ArticlePDF Available

Effect of Reversible Intermittent Intra-abdominal Vagal Nerve Blockade on Morbid Obesity The ReCharge Randomized Clinical Trial

Authors:

Abstract and Figures

Importance Although conventional bariatric surgery results in weight loss, it does so with potential short-term and long-term morbidity.Objective To evaluate the effectiveness and safety of intermittent, reversible vagal nerve blockade therapy for obesity treatment.Design, Setting, and Participants A randomized, double-blind, sham-controlled clinical trial involving 239 participants who had a body mass index of 40 to 45 or 35 to 40 and 1 or more obesity-related condition was conducted at 10 sites in the United States and Australia between May and December 2011. The 12-month blinded portion of the 5-year study was completed in January 2013.Interventions One hundred sixty-two patients received an active vagal nerve block device and 77 received a sham device. All participants received weight management education.Main Outcomes and Measures The coprimary efficacy objectives were to 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. The primary safety objective was to determine whether the rate of serious adverse events related to device, procedure, or therapy in the vagal block group was less than 15%.Results In the intent-to-treat analysis, the vagal nerve block group had a mean 24.4% excess weight loss (9.2% of their initial body weight loss) vs 15.9% excess weight loss (6.0% initial body weight loss) in the sham group. The mean difference in the percentage of the excess weight loss between groups was 8.5 percentage points (95% CI, 3.1-13.9), which did not meet the 10-point target (P = .71), although weight loss was statistically greater in the vagal nerve block group (P = .002 for treatment difference in a post hoc analysis). At 12 months, 52% of patients in the vagal nerve block group achieved 20% or more excess weight loss and 38% achieved 25% or more excess weight loss vs 32% in the sham group who achieved 20% or more loss and 23% who achieved 25% or more loss. The device, procedure, or therapy–related serious adverse event rate in the vagal nerve block group was 3.7% (95% CI, 1.4%-7.9%), significantly lower than the 15% goal. The adverse events more frequent in the vagal nerve block group were heartburn or dyspepsia and abdominal pain attributed to therapy; all were reported as mild or moderate in severity.Conclusion and Relevance Among patients with morbid obesity, the use of vagal nerve block therapy compared with a sham control device did not meet either of the prespecified coprimary efficacy objectives, although weight loss in the vagal block group was statistically greater than in the sham device group. The treatment was well tolerated, having met the primary safety objective.Trial Registration clinicaltrials.gov Identifier: NCT01327976
Content may be subject to copyright.
Copyright 2014 American Medical Association. All rights reserved.
Effect of Reversible Intermittent Intra-abdominal Vagal Nerve
Blockade on Morbid Obesity
The ReCharge Randomized Clinical Trial
Sayeed Ikramuddin, MD; Robin P. Blackstone, MD; Anthony Brancatisano, MD, PhD; James Toouli, MD, PhD; Sajani N. Shah, MD; Bruce M. Wolfe, MD;
Ken Fujioka, MD; James W. Maher, MD; James Swain, MD; Florencia G. Que, MD; John M. Morton, MD; Daniel B. Leslie, MD; Roy Brancatisano, MD;
Lilian Kow, MD, PhD; Robert W. O’Rourke, MD; Clifford Deveney,MD; Mark Takata, MD; Christopher J. Miller,MS; Mark B. Knudson, PhD;
Katherine S. Tweden, PhD; ScottA . Shikora, MD; Michael G. Sarr, MD; Charles J. Billington, MD
IMPORTANCE Although conventional bariatric surgery results in weight loss, it does so with
potential short-term and long-term morbidity.
OBJECTIVE To evaluate the effectiveness and safety of intermittent, reversible vagal nerve
blockade therapy for obesity treatment.
DESIGN, SETTING, AND PARTICIPANTS A randomized, double-blind, sham-controlled clinical
trial involving 239 participants who had a body mass index of 40 to 45 or 35 to 40 and 1 or
more obesity-related condition was conducted at 10 sites in the United States and Australia
between May and December 2011. The 12-month blinded portion of the 5-year study was
completed in January 2013.
INTERVENTIONS One hundred sixty-two patients received an active vagal nerve block device
and 77 received a sham device. All participants received weight management education.
MAIN OUTCOMES AND MEASURES The coprimary efficacy objectives were to 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. The primary safety objective was to determine whether
the rate of serious adverse events related to device, procedure, or therapy in the vagal block
group was less than 15%.
RESULTS In the intent-to-treat analysis, the vagal nerve block group had a mean 24.4%
excess weight loss (9.2% of their initial body weight loss) vs 15.9% excess weight loss (6.0%
initial body weight loss) in the sham group. The mean difference in the percentage of the
excess weight loss between groups was 8.5 percentage points (95% CI, 3.1-13.9), which did
not meet the 10-point target (P= .71), although weight loss was statistically greater in the
vagal nerve block group (P= .002 for treatment difference in a post hoc analysis). At 12
months, 52% of patients in the vagal nerve block group achieved 20% or more excess weight
loss and 38% achieved 25% or more excess weight loss vs 32% in the sham group who
achieved 20% or more loss and 23% who achieved 25% or more loss. The device, procedure,
or therapy–related serious adverse event rate in the vagal nerve block group was 3.7% (95%
CI, 1.4%-7.9%), significantly lower than the 15% goal. The adverse events more frequent in
the vagal nerve block group were heartburn or dyspepsia and abdominal pain attributed to
therapy; all were reported as mild or moderate in severity.
CONCLUSION AND RELEVANCE Among patients with morbid obesity, the use of vagal nerve
block therapy compared with a sham control device did not meet either of the prespecified
coprimary efficacy objectives, although weight loss in the vagal block group was statistically
greater than in the sham device group. The treatment was well tolerated, having met the
primary safety objective.
TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT01327976
JAMA. 2014;312(9):915-922. doi:10.1001/jama.2014.10540
Editorial page 898
Supplemental content at
jama.com
Author Affiliations: Author
affiliations are listed at the end of this
article.
Corresponding Author: Charles J.
Billington, MD, Minneapolis VA
Medical Center, 1 Veterans Dr,
Minneapolis, MN 55417 (billi005
@umn.edu).
Research
Original Investigation
915
Copyright 2014 American Medical Association. All rights reserved.
Downloaded From: http://jama.jamanetwork.com/ by a Oregon Health & Science University User on 09/25/2014
Copyright 2014 American Medical Association. All rights reserved.
Illness imposed by obesity, especially by morbid obesity,has
inspired an array of treatment options, typically applied in
stepwise fashion. Although changes in food intake and ac-
tivity levels can be effective for some, the mean effect is
modest.
1
The most effective treatment, bariatric surgery, can
produce significant weight loss and improvementin health but
is associated with risks of morbidity and distortion of anatomy
that are unacceptable to some.
2
Medications can be helpful,
but their application is limited by adverse effects and reluc-
tance of insurance companies to cover them.
3
There is great
interest in the development of a device that could be as effec-
tive or nearly as effective as bariatric surgery but that has fewer
risks and that is less invasive. One such possibility is vagal
blockade using electrodes implanted through minimally in-
vasive laparoscopic surgery.
4
The vagus nerve is known to play a key role in satiety, me-
tabolism, and autonomic control in upper gastrointestinal track
function.
5
The EMPOWER
6
study, a recent randomized trial
testing vagal blockade, found substantial weight loss, but the
difference in weight loss between treatmentand control groups
was not significant. However, treatment group participants
who received at least 12 hours of vagal block therapy a day
achieved the level of weight loss anticipated in the design. Fur-
thermore, a significant dose response of weight loss in rela-
tion to hours of device use for both groups coupled with the
possibility that control patients may have received partial va-
gal blockade through low-energy safety or device checks con-
founded the interpretation of the trial’s results.
6
These find-
ings required a new study to determine the efficacy of vagal
nerve block therapy with a treatment device that consis-
tently delivered at least 12 hours of therapy a day and a sham
control device that had no possibility of delivering therapy. The
ReCharge Study, a multicenter,randomized, double-blind trial
that addressed these design limitations, is the focus of this ar-
ticle.
Methods
Participants
Participants at 2 sites in Australia and 8 sites in the UnitedStates
were eligible for inclusion in the study if their body mass in-
dex (BMI, calculated as weight in kilograms divided by height
in meters squared) was 40 to 45 (class III obesity) or 35 to 40
(class II obesity) and had 1 or more obesity-related comorbid
conditions, including type 2 diabetes, hypertension (systolic
blood pressure ≥140 mm Hg or diastolic blood pressure ≥90 mm
Hg), dyslipidemia (total cholesterol ≥200 mg/dL or low-
density lipoprotein [LDL] ≥130 mg/dL), sleep apnea syn-
drome, or obesity-related cardiomyopathy. The participation
of those with type 2 diabetes was limited so that potential
weight loss limiting effects of diabetes and its treatment did
not affect the study outcomes. A detailed description of in-
clusion and exclusion criteria can be found in Supplement 1.
