescents after Roux-en-Y gastric
bypass for severe obesity. Pediatr Obes. Pediatr Obes. 2017 Jun;12(3):239-246.
Beckmann JH, Bernsmeier A, Kersebaum JN, et al. The impact of robotics in learning Roux-en-Y gastric bypass: a
retrospective analysis of 214 laparoscopic and robotic procedures: robotic vs. laparoscopic RYGB. Obes Surg. 2020
Jun;30(6):2403-2410.
Bariatric Surgery
Page 47 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Benedix F, Krause T, Adolf D, et al. Perioperative course, weight loss and resolution of comorbidities after primary sleeve
gastrectomy for morbid obesity: are there differences between adolescents and adults? Obes Surg. 2017 Sep;27(9):2388-
2397.
Betzel B, Homan J, Aarts EO, et al. Weight reduction and improvement in diabetes by the duodenal-jejunal bypass liner: a
198-patient cohort study. Surg Endosc. 2017 Jul;31(7):2881-2891.
Bindal V, Bhatia P, Dudeja U, et al. Review of contemporary role of robotics in bariatric surgery. J Minim Access Surg.
2015 Jan-Mar; 11(1):1621.
Brethauer SA, Hammel JP, Schauer PR. Systematic review of sleeve gastrectomy as staging and primary bariatric
procedure. Surg Obes Relat Dis. 2009 Jul-Aug;5(4):469-475.
Brethauer SA, Kothari S, Sudan R, et al. Systematic review on reoperative bariatric surgery: American Society for
Metabolic and Bariatric Surgery Revision Task Force. Surg Obes Relat Dis. 2014. Sep-Oct;10(5):952-72.
Buttelmann K, Linn JG, Denham W, Ruiz M, Yetasook A, Ujiki M. Management options for obesity after bariatric surgery.
Surg Laparosc Endosc Percutan Tech. 2015 Feb;25(1):15-18.
Bužga M, Švagera Z, Tomášková H, et al. Metabolic effects of sleeve gastrectomy and laparoscopic greater curvature
plication: an 18-month prospective, observational, open-label study. Obes Surg. 2017 Dec;27(12):3258-3266.
California Technology Assessment Forum. Institute for Clinical and Economic Review 2015. Controversies in obesity
management. A technology assessment. June 2015.
Camilleri M, Toouli J, Herrera MF, et al. Intra-abdominal vagal blocking (VBLOC therapy): Clinical results with a new
implantable medical device. Surgery. 2008;143(6):723-731.
Carbajo MA, Jiménez JM, Luque-de-León E, et al. Evaluation of weight loss indicators and laparoscopic one-anastomosis
gastric bypass outcomes. Sci Rep. 2018 Jan 31;8(1):1961.
Carbajo MA, Fong-Hirales A, Luque-de-León E, et al. Weight loss and improvement of lipid profiles in morbidly obese
patients after laparoscopic one-anastomosis gastric bypass: 2-year follow-up. Surg Endosc. 2017 Jan;31(1):416-421.
Centers for Disease Control and Prevention (CDC), National Center for Health Statistics. Clinical growth charts. Updated
December 2022. Available at: https://www.cdc.gov/growthcharts/extended-bmi.htm
. Accessed October 5, 2023.
Centers for Disease Control and Prevention (CDC). National Center for Health Statistics. Obesity and Overweight.
https://www.cdc.gov/nchs/fastats/obesity-overweight.htm
. Accessed August 24, 2023.
Centers for Disease Control and Prevention (CDC). What is BMI. August 2021. Available at:
https://www.cdc.gov/healthyweight/assessing/bmi/adult_bmi/index.html
. Accessed August 24, 2023.
Cha R, Marescaux J, Diana M. Updates on gastric electrical stimulation to treat obesity: Systematic review and future
perspectives. World J Gastrointest Endosc. 2014 Sep 16;6(9):419-31.
Chaar ME, Lundberg P, Stoltzfus J. Thirty-day outcomes of sleeve gastrectomy versus Roux-en-Y gastric bypass: first
report based on Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program database. Surg Obes
Relat Dis. 2018 May;14(5):545-551.
Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of nonalcoholic fatty liver disease: Practice
guidance from the American Association for the Study of Liver Diseases. Hepatology. 2018 Jan;67(1):328-357.
Chang SH, Stoll CR, Song J, et al. The effectiveness and risks of bariatric surgery: an updated systematic review and
meta-analysis, 2003-2012. JAMA Surg. 2014 Mar;149(3):275-87.
Chierici A, Chevalier N, Iannelli A. Postoperative morbidity and weight loss after revisional bariatric surgery for primary
failed restrictive procedure: A systematic review and network meta-analysis. Int J Surg. 2022 Jun;102:106677.
Christou NV, Sampalis JS, Liberman M, et al. Surgery decreases long-term mortality, morbidity, and health care use in
morbidly obese patients. Ann Surg. 2004 Sep;240(3):416-23.
Clapp B, Wynn M, Martyn C, et al. Long term (7 or more years) outcomes of the sleeve gastrectomy: a meta-analysis.
Surg Obes Relat Dis. 2018 Jun;14(6):741-747.
Coffin B, Maunoury V, Pattou F, et al. Impact of intragastric balloon before laparoscopic gastric bypass on patients with
super obesity: a randomized multicenter study. Obes Surg.2017;27(4):902-909.
Colquitt JL, Pickett K, Loveman E, et al. Surgery for weight loss in adults. Cochrane Database Syst Rev. 2014 Aug
8;(8):CD003641.
Cooper TC, Simmons EB, Webb K, et al. Trends in weight regain following Roux-en-y gastric bypass (RYGB) bariatric
surgery. Obes Surg. 2015 Aug;25(8):1474-81.
Bariatric Surgery
Page 48 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Cottam A, Cottam D, Portenier D, et al. A matched cohort analysis of stomach intestinal pylorus saving (SIPS) surgery
versus biliopancreatic diversion with duodenal switch with two-year follow-up. Obes Surg. 2017;27(2):454-461.
Cottam A, Cottam D, Medlin W, et al. A matched cohort analysis of single anastomosis loop duodenal switch versus
Roux-en-Y gastric bypass with 18-month follow-up. Surg Endosc. 2016 Sep;30(9):3958-64.
Cottam D, Roslin M, Enochs P, et al. Single Anastomosis Duodenal Switch: 1-Year Outcomes. Obes Surg. 2020
Apr;30(4):1506-1514.
Courcoulas A, Abu Dayyeh BK, Eaton L, et al. Intragastric balloon as an adjunct to lifestyle intervention: a randomized
controlled trial. Int J Obes. 2017;41(3):427-433.
Dardamanis D, Navez J, Coubeau L, et al. A retrospective comparative study of primary versus revisional Roux-en-Y
gastric bypass: long-term results. Obes Surg. 2018 Aug;28(8):2457-2464.
David MB, Abu-Gazala S, Sadot E, et al. Laparoscopic conversion of failed vertical banded gastroplasty to Roux-en-Y
gastric bypass or biliopancreatic diversion. Surg Obes Relat Dis. 2015 Sep-Oct;11(5):1085-91.
Desai NK, Wulkan ML, Inge TH. Update on adolescent bariatric surgery. Endocrinol Metab Clin North Am. 2016
Sep;45(3):667-76.