All participants provided written informed consent, and the
institutional review boards at each study site approvedall study
protocols. (To convert LDL from mg/dL to mmol/L, multiply
by 0.0259.)
Study Design
This multicenter, randomized, double-blind trial compared
active vagal nerve blocking with an implanted sham device.
The randomization allocation was 2:1 for active vagal nerve
block to sham devices in permuted block sizes of 3 or 6,
stratified by study site and type 2 diabetes status (Figure 1).
To ensure comparability between groups with respect to
potential weight loss, participants with type 2 diabetes were
limited to 10% of enrollment at each site; their randomiza-
tion was not stratified by site. Randomization to active or
sham group occurred once patients were admitted to the
hospital. The participant, sponsor, and follow-up staff at the
clinical site were blinded to treatment assignment. The sur-
geons and surgery support staff could not be blinded, so
their interaction with participants after placing the device
was limited.
Implanting vagal nerve block therapy electrodes requires
standard laparoscopic techniques and general anesthesia.
4
The
anterior and posterior vagus nerves were identified and dis-
sected free at the gastroesophageal junction. Customized elec-
trodes were placed around the nerves and then secured with
sutures.
4
These electrodes were connected to a transcutane-
ously rechargeable neuroregulator placed in a subcutaneous
pocket on the thoracic side wall. Both treatment and control
devices were 8.6 cm in diameter, 7.1 cm in width, and 1.6 cm
thick.
Participants in the sham group were implanted similarly
but with a neuroregulator that dissipated charges intoan elec-
tronic circuit within the device without leads. Their proce-
dure consisted of the same number of skin incisions as vagal
nerve block participants without peritoneal penetration.
Neuroregulators were programmed to deliver a charge
for at least 12 hours daily. An external programming device
allowed the investigator to increase the amplitude in both
active and sham devices at scheduled intervals during
follow-up visits in accordance with the protocol to the
desired amplitude of 6 to 8 mA, depending on patient toler-
ance. The active or sham neuroregulator battery required
recharging at least twice a week for approximately 30 min-
utes to 90 minutes using an external charging device. Each
charging session was recorded by the device and data were
monitored by the clinic staff for safety and recharging
adherence.
Data Collection and Study Conduct
Data collected prior to randomization included height, weight,
medical histories, and medication usage. Follow-up visits
following randomization were weekly for the first month,
then every 2 weeks between weeks 4 and 12, and monthly
thereafter from 3 to 12 months to collect primary and second-
ary measures including weight and adverse events. All par-
ticipants attended a weight management program, consist-
ing of 17 face-to-face educational counseling sessions that
ranged from 15 minutes to 45 minutes and discussed such top-
ics as healthful food choices, physical fitness, and social sup-
port. No diet or exercise regimen was prescribed.
Coprimary efficacy objectives for the study were based on
the percentage excess weight loss, which is calculated using
Research Original Investigation Intra-abdominal Vagal Nerve Blockade for Morbid Obesity
916 JAMA September 3, 2014 Volume 312, Number 9 jama.com
Copyright 2014 American Medical Association. All rights reserved.
Downloaded From: http://jama.jamanetwork.com/ by a Oregon Health & Science University User on 09/25/2014
Copyright 2014 American Medical Association. All rights reserved.
the formula: percent of excess weight loss = 100% × [weight
loss/excess body weight at implant placement]. Excess body
weight was defined as the difference between weight at im-
plantation and the ideal body weight to achieve a BMI of 25.
The first efficacy objective was based on a mean percent ex-
cess weight loss comparison between groups at a superiority
margin of 10% (super-superiority). The second efficacy objec-
tive was to demonstrate that 55% of patientsin the vagal ner ve
block group would achieve 20% excessweight loss and that 45%
would achieve 25% excess weight loss.
Safety data were monitored by an independent data and
safety monitoring board that was chartered to meet atleast an-
nually and adjudicated all serious adverse events. No interim
analyses or early stopping boundaries were used during this
study. Serious adverse events were defined as any untoward
event that led to death or serious deterioration in the health
of the participant, including prolonged hospitalization. The pri-
mary safety objective was to demonstrate that the serious ad-
verse event rate related tothe dev ice, the implantor rev ision,
or the therapy was less than 15% in the vagal nerve block group.
This single-group comparison with a performance criterion was
chosen in consultation with the US Food and Drug Adminis-
tration (FDA) to demonstrate a lowerserious adverse event rate
than studies used to support FDA approval for the laparo-
scopic adjustable gastric band. The site investigator deter-
mined the severity and attribution of adverse eventsthat were
not serious.
Statistical Analysis
The assumptions of the trial were that, on average, patients
in the vagal nerve block group would achieve 25% excess
weight loss and the sham group would achieve 5% excess
weight loss at 12 months. These estimates were based on
experience with the dose-response effect observed in the
EMPOWER trial and the 12-month weight loss observed in
the VBLOC DM2 study among participants with type 2
diabetes.
6,7
In EMPOWER, participants with fewer than 6
hours of device use per day in the first 12 months achieved a
mean 5% excess weight loss, and those who received at least
12 hours of therapy per day achieved a mean 29% of excess
weight loss.
6
In the observational VBLOC DM2 study, par-
ticipants achieved an average of 25% of excess weight loss.
7
Therefore, it was determined that a minimum of 233 partici-
pants (allocated 2:1) would be required to achieve 85% sta-
tistical power to detect a 20-point mean difference between
groups with a superiority margin of 10 points at 12 months,
accounting for up to 15% attrition.
Data were analyzed according to the intention-to-treat
(ITT) principle, in which all participants were analyzed as ran-
domized. Per protocol, missing 12-month values for the per-
centage of excess weight lost were imputed using the last-
observation-carried-forward method. We also conducted a post
hoc multiple imputation analysis with 50 imputation data sets
using multivariate normal regression for 26 missing 12-
month values for the percentage of excess weight loss using
Figure 1. CONSORT Diagram of Participants in the ReCharge Study
420 Patients assessed for eligibility
181 Excluded
30 BMI criteria not met
25 Failed psychiatric evaluation
12 Medical condition making
subject unfit for surgery
10 Unable to complete study visits
9Has history of hiatal hernia
7Diabetes inclusion not met
88 Refused participation/other
239 Randomized
162 Included in the primary analysis 77 Included in the primary analysis
147 Had 12-month data
10 Did not have 12-mo data
7Missed follow-up visit
2Lost to follow-up
1Withdrew (adverse event)
66 Had 12-month data
10 Did not have 12-mo data
4Missed follow-up visit
3Withdrew (because of subject decision)
3Withdrew (adverse event)
162 Randomly assigned to weight
management program and active vagal
nerve block device
157 Received intervention as
randomized
5Not implanted because of
intraoperative exclusion and
withdrawn
77 Randomly assigned to weight
management program and sham
control device
75 Received intervention as
randomized
2Did not receive the sham device
as randomized
1Received active vagal nerve
block device
1Not implanted for subject decision
and withdrawn
BMI indicates body mass index.
Intra-abdominal Vagal Nerve Blockade for Morbid Obesity Original Investigation Research
jama.com JAMA September 3, 2014 Volume 312, Number 9 917
Copyright 2014 American Medical Association. All rights reserved.
Downloaded From: http://jama.jamanetwork.com/ by a Oregon Health & Science University User on 09/25/2014
Copyright 2014 American Medical Association. All rights reserved.
treatment group, age, sex, race, diabetic status, weight at im-
plantation, and site as predictors.
The primary safety objective was analyzed using a 1-sided
exact binomial test at the 0.025 significance level. Superior-
ity of the mean percentage of excess weight lost was com-
pared between groups using a 1-sided ttest at the 0.025 sig-
nificance level with a 10-point superiority margin. Post hoc
testing with a 2-sided ttest was used to compare weight loss
between groups with no superiority margin. In addition, the
odds ratio comparing the percentage of excess weight loss
thresholds by treatment group are presented as post hoc ex-
ploratory analyses. Statistical analyses were conducted using
SAS version 9.3 (SAS Institute Inc).
Results
Baseline Characteristics
The screening and randomization procedure is summarized in
Figure 1. Of the 239 participants, 162 to were randomized from
May 16 to December 27, 2011, to receive the vagal nerve block
and 77 to receive the sham device. Twelve-month visits were
completed by January 4, 2013. Participant characteristics at
baseline are summarized in Table 1.
Participant Disposition
Seven participants did not receive the implants as random-
ized. One participant randomized to the sham control group
changed his/her mind immediately before the procedure and
withdrew. Another participant in the sham group was anes-
thetized in the operating room, but because no sham kits were
available, an active device was implanted. Five participants in
the vagal nerve block group did not receive the implant due
to intraoperative exclusions (ie, hiatal hernia >5 cm, previous
Nissen fundoplication, food in stomach, cirrhosis or hepatitis
C, and inability to locate anterior vagal nerve) and were with-
drawn.