Dixon J, O'Brien P, Playfair J, et al. Adjustable gastric banding and conventional therapy for type 2 diabetes. JAMA.
2008;299(3):316-323.
Doležalova-Kormanova K, Buchwald JN, Skochova D, et al. Five-year outcomes: laparoscopic greater curvature plication
for treatment of morbid obesity. Obes Surg. 2017 Nov;27(11):2818-2828.
Economopoulos KP, Theocharidis V, McKenzie TJ, et al. Robotic vs. laparoscopic Roux-En-Y gastric bypass: a
systematic review and meta-analysis. Obes Surg. 2015 Nov;25(11):2180-9.
ECRI Institute. Clinical Evidence Assessment. AspireAssist Gastric Aspiration Port (Aspire Bariatrics, Inc.) for Treating
Obesity. February 2021.
ECRI Institute. Clinical Evidence Assessment. Ingestible intragastric balloons for treating obesity. April 2020.
ECRI Institute. Clinical Evidence Assessment. Orbera Intragastric Balloon (Apollo Endosurgery, Inc.) for Treating Obesity.
September 2015. Updated February 2021.
ECRI Institute. Clinical Evidence Assessment. Rechargeable vagal blocking system (Maestro) for treating obesity. July
2016. Updated May 2017.
ECRI Institute. Clinical Evidence Assessment. Spatz3
®
Adjustable Gastric Balloon (Spatz FGIA, Inc.) for Treating Obesity.
January 2022.
ECRI Institute. Custom Product Brief. TransPyloric Shuttle implant (BAROnova, Inc.) for Treating Obesity. May 2019.
ECRI. Health Information Assessment. Ingestible Intragastric Balloons for Treating Obesity April 2020.
Eid GM, McCloskey CA, Eagleton JK, et al. StomaphyX vs a sham procedure for revisional surgery to reduce regained
weight in Roux-en-Y gastric bypass patients: a randomized clinical trial. JAMA Surg. 2014 Apr;149(4):372-9.
Eisenberg D, Shikora SA, Aarts E, et al. 2022 American Society of Metabolic and Bariatric Surgery (ASMBS) and
International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO) Indications for Metabolic and Bariatric
Surgery. Obes Surg. 2023 Jan;33(1):3-14. Erratum in: Obes Surg. 2022 Nov 29.
El Chaar M, Stoltzfus J, Claros L, et al. Indications for revisions following 630 consecutive laparoscopic sleeve
gastrectomy cases: Experience in a single accredited center. J Gastrointest Surg. 2017 Jan;21(1):12-16.
Elhag W, El Ansari W, Abdulrazzaq S, et al. Evolution of 29 anthropometric, nutritional, and cardiometabolic parameters
among morbidly obese adolescents 2 years post sleeve gastrectomy. Obes Surg. 2018 Feb;28(2):474-482.
English WJ, DeMaria EJ, Brethauer SA, et al. American Society for Metabolic and Bariatric Surgery estimation of
metabolic and bariatric procedures performed in the United States in 2016. Surg Obes Relat Dis. 2018 Mar;14(3):259-
263.
Epstein LJ, Kristo D, Strollo PJ, et al. Adult Sleep Apnea Taskforce of the American Academy of Sleep Medicine. Clinical
guideline for the evaluation, management and long-term care of obstructive sleep apnea in adults. Journal of Clinical
Sleep Medicine. March 2009. 5(3): 263-276.
Eskandaros MS, Abbass A, Zaid MH, et al. Laparoscopic one anastomosis gastric bypass versus laparoscopic Roux-en-Y
gastric bypass effects on pre-existing mild-to-moderate gastroesophageal reflux disease in patients with obesity: a
randomized controlled study. Obes Surg. 2021 Aug 18.
Bariatric Surgery
Page 49 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Felsenreich DM, Kefurt R, Schermann M, et al. Reflux, sleeve dilation, and Barrett's Esophagus after laparoscopic sleeve
gastrectomy: long-term follow-up. Obes Surg. 2017 Dec;27(12):3092-3101.
Flølo TN, Andersen JR, Kolotkin RL, et al. Five-year outcomes after vertical sleeve gastrectomy for severe obesity: a
prospective cohort study. Obes Surg. 2017 Aug;27(8):1944-1951.
Forner PM, Ramacciotti T, Farey JE, et al. Safety and effectiveness of an endoscopically placed duodenal-jejunal bypass
device (EndoBarrier
®
): outcomes in 114 patients. Obes Surg. 2017 Dec;27(12):3306-3313.
Forssell H, Norén E. A novel endoscopic weight loss therapy using gastric aspiration: results after 6 months. Endoscopy.
2015 Jan;47(1):68-71.
Froylich D, Abramovich-Segal T, Pascal G, et al. Long-term (over 10 years) retrospective follow-up of laparoscopic
adjustable gastric banding. Obes Surg. 2018 Apr;28(4):976-980.
Fulton C, Sheppard C, Birch D, et al. A comparison of revisional and primary bariatric surgery. Can J Surg. 2017
Jun;60(3):205-211.
Gallas S, Fetissova S. Ghrelin, appetite and gastric electrical stimulation. Peptides. Volume 32, Issue 11, November 2011,
Pages 22832289.
Garvey WT, Mechanick JI, Brett EM, et al. American Association of Clinical Endocrinologists and American College of
Endocrinology comprehensive clinical practice guidelines for medical care of patients with obesity. Endocr Pract. 2016
Jul;22 Suppl 3:1-203.
Gersin KS, Rothstein RI, Rosenthal RJ, et al. Open-label, sham-controlled trial of an endoscopic duodenojejunal bypass
liner for preoperative weight loss in bariatric surgery candidates. Gastrointest Endosc. 2010;71(6):976-982.
Giet L, Baker J, Favretti F, et al. Medium and long-term results of gastric banding: outcomes from a large private clinic in
UK. BMC Obes. 2018 Apr 12;5:12.
Giordano S. Laparoscopic Roux-en-Y gastric bypass versus laparoscopic adjustable gastric banding in the super-obese:
peri-operative and early outcomes. Scandinavian Journal of Surgery. 2015; 104(1):5-9.
Gray KD, Moore MD, Elmously A, et al. Perioperative outcomes of laparoscopic and robotic revisional bariatric surgery in
a complex patient population. Obes Surg. 2018 Jul;28(7):1852-1859.
Greenberg I, Perna F, Kaplan M, et al. Behavioral and psychological factors in the assessment and treatment of obesity
surgery patients. Obesity Research. 2005;13(2),244149.
Grubnik VV, Ospanov OB, Namaeva KA, et al. Randomized controlled trial comparing laparoscopic greater curvature
plication versus laparoscopic sleeve gastrectomy. Surg Endosc. 2016 Jun;30(6):2186-91.
Gudur AR, Geng CX, Kshatri S, et al. Comparison of endoscopic sleeve gastroplasty versus surgical sleeve gastrectomy:
a Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program database analysis. Gastrointest
Endosc. 2023 Jan;97(1):11-21.
Hardin AP, Hackell JM, American Academy of Pediatrics Committee on Practice and Ambulatory Medicine. Age limit of
pediatrics. Pediatrics. 2017 Sep;140(3). pii: e20172151.