Followingimplantation, 6 patients in the sham group with-
drew (3 for patient decision, 3 for adverse events) as did 3 pa-
tients in the vagal nerve block group (2 lost to follow-up, 1 for
an adverse event). The completion rate of the 12-month visit
was 91% in the vagal nerve block group and 86% in the sham
control group.
During the 12-month blinded study period, 8 patients
(4.9%) in the vagal nerve block group had undergone a revi-
sion procedure to reposition or replace the neuroregulator; no
revisions were required in the sham group. Five participants
(3.1%) in the vagal nerve block group and 8 (10.4%) in the sham
group had the device removed by 12 months. Two patients in
the vagal nerve block did so because 1 had experienced pain
at the neuroregulator site and the other, heartburn. Of the 4
patients in the sham group, 1 had pain at the neuroregulator
site, 1 needed to undergo magnetic resonance imaging to in-
vestigate causes for shoulder pain, 1 had worsening of irri-
table bowel syndrome symptoms, and 1 had breast cancer.
Three patients in the vagal nerve block group and 4 in the sham
group asked to have the devices removed.
Efficacy
At 12 months in the ITT population (Table 2), the mean per-
centages of excess weight loss was 24.4% in the vagal nerve
block group and 15.9% in the sham group with a mean differ-
ence of 8.5 percentage points (95% CI, 3.1-13.9). This differ-
ence did not meet the primary efficacy objective of achieving
superiority with a 10 percentage-point margin (P=.71), al-
though weight loss was statistically greater in the vagal nerve
block group (P= .002 for treatment difference in post hoc test-
ing). When analyzed using multiple imputation analysis, the
mean percentage of excess weight loss was 26.1% in the vagal
nerve block group and 16.9% in the sham group with a mean
difference of 9.2 percentage points (95% CI, 2.7-15.6). Figure 2
illustrates the percentage of excess weight loss as observed
through 12 months. The mean percentage of initial body weight
loss at 12 months in the ITT population was 9.2% in the vagal
nerve block group and 6.0% in the sham group for a mean dif-
ference of 3.2 percentage points (95% CI, 1.1-5.2).
At 12 months, 52% of participants in the vagal nerve block
group achieved at least 20%; and 38%, at least 25% of excess
weight loss (Table 2), which did not meet the primary effi-
cacy objective performance goals of at least 55% of partici-
pants achieving a 20% excess weight loss and 45% achieving
a 25% excess weight loss. Table 2 presents the percentage of
participants reaching weight loss thresholds from 20% to 50%
as well as the post hoc exploratory odds ratios showing higher
odds of participants in the vagal nerve block group achieving
those thresholds than participants in the sham group.
Table 1. Baseline Characteristics byTreatment Group
Vagal Nerve Block
(n=162)
Sham
(n=77)
Demographics
Women, No. (%) 141 (87) 62 (81)
Age, mean (SD), y 47 (10) 47 (9)
Race, No. (%)
White 149 (92) 73 (95)
Black 8 (5) 3 (4)
American Indian 2 (1) 1 (1)
Asian 1 (1) 1 (1)
Hawaiian/Pacific Islander 1 (1) 0
Body size measures when device
placed, mean (SD)
Height, cm 166 (8) 168 (9)
Weight, kg 113 (13) 116 (14)
Body mass index 41 (3) 41 (3)
Excess weight, kg
a
44 (9) 45 (10)
Waist circumference, cm 121 (12) 123 (11)
Type 2 diabetes mellitus, No. (%) 9 (6) 6 (8)
Hypertension, No. (%) 63 (39) 32 (42)
Dyslipidemia, No. (%) 91 (56) 46 (60)
Obstructive sleep apnea, No. (%) 33 (20) 23 (30)
a
Excess weight was calculated as the difference between the weight at the time
of implantation and the ideal body weight corresponding to a body mass index
of 25, which is calculated as weight in kilograms divided by height in meters
squared.
Research Original Investigation Intra-abdominal Vagal Nerve Blockade for Morbid Obesity
918 JAMA September 3, 2014 Volume 312, Number 9 jama.com
Copyright 2014 American Medical Association. All rights reserved.
Downloaded From: http://jama.jamanetwork.com/ by a Oregon Health & Science University User on 09/25/2014
Copyright 2014 American Medical Association. All rights reserved.
Safety
The primary safety end point, the rate of serious adverse events
that were related directly to the device, implantation or revi-
sion, or therapy in the vagal nerve block group, was 3.7% (95%
CI, 1.4%-7.9%); therefore, the primary safety objective was met
(P<.001). Two serious adverse events were for neuroregula-
tor malfunction requiring replacement and 1 was pain at the
neuroregulator site following a large weight loss (80% of ex-
cess weight loss), which required repositioning; these partici-
pants were hospitalized overnight and discharged. One par-
ticipant developed atelectasis, which prolonged hospitalization
by 2 days. One patient developed emesis and needed a hiatus
hernia repair and was discharged 2 days following repair. One
participant had gallbladder disease, which was determined to
be possibly related to therapy due to weight loss.
Nine serious adverse events were related to general intra-
abdominal surgery. Six were for nausea resulting in pro-
longed hospitalization of at least 1 daylonger than what is typi-
cal for the procedure. One was for intraoperative oozing, which
resulted in overnight hospitalizationand hemodynamic moni-
toring. A participant who had not received an implant due to
an intraoperative exclusion for cirrhosis was hospitalized be-
cause of a complicated liver biopsy.One partic ipanthad a mod-
erate ileus of the stomach, was treated with pain medica-
tions, and was discharged on the second postoperative day.
Combining the serious adverse events thatwere related to intra-
abdominal surgery with those related to vagal nerve block, the
rate was 8.6% (95% CI, 4.8% to 14.1%). eTable 1 in Supplement
2 lists all serious adverse events by the adjudicated cause of
the event.
The most common adverse events related to treatment are
reported in Table 3. Pain at the neuroregulatorsite was the most
common event in both groups. Ninety-six percent of these
events in the vagal nerve block group were deemed to be mild
or moderate, although a revision was performed in 3 cases and
an explant in 1 due to pain. Heartburn and dyspepsia, abdomi-
nal pain, other or nonspecific pain, dysphagia, and eructa-
tion or belching were reported more frequently by partici-
pants in the vagal nerve block group and were attributed by
investigators to be primarily related totherapy. All events were
Figure 2. Weight Loss From Baseline as ObservedWithout Imputation
0
–5
–10
–15
–20
–25
–30
0
–2
–4
–6
–8
–10
–12
Mean Percentage of Excess
Weight Loss, BMI Method
Mean Percentage of Initial
Body Weight Loss
Visit Month
0
162
77
3
151
71
6
149
69
9
135
60
12
147
66
No. of patients
Vagal nerve block
Vagal nerve block
Sham
Sham Error bars indicate 95% CIs. BMI
indicates body mass index.
Table 2. Twelve-Month Percentage Excess Weight Loss
Mean (95% CI)
Difference (95% CI)
Vagal Nerve Block
(n=162)
Sham
(n=77)
Continuous outcomes
Percentage of excess weight loss
LOCF 24.4 (20.8-28.1) 15.9 (11.9-19.9) 8.5 (3.1-13.9)
Multiple Imputation 26.1 (22.2-29.9) 16.9 (11.6-22.2) 9.2 (2.7-15.6)
Categorical Outcomes No. (%) of Patients Odds Ratio (95% CI)
Percentage of excess weight loss
threshold, LOCF
20 85 (52) 25 (32) 2.3 (1.3-4.1)
25 62 (38) 18 (23) 2.0 (1.1-3.8)
30 49 (30) 14 (18) 2.0 (1.0-3.8)
35 42 (26) 7 (9) 3.5 (1.5-8.2)
40 35 (22) 4 (5) 5.0 (1.7-14.7)
45 30 (19) 3 (4) 5.6 (1.7-19.0)
50 24 (15) 1 (1) 13.2 (1.8-99.6) Abbreviation: LOCF, last observation
carried forward.
Intra-abdominal Vagal Nerve Blockade for Morbid Obesity Original Investigation Research
jama.com JAMA September 3, 2014 Volume 312, Number 9 919
Copyright 2014 American Medical Association. All rights reserved.
Downloaded From: http://jama.jamanetwork.com/ by a Oregon Health & Science University User on 09/25/2014
Copyright 2014 American Medical Association. All rights reserved.
adjudicated as mild or moderate in severity, and most were re-
solved with alteration to the therapy algorithm. Nausea was
reported more frequently by participants in the vagal nerve
block group, which was expected given that sham implantpro -
cedures had no peritoneal penetration. Eighty-six percent of
nausea events were mild or moderate.