Hayes, Inc., Clinical Research Response. AspireAssist (Aspire Bariatrics) Aspiration Therapy for Weight Loss in Obese
Individuals. Lansdale PA: Hayes, Inc., April 2021.
Hayes, Inc. Comparative Effectiveness Review. Mini-Gastric Bypass-One Anastomosis Gastric Bypass for the Treatment
of Obesity: A Review of Reviews. Lansdale PA: Hayes Inc., May 2019. Updated November 2021.
Hayes, Inc. Evolving Evidence Review. OverStitch Endoscopic Suturing System (Apollo Endosurgery Inc.) for Endoscopic
Sleeve Gastroplasty. Lansdale PA: Hayes, Inc., May 2022.
Hayes, Inc. Health Technology Brief. Single-anastomosis duodenal switch for weight loss. Landsale PA: Hayes, Inc.,
February 2018. Updated March 2020. Archived March 2021.
Hayes, Inc. Medical Technology Directory. Comparative Effectiveness Review of Bariatric Surgeries for Treatment of
Obesity in Adolescents. Lansdale, PA: Hayes Inc.; January 2019. Updated April 2021.
Hayes, Inc. Hayes Medical Technology Directory. Intragastric balloons for treatment of obesity. Lansdale, PA: Hayes, Inc.;
March 2018. Updated May 2021.
Hafezi-Nejad N, Bailey CR, Gunn AJ, et al. Weight loss after left gastric artery embolization: a systematic review and
meta-analysis. J Vasc Interv Radiol. 2019 Oct;30(10):1593-1603.
Hedjoudje A, Abu Dayyeh BK, Cheskin LJ, et al. Efficacy and safety of endoscopic sleeve gastroplasty: a systematic
review and meta-analysis. Clin Gastroenterol Hepatol. 2020 May;18(5):1043-1053.e4.
Bariatric Surgery
Page 50 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Hervieux E, Baud G, Dabbas M, et al. Comparative results of gastric banding in adolescents and young adults. J Pediatr
Surg. 2016 Jul;51(7):1122-5.
Hoeltzel GD, Swendiman RA, Tewksbury CM, et al. How safe is adolescent bariatric surgery? An analysis of short-term
outcomes. J Pediatr Surg. 2021 Sep 5:S0022-3468(21)00583-2.
Hudgel DW, Patel SR, Ahasic AM, et al; American Thoracic Society Assembly on Sleep and Respiratory Neurobiology.
The role of weight management in the treatment of adult obstructive sleep apnea. An Official American Thoracic Society
Clinical Practice Guideline. Am J Respir Crit Care Med. 2018 Sep 15;198(6):e70-e87.
Ikramuddin S, Korner J, Lee WJ, et al. Lifestyle intervention and medical management with vs without Roux-en-Y gastric
bypass and control of hemoglobin A1c, LDL cholesterol, and systolic blood pressure at 5 years in the Diabetes Surgery
Study. JAMA. 2018 Jan 16;319(3):266-278.
Ikramuddin S, Blackstone RP, Brancatisano A, et al. Effect of reversible intermittent intra-abdominal vagal nerve blockade
on morbid obesity: the ReCharge randomized clinical trial. JAMA. 2014; 312(9):915-922.
Inge TH, Laffel LM, Jenkins TM, et al. Comparison of surgical and medical therapy for type 2 diabetes in severely obese
adolescents. JAMA Pediatr. 2018 May 1;172(5):452-460.
Inge TH, Courcoulas AP, Jenkins TM, et al. Weight loss and health status 3 years after bariatric surgery in adolescents. N
Engl J Med. 2016 Jan 14;374(2):113-23.
Iranmanesh P, Boudreau V, Barlow K, et al. Comparison of single- versus double-anastomosis duodenal switch: a single-
center experience with 2-year follow-up. Int J Obes (Lond). 2021 Aug;45(8):1782-1789.
Jambhekar A, Maselli A, Robinson S, et al. Demographics and socioeconomic status as predictors of weight loss after
laparoscopic sleeve gastrectomy: A prospective cohort study. Int J Surg. 2018 Jun;54(Pt A):163-169.
Janik MR, Rogula TG, Mustafa RR, et al. Safety of revision sleeve gastrectomy compared to Roux-Y Gastric Bypass after
failed gastric banding: analysis of the MBSAQIP. Ann Surg. 2019 Feb;269(2):299-303.
Jensen MD, Ryan DH, Apovian CM, et al; American College of Cardiology/American Heart Association Task Force on
Practice Guidelines; Obesity Society. 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in
adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and
The Obesity Society. Circulation. 2014 Jun 24;129(25 Suppl 2):S102-38.
Jirapinyo P, Abu Dayyeh BK, Thompson CC. Weight regain after Roux-en-Y gastric bypass has a large negative impact
on the bariatric quality of life index. BMJ Open Gastro 2017;4:e000153.
Jirapinyo P, de Moura DTH, Horton LC, et al. Effect of Aspiration Therapy on Obesity-Related Comorbidities: Systematic
Review and Meta-Analysis. Clin Endosc. 2020 Nov;53(6):686-697.
Jung SH, Yoon JH, Choi HS, et al; Korean Research Group for Endoscopic Management of Metabolic Disorder and
Obesity. Comparative efficacy of bariatric endoscopic procedures in the treatment of morbid obesity: a systematic review
and network meta-analysis. Endoscopy. 2020 Nov;52(11):940-954.
Kadeli DK, Sczepaniak JP, Kumar K, et al. The effect of preoperative weight loss before gastric bypass: A systematic
review. J Obes. 2012;2012:867540.
Kallies K, Rogers AM; American Society for Metabolic and Bariatric Surgery Clinical Issues Committee. American Society
for Metabolic and Bariatric Surgery updated statement on single-anastomosis duodenal switch. Surg Obes Relat Dis.
2020 Jul;16(7):825-830.
Kang JH, Le QA. Effectiveness of bariatric surgical procedures: A systematic review and network meta-analysis of
randomized controlled trials. Medicine (Baltimore). 2017 Nov;96(46):e8632.
Kansou G, Lechaux D, Delarue J, et al. Laparoscopic sleeve gastrectomy versus laparoscopic mini gastric bypass: One
year outcomes. Int J Surg. 2016 Jul 22;33(Pt A):18-22.
Kapeluto JE, Tchernof A, Masckauchan D, et al. Ten-year remission rates in insulin-treated type 2 diabetes after
biliopancreatic diversion with duodenal switch. Surg Obes Relat Dis. 2020 Nov;16(11):1701-1712.
Katz PO, Dunbar KB, Schnoll-Sussman FH, et al. ACG Clinical Guideline for the Diagnosis and Management of
Gastroesophageal Reflux Disease. Am J Gastroenterol. 2022 Jan 1;117(1):27-56.
Kelly AS, Barlow SE, Rao G, et al. Severe obesity in children and adolescents: identification, associated health risks, and
treatment approaches: a scientific statement from the American Heart Association. Circulation. 2013 Oct 8;128(15):1689-
712.
Khalaj A, Tasdighi E, Hosseinpanah F, et al. Two-year outcomes of sleeve gastrectomy versus gastric bypass: first report
based on Tehran obesity treatment study (TOTS). BMC Surg. 2020 Jul 20;20(1):160.