Discussion
In this study of patients with morbid obesity, the percentage
of excess weight loss among participants treated with vagal
block did not meet either of the coprimary efficacy objec-
tives, although weight loss in the intervention group was sta-
tistically greater than in the sham group. The vagal nerve block
group nearly achieved the 25% mean excess weight loss re-
sponse predicted in the trial design, but the sham group re-
sponse was 3 times greater than predicted. Fifty-two percent
of participants in the vagal nerve block group achieved 20%
and 38% achieved 25% excessweight loss; however, those were
lower percentages than the study’s objective of 55% achiev-
ing a 20% loss and 45% achieving a 25% loss. Weight loss was
achieved with a low rate of serious adverse events related to
device implantation or function, and most adverse events were
mild or moderate in severity.
This is 1 of 3 reported trials testing devices to treat obe-
sity in which a randomized, sham-control method was used.
6-8
To our knowledge, it is the first to report a statistically greater
weight loss among treated patients than among those in the
control group. Previous trials with an earlier version of the va-
gal nerve block device
6
and with an implantable gastric
stimulator
8
did not produce statistically different treatment
effects. Randomized placebo controlled trials have been the
standard in obesity trials involving medication and lifestyle in-
tervention, but up to this time, obesity device trials, such as
those for laparoscopic adjustable gastric band, have rarely in-
cluded a sham comparator or a 10%-point superiority mar-
gin. Consequently, the trial used an unusually strong design
to study this obesity-treatment device.
It is likely that the sham group weight loss was due to a
placebo effect of surgery, daily self-monitoring reinforced by
interaction with the sham device to recharge the battery, and
participation in the weight management program. Placebo con-
trol groups in recent obesity medication studies have shown
1% to 2% initial body weight loss.
9-11
Lifestyle counseling with-
out medication or meal replacements produced a 3.5% initial
body weight loss at 12 months in a controlled trial.
12
The 6%
initial body weight loss seen in the sham group in this study
is similar to the sham effect in the previous vagal nerve block
trial (EMPOWER)
6
and to the control group in the recent SHAPE
trial of an implantable gastric stimulator.
8
At the time this trial
was designed, weight loss in the previous vagal nerve block
sham treatment was thought to be due to inadvertent active
treatment. In the EMPOWER study, there was a relationship
between hours of use and weight loss, regardless of assign-
ment to active or control groups.
6
Sham surgeries are known to have substantial placebo ef-
fects in other contexts, such as arthroscopic knee surgery,
13,14
vertebroplasty for back pain,
15,16
and internal mammary ar-
tery ligation for angina.
17
The sham effect was sufficient in each
of these cases to result in a conclusion of ineffectiveness for
the intervention. A recent review article highlights the large
sham effects observed with surgical interventions.
18
Further-
more, the structure of a clinical weight loss trial imposes more
than usual behavioral changes, notably in the form of self-
monitoring of behavior and weight, which may have impor-
tant effects.
19
The results in the ReCharge trial are therefore
of heightened interest because the post hoc analysis showed
greater weight loss in the active group despite a robust sham
effect.
Adverse events in this trial were less severe than those as-
sociated with conventional bariatric surgical procedures, pri-
Table 3. AdverseEvents Related to Treatment Through 12 Months
Adverse Event
Vagal Nerve Block
(n=162)
Sham
(n=77)
No. (%) Patients No. Events No. (%) Patients No. Events
Pain, neuroregulator site 61 (38) 73 32 (42) 35
Heartburn/dyspepsia 38 (23) 42 3 (4) 3
Pain, other 37 (23) 42 0 0
Pain, abdominal 20 (12) 26 2 (3) 2
Nausea 11 (7) 14 1 (1) 1
Dysphagia 13 (8) 13 0 0
Eructation/belching 13 (8) 13 0 0
Incision pain 12 (7) 13 7 (9) 7
Chest pain 9 (6) 9 2 (3) 2
Cramps, abdominal 7 (4) 7 0 0
Wound redness or irritation 7 (4) 7 5 (6) 5
Appetite increased 5 (3) 6 2 (3) 3
Constipation 6 (4) 6 7 (9) 8
Emesis/vomiting 5 (3) 6 2 (3) 2
Bloating, abdominal 5 (3) 5 1 (1) 2
Headache 5 (3) 5 2 (3) 2
Only adverse events attributed by the
investigator to the device, procedure,
or therapy that occurred in at least
3% of vagal nerve block group
participants are displayed.
Research Original Investigation Intra-abdominal Vagal Nerve Blockade for Morbid Obesity
920 JAMA September 3, 2014 Volume 312, Number 9 jama.com
Copyright 2014 American Medical Association. All rights reserved.
Downloaded From: http://jama.jamanetwork.com/ by a Oregon Health & Science University User on 09/25/2014
Copyright 2014 American Medical Association. All rights reserved.
marily mild or moderate events related to therapy. Recent re-
ports of uncontrolled clinical trials and registry data provide
comparative safety data on laparoscopic adjustable gastric
band.
20-22
Overall, the rate of device-related adverse eventsin
the Helping Evaluate Reduction in Obesity (HERO)registr y ap-
pears to be 4.7%.
20
Phillips et al
21
reported adverse events that
included dysphagia (9.4%), gastroesophageal reflux (19.2%),
and vomiting (40.6%) and a reoperation rate of 15.2%.
Cobourn et al
20
reported a low rate of band-related complica-
tions, but a reoperation rate of 15.2%. Late complications for
laparoscopic adjustable gastric band include band slippage and
pouch dilation, which occurs in up to 20% of patients, and band
erosion occurring in up to 4% of patients.
23
However, adverse
events of vagal nerve block have not yet been reportedfollow-
ing widespread use. Rates of adverse events often increase
when therapies are used in general populations.
This study has limitations including the demographics of
the study participants, which were primarily white women, so
inference to other groups must be made with care. Further-
more, the study population had a low rate of diabetes, a com-
mon comorbidity of significant obesity, and a low rate of other
metabolic complications such as hypertension and dyslipid-
emia. The study focused on obesity in the BMI range of 35 to
45, so application of the treatment to individuals with higher
BMI will need careful consideration.
Conclusions
Among patients with morbid obesity, the use of vagal nerve
block therapy compared with a sham control device did not
meet either of the coprimary prespecified efficacy objec-
tives, although the intervention group had statistically greater
weight loss than the sham control group. The treatment met
the primary safety objective and was well tolerated. Addi-
tional studies are needed to compare effectiveness of vagal
nerve block with other obesity treatments and to assess long-
term durability of weight loss and safety.
ARTICLE INFORMATION
Author Affiliations: Department of Surgery,
University of Minnesota, Minneapolis (Ikramuddin,
Leslie); University of Minnesota, Minneapolis;
Scottsdale Healthcare Bariatric Center, Scottsdale,
Arizona (Blackstone, Swain); Institute of Weight
Control, Sydney, Australia (A. Brancatisano,
R. Brancatisano); Adelaide Bariatric Centre,
Adelaide, Australia (Toouli,Kow); Department of
Surgery, Tufts Medical Center, Boston,
Massachusetts (Shah); Department of Surgery,
Oregon Health & Science University,Portland
(Wolfe, Deveney); Scripps Clinic, San Diego,
California (Fujioka, Takata); Division of General
Surgery, Virginia Commonwealth University,
Richmond (Maher); Department of
Gastroenterologic and General Surgery,Mayo Clinic
Rochester,Rochester, Minnesota (Que, Sarr);
Division of General Surgery, StanfordUniversity
School of Medicine, Stanford, California (Morton);
Department of Surgery, University of Michigan and
Ann Arbor VA Hospital (O’Rourke);North American
Science Associates, Minneapolis, Minnesota
(Miller); EnteroMedics Inc, St Paul, Minnesota
(Knudson, Tweden, Shikora);Division of General
and Gastrointestinal Surgery,Brigham and Women’s
Hospital, Boston, Massachusetts (Shikora); Division
of Endocrinology and Diabetes, Minneapolis VA
Medical Center and University of Minnesota,
Minneapolis (Billington).
Author Contributions: Drs. Billington and Sarr had
full access to all of the data in the study and take
responsibility for the integrity of the data and the
accuracy of the data analysis.
Study concept and design: Toouli,Kow, Knudson,
Tweden, Shikora,Sarr, Billington.
Acquisition, analysis, or interpretation of data: All
authors.
Drafting of the manuscript: Miller,Knudson,
Tweden, Shikora,Sarr, Billington.
Critical revision of the manuscript for important
intellectual content: Ikramuddin, Blackstone, A
Brancatisano, Toouli,Wolfe, Fujioka, Maher, Swain,
Que, Morton, Leslie, R Brancatisano, Kow,
O’Rourke, Deveney, Takata,Miller, Knudson,
Tweden, Shikora,Sarr, Billington.
Statistical analysis: Miller.
Obtained funding: Knudson.
Administrative, technical, or material support: All
authors.
Study supervision: Ikramuddin, Blackstone, A
Brancatisano, Toouli,Shah, Wolfe, Fujioka, Maher,
Swain, Que, Morton, Tweden,Shikora, Sarr,
Billington.
Conflict of Interest Disclosures: All authors have
completed and submitted the ICMJE Form for
Disclosure of Potential Conflicts of Interest. Dr
Ikramuddin reported that he serves on the advisory
board for Novo Nordisk Inc and Medica; has served
as a consultant for Metamodix Inc and on an expert
panel for OptumHealth; and has received grant
support from USGI Medical Inc, ReShape Medical,
and Covidien. Dr Brancatisano reported that he is a
consultant for EnteroMedics. Mr Miller reports
consulting support from EnteroMedics, Inc. Dr
Fujioka reported receiving personal fees from
EnteroMedics for help and consulting with
European device approval. Dr Swain reports
personal fees from surgical proctoring for
Enteromedics. Dr Knudson reported that he is the
chief executive officer of EnteroMedics and
codeveloped the patented vagal nerve blockade
device, which is owned by EnteroMedics. Dr
Tweden reported that she is an employee of
EnteroMedics and codeveloped the patented vagal
nerve blockade device, which is owned by
EnteroMedics. Dr Sarr reported that he is a
consultant for EnteroMedics. Dr Billington reported
that he is a consultant for EnteroMedics and
NovoNordisk and has received pgrant support from
Covidien.
Funding/Support: The ReCharge Study was
supported by EnteroMedics Inc, St Paul, Minnesota.
Role of the Funder/Sponsor:EnteroMedics Inc
was involved in the design and conduct of the
study,site selec tion, database management. The
sponsor provided funding to the clinical sites for
patient enrollment, core laboratory analyses,
clinical events adjudications, and database entry.
The sponsor provided fees to North American
Science Associates for independent statistical
analyses. Two sponsor representatives (Knudson
and Tweden) were allowedto review and
participate in the critical revision of the manuscript
prior to submission.
National Primary Investigators: Michael G. Sarr,
MD, and Charles J. Billington, MD.
Study and Weight Management Coordinators:
University of Minnesota: Bridget Slusarek, Elsie
Waddick, Shannon Johnson, and Nikolaus Rasmus;
Scottsdale Bariatric Center: Melisa Celaya, Heather
Lane, Samantha Easterly,Cassie Truran, Arrin
Larson, Dorrie Wilson, Katherine Emershad,
Jennifer Childress, and Angelia Seitz; Institute of
Weight Control, Sydney: RadhikaButala, Gre tel
Young, Ruth Hutchinson, Kerry McCurley, and
Geane Sharman; Adelaide Bariatric Centre,
Adelaide: Jane Collins and Fiona McDonald; Tufts
Medical Center: Ann Marie Melanson, Meghan
Ariagno, Danielle DeMarco, and Jillian Regan;
OHSU: Wencesly Paez, Chad Sorenson, Shannon
Rentz, Gloria Scalzo, and Tracy Severson; Scripps
Clinic: Jennifer Wagner,Mary Collard, Sandy Grad,
Cindy Galm, and Melissa Susak; VCU: Donna
Neatrour, Jill Meador, Sakita Sistrun, Melanie
Wiggins, Carly Sopko, and Leah Loomis; Mayo
Clinic: Amy Reynolds, Joe Hockert, Sue Starkson,
Margaret Gall, and Debbie Dixon; and Stanford:
Dana Schroeder and Kristine Birge.
Data and Safety Monitoring Board and Clinical
Events Committee Members: James Freston,MD,
(chair) Department of Medicine, University of
Connecticut School of Medicine, Farmington; Daniel
Bessesen, MD, Division of Endocrinology,University
of Colorado Denver,Denver; Miguel Herrera, MD,
Division of General Surgery, UniversidadNacional
Autónoma de México at Instituto Nacional de la
Nutrición Salvador Zubirán, Mexico City; Melissa
Martinson, PhD, Division of Health Policy and
Management, School of Public Health, University of
Minnesota, Minneapolis; Frank Moody, MD,
Department of Surgery, University of Texas Health
Science Center, San Antonio.
REFERENCES
1. Wing RR, Bolin P, Brancati FL, et al; The Look
AHEAD Research Group. Cardiovascular effects of
intensive lifestyle intervention in type 2 diabetes.
N Engl J Med. 2013;369(2)145-154.
Intra-abdominal Vagal Nerve Blockade for Morbid Obesity Original Investigation Research
jama.com JAMA September 3, 2014 Volume 312, Number 9 921
Copyright 2014 American Medical Association. All rights reserved.
Downloaded From: http://jama.jamanetwork.com/ by a Oregon Health & Science University User on 09/25/2014
Copyright 2014 American Medical Association. All rights reserved.
2. Picot J, Jones J, Colquitt JL, et al The clinical
effectiveness and cost-effectiveness of bariatric
(weight loss) surgery for obesity: a systematic
review and economic evaluation. Health Technol
Assess. 2009;13(41):1-190, 215-357, iii-iv.
3. Bray GA. Medications for weight reduction. Med
Clin North Am. 2011;95(5):989-1008.
4. 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.
5. Berthoud H-R. Vagal and hormonal gut-brain
communication: from satiation to satisfaction.
Neurogastroenterol Motil. 2008;20(suppl 1):64-72.
6. Sarr MG, Billington CJ, Brancatisano R, et al;
EMPOWER Study Group. The EMPOWER study:
randomized, prospective, double-blind, multicenter
trial of vagal blockade to induce weight loss in
morbid obesity. Obes Surg.2012;22(11):1771-1782.
7. Shikora S, Toouli J, Herrera MF,et al. Vagal
blocking improves glycemic control and elevated
blood pressure in obese subjects with type 2
diabetes mellitus. J Obes. 2013;2013:245683.
8. 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;5(1):31-37.
9. Smith SR, Weissman NJ, Anderson CM, et al;
Behavioral Modification and Lorcaserin for
Overweight and Obesity Management (BLOOM)
Study Group. Multicenter,placebo-controlled trial
of lorcaserin for weight management. N Engl J Med.
2010;363(3):245-256.
10. Allison DB, Gadde KM, Garvey WT,et al.
Controlled-release phentermine/topiramate in
severely obese adults: a randomized controlled trial
(EQUIP). Obesity (Silver Spring). 2012;20(2):330-342.
11. Apovian CM, Aronne L, Rubino D, et al; COR-II
Study Group. A randomized, phase 3 trial of
naltrexone SR/bupropion SR on weight and
obesity-related risk factors (COR-II). Obesity (Silver
Spring). 2013;21(5):935-943.
12. Wadden TA, VolgerS, Sarwer DB, et al. A
two-year randomized trial of obesity treatment in
primary care practice. N Engl J Med. 2011;365(21):
1969-1979.
13. Moseley JB, O’Malley K, Petersen NJ, et al. A
controlled trial of arthroscopic surgery for
osteoarthritis of the knee. N Engl J Med. 2002;347
(2):81-88.
14. Sihvonen R, Paavola M, Malmivaara A, et al;
Finnish Degenerative Meniscal Lesion Study
(FIDELITY) Group. Arthroscopic partial
meniscectomy versus sham surgery for a
degenerative meniscal tear.N Engl J Med. 2013;369
(26):2515-2524.
15. Buchbinder R, Osborne RH, Ebeling PR, et al. A
randomized trial of vertebroplasty for painful
osteoporotic vertebral fractures. N Engl J Med.
2009;361(6):557-568.
16. Kallmes DF,Comstock BA, Heager tyPJ, et al. A
randomized trial of vertebroplasty for osteoporotic
spinal fractures. N Engl J Med. 2009;361(6):569-579.
17. Miller FG. The enduring legacy of
sham-controlled trials of internal mammary artery
ligation. Prog Cardiovasc Dis. 2012;55(3):246-250.
18. Birch S. A review and analysis of placebo
treatments, placebo effects, and placebo controls
in trials of medical procedures when sham is not
inert. J Altern Complement Med. 2006;12(3):303-310.
19. Burke LE, Wang J, Sevick MA. Self-monitoringin
weight loss: a systematic review of the literature.
JAmDietAssoc. 2011;111(1):92-102.
20. Cobourn C, Degboe A, Super PA, et al. Safety
and effectiveness of LAP-BAND AP System: results
of Helping Evaluate Reduction in Obesity (HERO)
prospective registry study at 1 year.J Am Coll Surg.
2013;217(5):907-918. doi:10.1016/j.jamcollsurg.2013
.06.01 0.