Bariatric Surgery
Page 51 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Khoraki J, Moraes MG, Neto APF, et al. Long-term outcomes of laparoscopic adjustable gastric banding. Am J Surg. 2018
Jan;215(1):97-103.
Kim J. American Society for Metabolic and Bariatric Surgery statement on single-anastomosis duodenal switch. Surg
Obes Relat Dis. 2016;12(5):944-945.
King K, Sudan R, Bardaro S, et al. Assessment and management of gastroesophageal reflux disease following bariatric
surgery. Surg Obes Relat Dis. 2021 Nov;17(11):1919-1925.
Koh ZJ, Chew CAZ, Zhang JJY, et al. Metabolic outcomes after revisional bariatric surgery: a systematic review and
meta-analysis. Surg Obes Relat Dis. 2020 Oct;16(10):1442-1454.
Kominiarek MA, Jungheim ES, Hoeger KM, et al. American Society for Metabolic and Bariatric Surgery position statement
on the impact of obesity and obesity treatment on fertility and fertility therapy. Endorsed by the American College of
Obstetricians and Gynecologists and the Obesity Society. Surg Obes Relat Dis. 2017 May;13(5):750-757.
Lager CJ, Esfandiari NH, Subauste AR, et al. Roux-En-Y gastric bypass vs. sleeve gastrectomy: balancing the risks of
surgery with the benefits of weight loss. Obes Surg. 2017 Jan;27(1):154-161.
Lainas P, De Filippo G, Di Giuro G, et al. Laparoscopic sleeve gastrectomy for adolescents under 18 years old with
severe obesity. Obes Surg. 2020 Jan;30(1):267-273.
Lannoo M, Dillemans B. Laparoscopy for primary and secondary bariatric procedures. Best Pract Res Clin Gastroenterol.
2014;28(1):159-173.
Lessing Y, Pencovich N, Khatib M, et al. One-anastomosis gastric bypass: first 407 patients in 1 year. Obes Surg. 2017
Oct;27(10):2583-2589.
Li X, Hu X, Fu C, et al. Efficacy and safety of one anastomosis gastric bypass versus Roux-en-Y gastric bypass for
obesity: a meta-analysis and systematic review. Obes Surg. 2023 Feb;33(2):611-622.
Liakopoulos V, Franzén S, Svensson AM, et al. Changes in risk factors and their contribution to reduction of mortality risk
following gastric bypass surgery among obese individuals with type 2 diabetes: a nationwide, matched, observational
cohort study. BMJ Open Diabetes Res Care. 2017 Jun 14;5(1):e000386.
Lopez-Nava G, Sharaiha RZ, Vargas EJ, et al. Endoscopic sleeve gastroplasty for obesity: a multicenter study of 248
patients with 24 months follow-up. Obes Surg. 2017 Oct;27(10):2649-2655.
Lundell LR, Dent J, Bennett JR, et al. Endoscopic assessment of oesophagitis: clinical and functional correlates and
further validation of the Los Angeles classification. Gut. 1999 Aug;45(2):172-80.
Magallares A. Mental and physical health-related quality of life in obese patients before and after bariatric surgery: a
meta-analysis. Psychology, Health & Medicine. 2015; 20(2):165-176.
Maciejewski ML, Arterburn DE, Van Scoyoc L, et al. Bariatric surgery and long-term durability of weight loss. JAMA Surg.
2016 Nov 1;151(11):1046-1055.
Malinka T, Zerkowski J, Katharina I, et al. Three-year outcomes of revisional laparoscopic gastric bypass after failed
laparoscopic sleeve gastrectomy: a case-matched analysis. Obes Surg. 2017 Sep;27(9):2324-2330.
Manco M, Mosca A, De Peppo F, et al. The benefit of sleeve gastrectomy in obese adolescents on nonalcoholic
steatohepatitis and hepatic fibrosis. J Pediatr. 2017 Jan;180:31-37.e2.
Marinos G, Eliades C, Raman Muthusamy V, et al. Weight loss and improved quality of life with a nonsurgical endoscopic
treatment for obesity: clinical results from a 3- and 6-month study. Surg Obes Relat Dis. 2014 Sep-Oct;10(5):929-34.
Mayer SB, Graybill S, Raffa SD, et al. Synopsis of the 2020 U.S. VA/DoD Clinical Practice Guideline for the Management
of Adult Overweight and Obesity. Mil Med. 2021 Aug 28;186(9-10):884-896.
Mechanick JI, Apovian C, Brethauer S, et al. Clinical practice guidelines for the perioperative nutritional, metabolic, and
nonsurgical support of patients undergoing bariatric procedures2019 update: cosponsored by American Association of
Clinical Endocrinologists/American College of Endocrinology, the Obesity Society, and American Society for Metabolic &
Bariatric Surgery, Obesity Medicine Association, and American Society of Anesthesiologists Executive Summary.
Endocr Pract. 2019 25(12):1346-1359.
Metabolic and Bariatric Surgery Accreditation and Quality Improvement Program (MBSAQIP). Available at:
https://www.facs.org/quality-programs/mbsaqip
. Accessed August 24, 2023.
Michalsky M, Reichard K, Inge T, et al. American Society for Metabolic and Bariatric Surgery. ASMBS pediatric committee
best practice guidelines. Surg Obes Relat Dis. 2012 Jan-Feb;8(1):1-7.
Mingrone G, Panunzi S, Gaetano A, et. al. Bariatric surgery versus conventional medical therapy for type 2 diabetes. N
Engl J Med. 2012;366(17):1577-1585.
Bariatric Surgery
Page 52 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Mirkin K, Alli VV, Rogers AM. Revisional Bariatric Surgery. Surg Clin North Am. 2021 Apr;101(2):213-222.
Mistry P, Currie V, Super P, et al. Changes in glycemic control, blood pressure and lipids 5 years following laparoscopic
adjustable gastric banding combined with medical care in patients with type 2 diabetes: a longitudinal analysis. Clin Obes.
2018 Jun;8(3):151-158.
Moore R, Rosenthal P. ASMBS proposed addendum to position statement on intragastric balloon therapy. January 2018.
Moore RL, Seger MV, Garber SM, et al. Clinical safety and effectiveness of a swallowable gas-filled intragastric balloon
system for weight loss: consecutively treated patients in the initial year of U.S. commercialization. Surg Obes Relat Dis.
2019 Mar;15(3):417-423.
Morales-Conde S, Alarcón Del Agua I, Busetto, et al. Implanted closed-loop gastric electrical stimulation (clges) system
with sensor-based feedback safely limits weight regain at 24 months. Obes Surg. 2018 Jun;28(6):1766-1774.
Morton JM, Shah SN, Wolfe BM, et al. Effect of vagal nerve blockade on moderate obesity with an obesity-related
comorbid condition: the ReCharge Study. Obes Surg. 2016 May;26(5):983-9.
Mullady DK, Lautz DB, Thompson CC. Treatment of weight regain after gastric bypass surgery when using a new
endoscopic platform: initial experience and early outcomes (with video). Gastrointest Endosc. 2009 Sep;70(3):440-4.
Muniraj T, Day LW, Teigen LM, et al. AGA Clinical Practice Guidelines on Intragastric Balloons in the Management of
Obesity. Gastroenterology. 2021 Apr;160(5):1799-1808.