21. Phillips E, Ponce J, Cunneen SA, et al Safety and
effectiveness of Realize adjustable gastric band:
3-year prospective study in the United States. Surg
Obes Relat Dis. 2009;5(5):588-597.
22. Cunneen SA, Brathwaite CEM, Joyce C, et al
Clinical outcomes of the REALIZE adjustable gastric
band-C at one year in a US population. Surg Obes
Relat Dis. 2012;8(3):288-295.
23. Hamdan K, Somers S, Chand M. Management
of late postoperative complications of bariatric
surgery. Br J Surg. 2011;98(10):1345-1355.
Research Original Investigation Intra-abdominal Vagal Nerve Blockade for Morbid Obesity
922 JAMA September3, 2014 Volume 312, Number 9 jama.com
Copyright 2014 American Medical Association. All rights reserved.
Downloaded From: http://jama.jamanetwork.com/ by a Oregon Health & Science University User on 09/25/2014
... The vagus nerve has an important role in satiety, metabolism and autonomic control in upper gastrointestinal function [7]. Targeted therapy of the vagus nerve may lead to an improvement in obesity-related conditions such as diabetes mellitus type 2 with an associated low rate of major complications [8][9][10]. The vagal nerve blockade (vBloc) device is inserted by standard laparoscopic surgical techniques without permanent anatomical alterations of the gastrointestinal tract. ...
... The vBloc device has been shown to clinically result in weight loss with good safety outcomes. The ReCharge randomised-controlled trial (RCT) demonstrated an estimated mean percentage excess weight loss (%EWL) of 26% (10% total weight loss, TWL) for vBloc and 17% (6%TWL) for sham at 12 months (p < 0.001) [8]. The VBLOC DM2 study also found that intermittent vagal blocking led to significant weight loss with improvements in obesity and glycaemic control after two years of treatment with a well-tolerated safety profile [14]. ...
... The study characteristics of each individual study included in this systematic review are shown in Table 1. There were nine relevant RCTs [8,10,25,27,28,30,31,33] out of 15 studies. Six [9,14,26,28,32,34] were cohort studies, two [9,32] of which were prospective. ...
Article
Full-text available
Introduction: The vagus nerve has an important role in satiety, metabolism and autonomic control in upper gastrointestinal function. However, the role and effects of vagal nerve therapy on weight loss remain controversial. This systematic review and meta-analysis assessed the effects of vagal nerve therapy on weight loss, body mass index (BMI) and obesity-related conditions. Methods: MEDLINE, EMBASE and CINAHL databases were searched for studies up to April 2022 that reported on % excess weight loss (%EWL) or BMI at 12 months or remission of obesity-related conditions following vagal nerve therapy from January 2000 to April 2022. Weighted mean difference (WMD) was calculated, and meta-analysis was performed using random-effect models and between-study heterogeneity was assessed. Results: Fifteen studies, of which nine were randomised-controlled trials, of 1447 patients were included. Vagal nerve therapy led to some improvement in %EWL (WMD 17.19%; 95% CI 10.94 to 23.44; p < 0.001) and BMI (WMD -2.24 kg/m2; 95% CI -4.07 to -0.42; p = 0.016). There was a general improvement found in HbA1c following vagal nerve therapy when compared to no treatment given. No major complications were reported. Conclusions: Vagal nerve therapy can safely result in a mild-to-moderate improvement in weight loss. However, further clinical trials are required to confirm these results and investigate the possibility of the long-term benefit of vagal nerve therapy as a dual therapy combined with standard surgical bariatric interventions.
... The VBLOC Therapy Maestro system is an FDAapproved device intended to restate the hungersuppressing effect of subdiaphragmatic vagotomy 3,4 . ...
... Although trial studies report treatment group of study participants experienced significant weight loss compared to sham group 3 and sustained the excessive weight loss compared to control group for 24 months 4 , the efficacy of the treatment is known to be modest 4,5 . Moreover, several animal studies reported that excitationnot the blockingof VN activity by the VBLOC system resulted in reduced food intake and weight loss [6][7][8] . ...
Article
Full-text available
The vagus nerve (VN) plays an important role in regulating physiological conditions in the gastrointestinal (GI) tract by communicating via the parasympathetic pathway to the enteric nervous system (ENS). However, the lack of knowledge in the neurophysiology of the VN and GI tract limits the development of advanced treatments for autonomic dysfunctions related to the VN. To better understand the complicated underlying mechanisms of the VN-GI tract neurophysiology, it is necessary to use an advanced device enabled by microfabrication technologies. Among several candidates including intraneural probe array and extraneural cuff electrodes, microchannel electrode array devices can be used to interface with smaller numbers of nerve fibers by securing them in the separate channel structures. Previous microchannel electrode array devices to interface teased nerve structures are relatively bulky with thickness around 200 µm. The thick design can potentially harm the delicate tissue structures, including the nerve itself. In this paper, we present a flexible thin film based microchannel electrode array device (thickness: 11.5 µm) that can interface with one of the subdiaphragmatic nerve branches of the VN in a rat. We demonstrated recording evoked compound action potentials (ECAP) from a transected nerve ending that has multiple nerve fibers. Moreover, our analysis confirmed that the signals are from C-fibers that are critical in regulating autonomic neurophysiology in the GI tract.
... Afferent vagal nerve fibers are sensitive to the mechanical stretch of the stomach after ingestion of food. Furthermore, gut hormones, including ghrelin, GLP-1, and PYY, have been suggested to interact with vagal nerve receptors [124] . Studies investigating the impact of intermittent vagal nerve blockade on weight loss have shown some promise, but more work is necessary before drawing conclusions [125,126] . ...
... Studies investigating the impact of intermittent vagal nerve blockade on weight loss have shown some promise, but more work is necessary before drawing conclusions [125,126] . It has been shown that after RYGB and VSG, gastric branches of the vagus nerve are cut, causing damage to efferent and afferent fibers [124] . The significance of this and whether or not it is related to the metabolic effects of surgery require further investigation. ...
Article
Full-text available
After metabolic surgery, patients with type 2 diabetes (T2DM) typically experience a rapid improvement in glycemic control before any significant weight loss occurs. Furthermore, a significant proportion of patients are able to achieve long-term T2DM remission and improvement in β-cell function. While historically believed to be related to weight loss and caloric restriction, multiple weight loss independent mechanisms have been identified to contribute to the long-term glycemic effects induced by metabolic surgery. There are changes in bile acid metabolism, the gut microbiome, incretins, and other gut hormones after surgery that are implicated. It is also becoming increasingly evident that adipose tissue, specifically visceral adipose tissue, is implicated in the pathogenesis of insulin resistance (IR) and T2DM through inflammatory changes involving the host immune system. Therefore, metabolic surgery may exert its effects by reducing the inflammatory response through reduction of adipose. While these mechanisms may seem discrete, there is a significant cross-talk between all these factors that contributes to the regulation of glucose homeostasis. Together, this leads to reduced gluconeogenesis, improved glucose tissue uptake, reduced IR, and improved β-cell function after metabolic surgery.
... In human studies, the vital role of the VN in modulating food intake, energy metabolism, and glycemic control has been demonstrated more recently [23][24][25][26][27]. Across disciplines the pathophysiology of several disorders is attributed to the VN on a behavioral and psychological level; VN stimulation, remarkably non-invasive VN stimulation, has been studied, allowing a better understanding of the mechanisms by which VN stimulation exerts psychological and physiological effects. ...
Chapter
Full-text available
The medical community recognized last decennia the multidimensional nature of pain and proposed multimodal biopsychosocial management. The most compelling reason to embrace integrative pain strategies is to mitigate patient risk. For patients with chronic pain and pain refractory to conservative medicine, it is essential to assess all factors involved with the chronicity. With significant themes, nutrition and microbiome, neuroplasticity, homeostasis, and the side effects of medication, acupuncture has progressively gained a place in this multimodal evaluation. Therapeutic multimodality approaches the perspective of physiological rehabilitation and chronobiological improvement of the quality of life. Illustrated by various clinical situations, the objective of management is to seek a synergy in the mechanisms of action of treatments to improve quality of life and reduce the need for xenobiotics and, consequently, the side effects. The mechanism of action of integrative medicine, and acupuncture improved with a better understanding of genetics, and epigenetics. As opposed to sham and placebo, acupuncture activates other brain regions. In controlled trials, the strict inclusion and exclusion criteria result in the treatment of a “selected” patient population, which is not always comparable to the patients seen in daily practice. The integrative approach is better illustrated by case reports.
... VNS is FDA-approved to treat epilepsy, depression, obesity, and for stroke rehabilitation (U.S. Food and Drug Administration, PMAs P970003, P130019, P210007). It is being investigated as a therapy for heart failure (Zile et al., 2020), hypertension (Ntiloudi et al., 2019), inflammatory conditions (Pavlov and Tracey, 2012;Kessler et al., 2012), traumatic brain injury (TBI) (Bansal et al., 2012), lung injury (Santos et al., 2011;Reys et al., 2013) Alzheimer's disease (Merrill, Bikson, and Jefferys, 2005), anxiety (George et al., 2008), chronic pain (Chakravarthy et al., 2015), tinnitus (Tyler et al., 2017), rheumatoid arthritis (Koopman et al., 2016), diabetes (Meyers et al., 2016) and obesity (Val-Laillet et al., 2010;Ikramuddin et al., 2014). to achieve a more precise stimulation, the search space of possible parameter sets expands significantly. ...
Preprint
Full-text available
Background: Vagus nerve stimulation (VNS) is an established therapy for treating a variety of chronic diseases, such as epilepsy, depression, obesity, and for stroke rehabilitation. However, lack of precision and side-effects have hindered its efficacy and extension to new conditions. Objective: To achieve a better understanding of the relationship between VNS parameters and neural and physiological responses to enable the design of personalized dosing procedures to improve precision and efficacy of VNS therapies. Methods: We used biomarkers from recorded evoked neural activity and short-term physiological responses (throat muscle, cardiac and respiratory activity) to understand the response to a wide range of VNS parameters in anaesthetised pigs. Using signal processing, Gaussian processes (GP) and parametric regression models we analyse the relationship between VNS parameters and neural and physiological responses. Results: Firstly, we observe inter-subject variability for both neural and physiological responses. Secondly, we illustrate how considering multiple stimulation parameters in VNS dosing can improve the efficacy and precision of VNS therapies. Thirdly, we describe the relationship between different VNS parameters and the evoked neural activity and show how spatially selective electrodes can be used to improve fibre recruitment. Fourthly, we provide a detailed exploration of the relationship between the activations of neural fibre types and different physiological effects, and show that recordings of evoked neural activity are powerful biomarkers for predicting the short-term physiological effects of VNS. Finally, based on these results, we discuss how recordings of evoked neural activity can help design VNS dosing procedures that optimize short-term physiological effects safely and efficiently. Conclusion: Understanding of evoked neural activity during VNS provide powerful biomarkers that could improve the precision, safety and efficacy of VNS therapies.
Article
italic xmlns:mml="http://www.w3.org/1998/Math/MathML" xmlns:xlink="http://www.w3.org/1999/xlink">Objective: The purpose of this paper is to investigate the persistence of nerve blockade beyond the duration of applying high frequency alternating current (HFAC) to thinly myelinated and non-myelinated fibers, also termed a “carry-over effect”. Methods: In this study, we used electrically-evoked compound action potentials from isolated rat vagus nerves to assess the influence of 5 kHz HFAC amplitude and duration on the degree of the carry-over effect. Current amplitudes from 1-10 mA and 5 kHz durations from 10-120 seconds were tested. Results: By testing 20 different combinations of 5 kHz amplitude and duration, we found a significant interaction between 5 kHz amplitude and duration on influencing the carry-over effect. Conclusion: The degree of carry-over effect was dependent on 5 kHz amplitude, as well as duration. Significance: Utilizing the carry-over effect may be useful in designing energy efficient nerve blocking algorithms for the treatment of diseases influenced by nerve activity.
Article
Full-text available
The correlation between body mass index (BMI) and the development of cough, shortness of breath, and dyspnea is unclear. Therefore, this study aimed to investigate the association between these parameters. Data from individuals who participated in the National Health and Nutrition Examination Survey between 2003 and 2012 were analyzed. Weighted logistic regression analysis and smoothed curve fitting were used to examine the correlation between BMI and respiratory symptoms. In addition, the relationship between BMI, chronic obstructive pulmonary disease (COPD), and bronchial asthma was examined. Stratified analysis was used to discover inflection points and specific groups. Weighted logistic regression and smoothed curve fitting revealed a U-shaped relationship between BMI and respiratory symptoms. The U-shaped relationship in BMI was also observed in patients with bronchial asthma and COPD. Stratified analysis showed that the correlation between BMI and wheezing and dyspnea was influenced by race. In addition, non-Hispanic black individuals had a higher risk of developing cough than individuals of the other three races [OR 1.040 (1.021, 1.060), p < 0.0001], and they also exhibited an inverted U-shaped relationship between BMI and bronchial asthma. However, the association of BMI with cough, wheezing, dyspnea, COPD, and asthma was not affected by sex. High or low BMI was associated with cough, shortness of breath, and dyspnea, and has been linked to bronchial asthma and COPD. These findings provide new insights into the management of respiratory symptoms and respiratory diseases.
Article
Full-text available
Background: Weight loss is recommended for overweight or obese patients with type 2 diabetes on the basis of short-term studies, but long-term effects on cardiovascular disease remain unknown. We examined whether an intensive lifestyle intervention for weight loss would decrease cardiovascular morbidity and mortality among such patients. Methods: In 16 study centers in the United States, we randomly assigned 5145 overweight or obese patients with type 2 diabetes to participate in an intensive lifestyle intervention that promoted weight loss through decreased caloric intake and increased physical activity (intervention group) or to receive diabetes support and education (control group). The primary outcome was a composite of death from cardiovascular causes, nonfatal myocardial infarction, nonfatal stroke, or hospitalization for angina during a maximum follow-up of 13.5 years. Results: The trial was stopped early on the basis of a futility analysis when the median follow-up was 9.6 years. Weight loss was greater in the intervention group than in the control group throughout the study (8.6% vs. 0.7% at 1 year; 6.0% vs. 3.5% at study end). The intensive lifestyle intervention also produced greater reductions in glycated hemoglobin and greater initial improvements in fitness and all cardiovascular risk factors, except for low-density-lipoprotein cholesterol levels. The primary outcome occurred in 403 patients in the intervention group and in 418 in the control group (1.83 and 1.92 events per 100 person-years, respectively; hazard ratio in the intervention group, 0.95; 95% confidence interval, 0.83 to 1.09; P=0.51). Conclusions: An intensive lifestyle intervention focusing on weight loss did not reduce the rate of cardiovascular events in overweight or obese adults with type 2 diabetes. (Funded by the National Institutes of Health and others; Look AHEAD ClinicalTrials.gov number, NCT00017953.).
Data
Full-text available
Background: Arthroscopic partial meniscectomy is one of the most common orthopedic procedures, yet rigorous evidence of its efficacy is lacking. Methods: We conducted a multicenter, randomized, double-blind, sham-controlled trial in 146 patients 35 to 65 years of age who had knee symptoms consistent with a degenerative medial meniscus tear and no knee osteoarthritis. Patients were randomly assigned to arthroscopic partial meniscectomy or sham surgery. The primary outcomes were changes in the Lysholm and Western Ontario Meniscal Evaluation Tool (WOMET) scores (each ranging from 0 to 100, with lower scores indicating more severe symptoms) and in knee pain after exercise (rated on a scale from 0 to 10, with 0 denoting no pain) at 12 months after the procedure. Results: In the intention-to-treat analysis, there were no significant between-group differences in the change from baseline to 12 months in any primary outcome. The mean changes (improvements) in the primary outcome measures were as follows: Lysholm score, 21.7 points in the partial-meniscectomy group as compared with 23.3 points in the sham-surgery group (between-group difference, -1.6 points; 95% confidence interval [CI], -7.2 to 4.0); WOMET score, 24.6 and 27.1 points, respectively (between-group difference, -2.5 points; 95% CI, -9.2 to 4.1); and score for knee pain after exercise, 3.1 and 3.3 points, respectively (between-group difference, -0.1; 95% CI, -0.9 to 0.7). There were no significant differences between groups in the number of patients who required subsequent knee surgery (two in the partial-meniscectomy group and five in the sham-surgery group) or serious adverse events (one and zero, respectively). Conclusions: In this trial involving patients without knee osteoarthritis but with symptoms of a degenerative medial meniscus tear, the outcomes after arthroscopic partial meniscectomy were no better than those after a sham surgical procedure. (Funded by the Sigrid Juselius Foundation and others; ClinicalTrials.gov number, NCT00549172.).
Article
Full-text available
Background: An active device that downregulates abdominal vagal signalling has resulted in significant weight loss in feasibility studies. Objective: To prospectively evaluate the effect of intermittent vagal blocking (VBLOC) on weight loss, glycemic control, and blood pressure (BP) in obese subjects with DM2. Methods: Twenty-eight subjects were implanted with a VBLOC device (Maestro Rechargeable System) at 5 centers in an open-label study. Effects on weight loss, HbA1c, fasting blood glucose, and BP were evaluated at 1 week to 12 months. Results: 26 subjects (17 females/9 males, 51 ± 2 years, BMI 37 ± 1 kg/m(2), mean ± SEM) completed 12 months followup. One serious adverse event (pain at implant site) was easily resolved. At 1 week and 12 months, mean excess weight loss percentages (% EWL) were 9 ± 1% and 25 ± 4% (P < 0.0001), and HbA1c declined by 0.3 ± 0.1% and 1.0 ± 0.2% (P = 0.02, baseline 7.8 ± 0.2%). In DM2 subjects with elevated BP (n = 15), mean arterial pressure reduced by 7 ± 3 mmHg and 8 ± 3 mmHg (P = 0.04, baseline 100 ± 2 mmHg) at 1 week and 12 months. All subjects MAP decreased by 3 ± 2 mmHg (baseline 95 ± 2 mmHg) at 12 months. Conclusions: VBLOC was safe in obese DM2 subjects and associated with meaningful weight loss, early and sustained improvements in HbA1c, and reductions in BP in hypertensive DM2 subjects. This trial is registered with ClinicalTrials.gov NCT00555958.