Musella M, Susa A, Manno E, et al. Complications following the mini/one anastomosis gastric bypass (MGB/OAGB): a
multi-institutional survey on 2678 patients with a mid-term (5 years) follow-up. Obes Surg. 2017 Nov;27(11):2956-2967.
National Heart, Lung and Blood Institute (NHLBI). Think tank on enhancing obesity research at the National Heart, Lunch
and Blood Institute. August 2004.
National Heart, Lung and Blood Institute (NHLBI). Managing Overweight and Obesity in Adults. Systematic Evidence
Review from the Obesity Panel, 2013.
National Heart, Lung and Blood Institute (NHLBI). Classification of overweight and obesity by BMI, waist circumference,
and associated disease risks. 2016.
National Institute for Health and Care Excellence (NICE). NICE guidelines [CG189]. Obesity: identification, assessment
and management. November 2014.
National Institute for Health and Care Excellence (NICE). NICE guidelines [IPG684]. Swallowable gastric balloon capsule
for weight loss. November 2020.
National Institute for Health and Care Excellence (NICE). NICE guidelines [IPG569]. Single-anastomosis duodeno-ileal
bypass with sleeve gastrectomy for treating morbid obesity. November 2016.
National Institute for Health and Care Excellence (NICE). Interventional procedures guidance [IPG518]. Implantation of a
duodenaljejunal bypass liner for managing type 2 diabetes. March 2015.
National Heart Lung and Blood Institute (NHLBI). The Practical Guide Identification, Evaluation and Treatment of
Overweight and Obesity in Adults. October 2020.
Neto MG, Moon RC, de Quadros LG, et al. Safety and short-term effectiveness of endoscopic sleeve gastroplasty using
overstitch: preliminary report from a multicenter study. Surg Endosc. 2020 Oct;34(10):4388-4394.
Neylan CJ, Dempsey DT, Tewksbury CM, et al. Endoscopic treatments of obesity: a comprehensive review. Surg Obes
Relat Dis. 2016 Jun;12(5):1108-15.
Nocca D, Loureiro M, Skalli EM, et al. Five-year results of laparoscopic sleeve gastrectomy for the treatment of severe
obesity. Surg Endosc. 2017 Aug;31(8):3251-3257.
Noel P, Nedelcu M, Eddbali I, et al. What are the long-term results 8 years after sleeve gastrectomy? Surg Obes Relat
Dis. 2017 Jul;13(7):1110-1115.
Norén E, Forssell H. Aspiration therapy for obesity; a safe and effective treatment. BMC Obes. 2016 Dec 28;3:56.
Nunes GC, Pajecki D, de Melo ME, et al. Assessment of weight loss with the intragastric balloon in patients with different
degrees of obesity. Surg Laparosc Endosc Percutan Tech. 2017 Aug;27(4):e83-e86.
Nyström M, Machytka E, Norén E, et al. Aspiration therapy as a tool to treat obesity: 1- to 4-year results in a 201-patient
multi-center post-market European registry study. Obes Surg. 2018 Jul;28(7):1860-1868.
O’Brien PE, Sawyer SM, Laurie C, et al. Laparoscopic adjustable gastric banding in severely obese adolescents: A
Randomized Trial. JAMA. 2010;303(6):519-526.
Bariatric Surgery
Page 53 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
O'Brien PE, Hindle A, Brennan L, et al. Long-term outcomes after bariatric surgery: a systematic review and meta-analysis
of weight loss at 10 or more years for all bariatric procedures and a single-center review of 20-year outcomes after
adjustable gastric banding. Obes Surg. 2019 Jan;29(1):3-14.
Olbers T, Beamish AJ, Gronowitz E, et al. Laparoscopic Roux-en-Y gastric bypass in adolescents with severe obesity
(AMOS): a prospective, 5-year, Swedish nationwide study. Lancet Diabetes Encocrinol 2017. Online January 5, 2017.
Osland E, Yunus RM, Khan S, et al. Diabetes improvement and resolution following laparoscopic vertical sleeve
gastrectomy (LVSG) versus laparoscopic Roux-en-Y gastric bypass (LRYGB) procedures: a systematic review of
randomized controlled trials. Surg Endosc. 2017a Apr;31(4):1952-1963.
Osland E, Yunus RM, Khan S, et al. Changes in non-diabetic comorbid disease status following laparoscopic vertical
sleeve gastrectomy (LVSG) versus laparoscopic Roux-En-Y Gastric Bypass (LRYGB) procedures: a systematic review of
randomized controlled trials. Obes Surg. 2017b May;27(5):1208-1221.
Osland E, Yunus RM, Khan S, et al. Postoperative early major and minor complications in laparoscopic vertical sleeve
gastrectomy (LVSG) versus laparoscopic Roux-en-Y gastric bypass (LRYGB) procedures: a meta-analysis and
systematic review. Obes Surg. 2016 Oct;26(10):2273-84.
Papasavas P, El Chaar M, Kothari SN. American Society for Metabolic and Bariatric Surgery Clinical Issues Committee.
American Society for Metabolic and Bariatric Surgery position statement on vagal blocking therapy for obesity. Surg Obes
Relat Dis. 2016 Mar-Apr;12(3):460-461.Parmar CD, Gan J, Stier C, et al. One Anastomosis/Mini Gastric Bypass (OAGB-
MGB) as revisional bariatric surgery after failed primary adjustable gastric band (LAGB) and sleeve gastrectomy (SG): A
systematic review of 1075 patients. Int J Surg. 2020 Sep;81:32-38.
Park JH, Kim SM. High-rate of long-term revision surgery due to weight regain after Laparoscopic Gastric Greater
Curvature Plication (LGGCP). Asian J Surg. 2023 Feb;46(2):850-855.
Parrott J, Frank L, Rabena R, et al. 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 May;13(5):727-741.
Paulus GF, de Vaan LE, Verdam FJ, et al. Bariatric surgery in morbidly obese adolescents: a systematic review and
meta-analysis. Obes Surg. 2015; 25(5): 860878.
Paulus GF, van Avesaat M, van Rijn S, et al. Multicenter, phase 1, open prospective trial of gastric electrical stimulation
for the treatment of obesity: first-in-human results with a novel implantable system. Obes Surg. 2020 May;30(5):1952-
1960.
Pereira AM, Guimarães M, Pereira SS, et al. Single and dual anastomosis duodenal switch for obesity treatment: a single-
center experience. Surg Obes Relat Dis. 2021 Jan;17(1):12-19.
Piazza L, Di Stefano C, Ferrara F, et al. Revision of failed primary adjustable gastric banding to mini-gastric bypass:
results in 48 consecutive patients. Updates Surg. 2015 Dec;67(4):433-7.
Pinto-Bastos A, Conceição EM, Machado PPP. Reoperative bariatric surgery: a systematic review of the reasons for
surgery, medical and weight loss outcomes, relevant behavioral factors. Obes Surg. 2017 Oct;27(10):2707-2715.
Plamper A, Lingohr P, Nadal J, et al. Comparison of mini-gastric bypass with sleeve gastrectomy in a mainly super-obese
patient group: first results. Surg Endosc. 2017 Mar;31(3):1156-1162.