Article
Full-text available
Objective To examine the effects of naltrexone/bupropion (NB) combination therapy on weight and weight-related risk factors in overweight and obese participants. Design and Methods CONTRAVE Obesity Research-II (COR-II) was a double-blind, placebo-controlled study of 1,496 obese (BMI 30-45 kg/m2) or overweight (27-45 kg/m2 with dyslipidemia and/or hypertension) participants randomized 2:1 to combined naltrexone sustained-release (SR) (32 mg/day) plus bupropion SR (360 mg/day) (NB32) or placebo for up to 56 weeks. The co-primary endpoints were percent weight change and proportion achieving ≥5% weight loss at week 28. Results Significantly (P < 0.001) greater weight loss was observed with NB32 versus placebo at week 28 (−6.5% vs. −1.9%) and week 56 (−6.4% vs. −1.2%). More NB32-treated participants (P < 0.001) experienced ≥5% weight loss versus placebo at week 28 (55.6% vs. 17.5%) and week 56 (50.5% vs. 17.1%). NB32 produced greater improvements in various cardiometabolic risk markers, participant-reported weight-related quality of life, and control of eating. The most common adverse event with NB was nausea, which was generally mild to moderate and transient. NB was not associated with increased events of depression or suicidality versus placebo. Conclusion NB represents a novel pharmacological approach to the treatment of obesity, and may become a valuable new therapeutic option.
Article
Full-text available
Calls for primary care providers (PCPs) to offer obese patients behavioral weight-loss counseling have not been accompanied by adequate guidance on how such care could be delivered. This randomized trial compared weight loss during a 2-year period in response to three lifestyle interventions, all delivered by PCPs in collaboration with auxiliary health professionals (lifestyle coaches) in their practices. We randomly assigned 390 obese adults in six primary care practices to one of three types of intervention: usual care, consisting of quarterly PCP visits that included education about weight management; brief lifestyle counseling, consisting of quarterly PCP visits combined with brief monthly sessions with lifestyle coaches who instructed participants about behavioral weight control; or enhanced brief lifestyle counseling, which provided the same care as described for the previous intervention but included meal replacements or weight-loss medication (orlistat or sibutramine), chosen by the participants in consultation with the PCPs, to potentially increase weight loss. Of the 390 participants, 86% completed the 2-year trial, at which time, the mean (±SE) weight loss with usual care, brief lifestyle counseling, and enhanced brief lifestyle counseling was 1.7±0.7, 2.9±0.7, and 4.6±0.7 kg, respectively. Initial weight decreased at least 5% in 21.5%, 26.0%, and 34.9% of the participants in the three groups, respectively. Enhanced lifestyle counseling was superior to usual care on both these measures of success (P=0.003 and P=0.02, respectively), with no other significant differences among the groups. The benefits of enhanced lifestyle counseling remained even after participants given sibutramine were excluded from the analyses. There were no significant differences between the intervention groups in the occurrence of serious adverse events. Enhanced weight-loss counseling helps about one third of obese patients achieve long-term, clinically meaningful weight loss. (Funded by the National Heart, Lung, and Blood Institute; POWER-UP ClinicalTrials.gov number, NCT00826774.).
Article
Laparoscopic adjustable gastric banding has several distinctive features, including band adjustability, easy reversibility, and lack of malabsorption, which contribute to its widespread use. The LAP-BAND AP System (LBAP; Allergan, Inc.), a redesigned and improved version of the original device, was approved by the US Food and Drug Administration in 2006. Because of limited information on LBAP, this study prospectively assesses the efficacy and safety of LBAP in real-world settings at clinical centers located in North America, Europe, and Australia. This interim report of the ongoing 5-year prospective, observational, international, multicenter registry, Helping Evaluate Reduction in Obesity (HERO) Study (NCT00953173), describes clinical efficacy and safety of LBAP in real-world settings at 1 year. One thousand one hundred and six patients were implanted with LBAP and 1-year data were available from 834 patients for efficacy analysis. At 1 year, the mean (SD) percentage of excess weight loss was 39.8% (22.3%), of weight loss was 16.9% (9.0%), and the mean (SD) body mass index decreased to 37.7 (7.0) kg/m(2) from 45.1 (6.9) kg/m(2) at baseline. Patients with type 2 diabetes mellitus or hypertension showed significant improvements at 1 year post LBAP (both p < 0.005). The most common device-related complications were port displacement (n = 20 [1.8%]), pouch dilation (n = 12 [1.1%]), band slippage (n = 7 [0.6%]), and band erosion (n = 5 [0.5%]). Eighteen (1.6%) patients had the device explanted. At 1 year post LBAP, progressive weight loss was associated with improvement and/or resolution of comorbid conditions and was safe and well tolerated. Patient follow-up continues.
Article
In 1959 and 1960 the results of two randomized controlled trials demonstrated that bilateral internal mammary artery ligation was no better than a sham intervention in the treatment of angina. These landmark trials were the first to use blinded placebo controls to evaluate invasive procedures. Revisiting the story of internal mammary artery ligation offers valuable insights into the ethics of sham-controlled trials and the implications of the placebo effect for thinking about risk-benefit assessment of sham procedures.
Article
Background: Intermittent, reversible intraabdominal vagal blockade (VBLOC® Therapy) demonstrated clinically important weight loss in feasibility trials. EMPOWER, a randomized, double-blind, prospective, controlled trial was conducted in USA and Australia. Methods: Five hundred three subjects were enrolled at 15 centers. After informed consent, 294 subjects were implanted with the vagal blocking system and randomized to the treated (n = 192) or control (n = 102) group. Main outcome measures were percent excess weight loss (percent EWL) at 12 months and serious adverse events. Subjects controlled duration of therapy using an external power source; therapy involved a programmed algorithm of electrical energy delivered to the subdiaphragmatic vagal nerves to inhibit afferent/efferent vagal transmission. Devices in both groups performed regular, low-energy safety checks. Data are mean ± SEM. Results: Study subjects consisted of 90 % females, body mass index of 41 ± 1 kg/m(2), and age of 46 ± 1 years. Device-related complications occurred in 3 % of subjects. There was no mortality. 12-month percent EWL was 17 ± 2 % for the treated and 16 ± 2 % for the control group. Weight loss was related linearly to hours of device use; treated and controls with ≥ 12 h/day use achieved 30 ± 4 and 22 ± 8 % EWL, respectively. Conclusions: VBLOC® therapy to treat morbid obesity was safe, but weight loss was not greater in treated compared to controls; clinically important weight loss, however, was related to hours of device use. Post-study analysis suggested that the system electrical safety checks (low charge delivered via the system for electrical impedance, safety, and diagnostic checks) may have contributed to weight loss in the control group.
Article
Background: To compare implantable gastric stimulation therapy with a standard diet and behavioral therapy regimen in a group of carefully selected class 2 and 3 obese subjects by evaluating the difference in the percentage of excess weight loss (EWL) between the control and treatment groups. The primary endpoint was the percentage of EWL from baseline to 12 months after randomization. Implantable gastric stimulation has been proposed as a first-line treatment for severely obese patients; however, previous investigations have reported inconclusive results. Methods: A total of 190 subjects were enrolled in this prospective, randomized, placebo-controlled, double-blind, multicenter study. All patients underwent implantation with the implantable gastric stimulator and were randomized to 1 of 2 treatment groups: the control group (stimulation off) or treatment group (stimulation on). The patients were evaluated on a monthly basis. All individuals who enrolled in this study agreed to consume a diet with a 500-kcal/d deficit and to participate in monthly support group meetings. Results: The procedure resulted in no deaths and a low complication rate. The primary endpoint of a difference in weight loss between the treatment and control groups was not met. The control group lost 11.7% +/- 16.9% of excess weight and the treatment group lost 11.8% +/- 17.6% (P = .717) according to an intent-to-treat analysis. Conclusion: Implantable gastric stimulation as a surgical option for the treatment of morbid obesity is a less complex procedure than current bariatric operations. However, the results of the present study do not support its application. Additional research is indicated to understand the physiology and potential benefits of this therapy.