Polega JR, Barreto TW, Kemmeter KD, et al. A matched cohort study of laparoscopic biliopancreatic diversion with
duodenal switch and sleeve gastrectomy performed by one surgeon. Surg Obes Relat Dis. 2017 Mar;13(3):411-414.
Ponce J, Woodman G, Swain J, et al. The REDUCE pivotal trial: a prospective, randomized controlled pivotal trial of a
dual intragastric balloon for the treatment of obesity. Surg Obes Relat Dis. 2015 Jul-Aug;11(4):874-81.
Pratt JSA, Browne A, Browne NT, et al. ASMBS pediatric metabolic and bariatric surgery guidelines, 2018. Surg Obes
Relat Dis. 2018 Jul;14(7):882-901.
Qi L, Guo Y, Liu CQ, et al. Effects of bariatric surgery on glycemic and lipid metabolism, surgical complication and quality
of life in adolescents with obesity: a systematic review and meta-analysis. Surg Obes Relat Dis. 2017 Dec;13(12):2037-
2055.
Qiu J, Lundberg PW, Javier Birriel T, et al. Revisional bariatric surgery for weight regain and refractory complications in a
single MBSAQIP accredited center: what are we dealing with? Obes Surg. 2018 Sep;28(9):2789-2795.
Quezada N, Hernández J, Pérez G, et al. 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 Sep - Oct;12(8):1611-1615.
Quezada N, Muñoz R, Morelli C, et al. Safety and efficacy of the endoscopic duodenal-jejunal bypass liner prototype in
severe or morbidly obese subjects implanted for up to 3 years. Surg Endosc. 2018 Jan;32(1):260-267.
Bariatric Surgery
Page 54 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Reddy VY, Neužil P, Musikantow D, et al. Transcatheter bariatric embolotherapy for weight reduction in obesity. J Am Coll
Cardiol. 2020 Nov 17;76(20):2305-2317.
Risstad H, Kristinsson JA, Fagerland MW, et al. Bile acid profiles over 5 years after gastric bypass and duodenal switch:
results from a randomized clinical trial. Surg Obes Relat Dis. 2017 Sep;13(9):1544-1553.
Rohde U, Hedbäck N, Gluud LL, et al. Effect of the EndoBarrier Gastrointestinal Liner on obesity and type 2 diabetes: a
systematic review and meta-analysis. Diabetes Obes Metab. 2016 Mar;18(3):300-5.
Ruban A, Miras AD, Glaysher MA, et al. Duodenal-jejunal bypass liner for the management of Type 2 diabetes mellitus
and obesity: A multicenter randomized controlled trial. Ann Surg. 2022 Mar 1;275(3):440-447.
Rubino F, Nathan DM, Eckel RH, et al. Metabolic surgery in the treatment algorithm for type 2 diabetes: a joint statement
by international diabetes organizations. Diabetes Care 2016 Jun; 39(6): 861-877.
Ryder JR, Gross AC, Fox CK, et al. Factors associated with long-term weight-loss maintenance following bariatric surgery
in adolescents with severe obesity. Int J Obes (Lond). 2018 Jan;42(1):102-107.
Ryder JR, Edwards NM, Gupta R, et al. Changes in functional mobility and musculoskeletal pain after bariatric surgery in
teens with severe obesity: Teen-Longitudinal Assessment of Bariatric Surgery (LABS) study. JAMA Pediatr. 2016 Sep
1;170(9):871-7.
Saber AA, Shoar S, Almadani MW, et al. Efficacy of first-time intragastric balloon in weight loss: a systematic review and
meta-analysis of randomized controlled trials. Obes Surg. 2017 Feb;27(2):277-287.
Salminen P, Helmiö M, Ovaska J, et al. Effect of laparoscopic sleeve gastrectomy vs laparoscopic Roux-en-Y gastric
bypass on weight loss at 5 years among patients with morbid obesity: The SLEEVEPASS randomized clinical trial. JAMA.
2018 Jan 16;319(3):241-254.
Sanchez B, Mohr C, Morton J, et al. Comparison of totally robotic laparoscopic Roux-en-Y gastric bypass and traditional
laparoscopic Roux-en-Y gastric bypass. 2005; 1(6):549-554.
Sandler BJ, Biertho L, Anvari M, et al. Totally endoscopic implant to effect a gastric bypass: 12-month safety and efficacy
outcomes. Surg Endosc. 2018 Apr 20. [Epub ahead of print].
Sarr MG, Billington CJ, Brancatisano A, et al. The EMPOWER study: randomized, prospective, double-blind, multicenter
trial of vagal blockade to induce weight loss in morbid obesity. Obes Surg. 2012 Nov;22(11):1771-1782.
Schauer P, Kashyap S, Wolski K, et. al. Bariatric surgery versus intensive medical therapy in obese patients with
diabetes. N Engl J Med. 2012;366(17):1567-1576.
Schauer PR, Bhatt DL, Kirwan JP, et al. Bariatric surgery versus intensive medical therapy for diabetes - 5-year
outcomes. N Engl J Med. 2017 Feb 16;376(7):641-651.
Schouten R, Rijs CS, Bouvy ND, et al. A multicenter, randomized efficacy study of the EndoBarrier Gastrointestinal Liner
for presurgical weight loss prior to bariatric surgery. Ann Surg. 2010 Feb;251(2):236-43.
Serrano OK, Zhang Y, Kintzer E, et al. Outcomes of bariatric surgery in the young: a single-institution experience caring
for patients under 21 years old. 2016 Nov;30(11):5015-5022. Epub 2016 Mar 11.
Sethi M, Chau E, Youn A, et al. Long-term outcomes after biliopancreatic diversion with and without duodenal switch: 2-,
5-, and 10-year data. Surg Obes Relat Dis. 2016 Nov;12(9):1697-1705.
Shah AS, Jenkins T, Gao Z, et al. Lipid changes 8 years post gastric bypass in adolescents with severe obesity (FABS-5+
study). Int J Obes (Lond). 2017 Oct;41(10):1579-1584.
Sharples AJ, Charalampakis V, Daskalakis M, et al. Systematic review and meta-analysis of outcomes after revisional
bariatric surgery following a failed adjustable gastric band. Obes Surg. 2017 Oct;27(10):2522-2536.
Sheikh L, Pearless LA, Booth MW. Laparoscopic silastic ring mini-gastric bypass (sr-mgbp): up to 11-year results from a
single center. Obes Surg. 2017 Sep;27(9):2229-2234.
Sheiner E, Levy A, Silverberg D, et al. Pregnancy after bariatric surgery is not associated with adverse perinatal outcome.
Am J Obstet Gynecol. 2004 May;190(5):1335-1340.
Shikora SA, Bergenstal R, Bessler M, et al. Implantable gastric stimulation for the treatment of clinically severe obesity:
results of the SHAPE trial. Surg Obes Relat Dis. 2009 Jan-Feb; 5(1):31-7.
Shikora SA, Toouli J, Herrera MF, et al. Intermittent vagal nerve block for improvements in obesity, cardiovascular risk
factors, and glycemic control in patients with type 2 diabetes mellitus: 2-year results of the VBLOC DM2 Study. Obes
Surg. 2016 May;26(5):1021-8.
Bariatric Surgery
Page 55 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Shoar S, Saber AA. Long-term and midterm outcomes of laparoscopic sleeve gastrectomy versus Roux-en-Y gastric
bypass: a systematic review and meta-analysis of comparative studies. Surg Obes Relat Dis. 2017 Feb;13(2):170-180.
Singh S, Hourneaux de Moura DT, Khan A, et al. Safety and efficacy of endoscopic sleeve gastroplasty worldwide for
treatment of obesity: a systematic review and meta-analysis. Surg Obes Relat Dis. 2020 Feb;16(2):340-351.
Society of American Gastrointestinal and Endoscopic Surgeons (SAGES). Guidelines for clinical application of
laparoscopic bariatric surgery. March 2008.
Sogg S, Lauretti J, L West-Smith. Recommendations for the presurgical psychosocial evaluation of bariatric surgery
patients. Surg Obes Relat Dis 2016;12:731749.
Stefanidis D, Hope WW, Kohn GP, et al. Practice/clinical guidelines by the Society of American Gastrointestinal and
Endoscopic Surgeons (SAGES). Guidelines for surgical treatment of gastroesophageal reflux disease. February 2010.
Still CD, Benotti P, Wood C, et al. Outcomes of preoperative weight loss in high-risk patients undergoing gastric bypass
surgery. Arch Surg. 2007;142(10):994-998.
Still CD., Sarwer D., Blankenship, J. The ASMBS Textbook of Bariatric Surgery, Volume 2: Integrated Health. Chapter 19.
Pages 185-191.
StomaphyX - Gastric bypass revision. About weight loss surgery. April 30, 2010.
Strain GW, Torghabeh MH, Gagner M, et al. The impact of biliopancreatic diversion with duodenal switch (bpd/ds) over 9
years. Obes Surg. 2017 Mar;27(3):787-794.
Styne DM, Arslanian SA, Connor EL, et al. Pediatric obesity-assessment, treatment, and prevention: an Endocrine
Society clinical practice guideline. J Clin Endocrinol Metab. 2017 Mar 1;102(3):709-757.
Sudan R, Nguyen NT, Hutter MM, et al. Morbidity, mortality, and weight loss outcomes after reoperative bariatric surgery
in the USA. J Gastrointest Surg. 2015 Jan;19(1):171-8.
Sullivan S, Kumar N, Edmundowicz SA, et al. ASGE position statement on endoscopic bariatric therapies in clinical
practice. Gastrointest Endosc. 2015 Nov;82(5):767-72.
Sullivan S, Stein R, Jonnalagadda S, et al. Aspiration therapy leads to weight loss in obese subjects: a pilot study.
Gastroenterology. 2013 Dec;145(6):1245-52.e1-5.
Surve A, Zaveri H, Cottam D, et al. A retrospective comparison of biliopancreatic diversion with duodenal switch with
single anastomosis duodenal switch (SIPS-stomach intestinal pylorus sparing surgery) at a single institution with two year
follow-up. Surg Obes Relat Dis. 2017 Mar;13(3):415-422.
Syed MI, Morar K, Shaikh A, et al. Gastric Artery Embolization Trial for the Lessening of Appetite Nonsurgically (GET
LEAN): six-month preliminary data. J Vasc Interv Radiol. 2016 Oct;27(10):1502-8.
Tang Y, Tang S, Hu S. Comparative efficacy and safety of laparoscopic greater curvature plication and laparoscopic
sleeve gastrectomy: a meta-analysis. Obes Surg. 2015 Nov;25(11):2169-75.
Thompson CC, Abu Dayyeh BK, Kushner R, et al. Percutaneous Gastrostomy Device for the Treatment of Class II and
Class III Obesity: Results of a Randomized Controlled Trial. Am J Gastroenterol. 2017 Mar;112(3):447-457.
Thompson CC, Abu Dayyeh BK, Kushnir V, et al. Aspiration therapy for the treatment of obesity: 4-year results of a
multicenter randomized controlled trial. Surg Obes Relat Dis. 2019 Aug;15(8):1348-1354.
Topart P, Becouarn G, Delarue J. Weight loss and nutritional outcomes 10 years after biliopancreatic diversion with
duodenal switch. Obes Surg. 2017 Jul;27(7):1645-1650.
Tran DD, Nwokeabia ID, Purnell S, et al. Revision of Roux-En-Y gastric bypass for weight regain: a systematic review of
techniques and outcomes. Obes Surg. 2016 Jul;26(7):1627-34.
United States Census Bureau. The Asian population: 2010. March 2012.
U.S. Preventive Services Task Force (USPSTF). Obesity in Adults: Screening and Management. June 2012.
Vilarrasa N, de Gordejuela AG, Casajoana A, et al. Endobarrier
®
in grade I obese patients with long-standing type 2
diabetes: role of gastrointestinal hormones in glucose metabolism. Obes Surg. 2017 Mar;27(3):569-577.
Vinzens F, Kilchenmann A, Zumstein V, et al. Long-term outcome of laparoscopic adjustable gastric banding (LAGB):
results of a Swiss single-center study of 405 patients with up to 18 years' follow-up. Surg Obes Relat Dis. 2017
Aug;13(8):1313-1319.
Wang FG, Yu ZP, Yan WM, et al. Comparison of safety and effectiveness between laparoscopic mini-gastric bypass and
laparoscopic sleeve gastrectomy: A meta-analysis and systematic review. Medicine (Baltimore). 2017 Dec;96(50):e8924.
Bariatric Surgery
Page 56 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Weiss CR, Abiola GO, Fischman AM, et al. Bariatric Embolization of Arteries for the Treatment of Obesity (BEAT Obesity)
Trial: Results at 1 Year. Radiology. 2019 Jun;291(3):792-800.
Weiss CR, Akinwande O, Paudel K, et al. Clinical safety of bariatric arterial embolization: preliminary results of the BEAT
Obesity Trial. Radiology. 2017 May;283(2):598-608.
Wijngaarden LH, Jonker FHW, van den Berg JW, et al. Impact of initial response of laparoscopic adjustable gastric
banding on outcomes of revisional laparoscopic Roux-en-Y gastric bypass for morbid obesity. Surg Obes Relat Dis. 2017
Apr;13(4):594-599.
Xie H, Doherty L, O'Boyle C. The positive impact of bariatric surgery on sleep. Ir Med J. 2016 Jan;109(1):328-30.
Yan Y, Sha Y, Yao G, et al. Roux-en-Y gastric bypass versus medical treatment for type 2 diabetes mellitus in obese
patients: a systematic review and meta-analysis of randomized controlled trials. Medicine (Baltimore). 2016
Apr;95(17):e3462.
Yska JP, van Roon EN, de Boer A, et al. 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 Dec;150(12):1126-33.
Zhao H, Jiao L. Comparative analysis for the effect of Roux-en-Y gastric bypass vs sleeve gastrectomy in patients with
morbid obesity: Evidence from 11 randomized clinical trials (meta-analysis). Int J Surg. 2019 Dec;72:216-223.
Zitsman JL, DiGiorgi MF, Fennoy I, et al. Adolescent laparoscopic adjustable gastric banding (LAGB): prospective results
in 137 patients followed for 3 years. Surg Obes Relat Dis. 2015 Jan-Feb;11(1):101-9.
Zou ZY, Zeng J, Ren TY, et al. Efficacy of intragastric balloons in the markers of metabolic dysfunction-associated fatty
liver disease: results from meta-analyses. J Clin Transl Hepatol. 2021 Jun 28;9(3):353-363.
Policy History/Revision Information
Date
07/01/2024
Removed reference link to the Medicare Advantage Coverage Summary titled Obesity:
03/01/2024
Revised list of proven and medically necessary indications for treating obesity; removed
vertical banded gastroplasty”
Added language to indicate a planned two-stage procedure is proven and medically necessary
when all of the following criteria are met:
o Initial BMI ≥ 50 kg/m2 prior to first stage bariatric procedure
o Second stage occurs within 2 years following the primary bariatric surgery procedure
o Individual has been compliant with nutrition and exercise
o Individual meets medical necessity criteria listed above at time of second stage procedure
Replaced language indicating “gastrointestinal liners (EndoBarrier
®
) are investigational,
unproven, and not medically necessary for treating obesity due to lack of U.S. Food and Drug
Administration (FDA) approval and insufficient evidence of efficacy” with “gastrointestinal liners
are unproven and not medically necessary for treating obesity due to insufficient evidence of
efficacy”
Revised list of unproven and not medically necessary indications for treating obesity; replaced:
o “Mini-gastric bypass (MGB)/Laparoscopic mini-gastric bypass (LMGBP)” with “mini-gastric
bypass (MGB)/Laparoscopic mini-gastric bypass (LMGBP)/One-Anastomosis Gastric
Bypass (OAGB)
o “Stomach aspiration therapy (AspireAssist
®
)” with “stomach aspiration therapy
Adults
Revised language to indicate in adults age 18 years or older, bariatric surgery using one of the
procedures identified [in the policy] for treating obesity is proven and medically necessary when
all of the following criteria are met:
o One of the following:
BMI ≥ 40 kg/m
2
(or BMI ≥ 37.5 kg/m
2
in individuals of Asian descent); or
BMI ≥ 35 kg/m
2
39.9 kg/m
2
(or BMI ≥ 32.5 kg/m
2
37.4 kg/m
2
in individuals of Asian
descent) in the presence of one or more of the following co-morbidities:
Bariatric Surgery
Page 57 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Date
controlled hypertension (systolic blood pressure greater than 140 mm Hg or
diastolic blood pressure 90 mm Hg or greater, despite pharmacotherapy), coronary
artery disease, hyperlipidemia]; or
History of cardiomyopathy; or
Obstructive Sleep Apnea (OSA) confirmed on polysomnography with an AHI or RDI
of ≥ 30; or
Evidence of Nonalcoholic Fatty Liver Disease (NAFLD), or
Idiopathic intracranial hypertension (pseudotumor cerebri)
and
o The individual must also meet the following criteria:
Both of the following:
Completion of a preoperative evaluation that includes a detailed weight history
along with dietary and physical activity patterns; and
Psychosocial-behavioral evaluation by an individual who is professionally
recognized as part of a behavioral health discipline to provide screening and
identification of risk factors or potential postoperative challenges that may
contribute to a poor postoperative outcome
or
Participation in a Multidisciplinary surgical preparatory regimen
Adolescents
Revised language to indicate in adolescents age 12-17 years, the bariatric surgical procedures
identified [in the policy] are proven and medically necessary for treating obesity when all of the
following criteria are met:
o One of the following:
Class III obesity; or
Class II obesity in the presence of one or more of the following co-morbidities:
Insulin resistance or Type 2 diabetes; or
Poorly controlled hypertension (systolic blood pressure greater than 140 mm Hg or
diastolic blood pressure 90 mm Hg or greater, despite pharmacotherapy); or
Hyperlipidemia; or
Obstructive Sleep Apnea confirmed on polysomnography with an AHI or RDI of ≥
30; or
Evidence of Nonalcoholic Fatty Liver Disease (NAFLD), or
Idiopathic intracranial hypertension (pseudotumor cerebri)
and
o The individual must also receive an evaluation at, or in consultation with, a Multidisciplinary
center focused on the surgical treatment of severe childhood obesity; this may include
adolescent centers that have received accreditation by the Metabolic and Bariatric Surgery
Accreditation and Quality Improvement Program (MBSAQIP) or can demonstrate similar
programmatic components
Documentation Requirements
Updated list of CPT codes with associated documentation requirements; removed 43842
Updated list of required clinical information:
o Added:
Provider attestation of Asian ancestry, when applicable, if individual is of Asian descent
For staged bariatric surgery for Body Mass Index (BMI) > 50, include [surgical] plan
o Removed:
Diet history
o Replaced:
“Detailed weight and BMI history” with “preoperative evaluation that includes a detailed
weight and BMI history along with dietary and physical activity patterns
Psychological evaluation by a licensed behavioral health professional” with
psychosocial-behavioral evaluation by a licensed behavioral health professional”
Definitions
Added definition of:
Bariatric Surgery
Page 58 of 58
UnitedHealthcare Commercial and Individual Exchange Medical Policy
Effective 03/01/2024
Proprietary Information of UnitedHealthcare. Copyright 2024 United HealthCare Services, Inc.
Date
Removed definition of “Adolescent”
Applicable Codes
Removed CPT codes 43842 and 43999
Benefit Considerations
For Fully Insured Group Policies in Maryland Only
Revised criteria specified in the Code of Maryland Regulations (COMAR) 31.10.33.03B:
o Added criterion requiring the individual is age 18 years or older
o Replaced criterion requiring “documentation that dietary attempts at weight control have
been ineffective through completion of a structured diet program” with “completion of a
structured diet program”
Added criteria specified in the COMAR 31.10.33.04 requiring documentation of completion of a
structured diet program, including:
o Physician notes; or
o Notes of health care providers other than physicians; or
o Receipts of payment for a structured diet program; or
o Diet or weight loss logs from a structured diet program
Supporting Information
Updated Description of Services, Clinical Evidence, FDA, and References sections to reflect the
most current information
Instructions for Use
This Medical Policy provides assistance in interpreting UnitedHealthcare standard benefit plans. When deciding coverage,
the member specific benefit plan document must be referenced as the terms of the member specific benefit plan may
differ from the standard plan. In the event of a conflict, the member specific benefit plan document governs. Before using
this policy, please check the member specific benefit plan document and any applicable federal or state mandates.
UnitedHealthcare reserves the right to modify its Policies and Guidelines as necessary. This Medical Policy is provided for
informational purposes. It does not constitute medical advice.
This Medical Policy may also be applied to Medicare Advantage plans in certain instances. In the absence of a Medicare
National Coverage Determination (NCD), Local Coverage Determination (LCD), or other Medicare coverage guidance,
CMS allows a Medicare Advantage Organization (MAO) to create its own coverage determinations, using objective
evidence-based rationale relying on authoritative evidence (Medicare IOM Pub. No. 100-16, Ch. 4, §90.5
).
UnitedHealthcare may also use tools developed by third parties, such as the InterQual
®
criteria, to assist us in
administering health benefits. UnitedHealthcare Medical Policies are intended to be used in connection with the
independent professional medical judgment of a qualified health care provider and do not constitute the practice of
medicine or medical advice.