ArticlePDF Available

Multi-dimensional cognitive behavior therapy for obesity applied by clinical psychologist using digital platform: an open-label, randomised controlled trial (Preprint)

Authors:

Abstract and Figures

Background: Developing effective, widely useful, weight management programs is a priority in health care because obesity is a major health problem. Objective: This study developed and investigated a new, comprehensive, multifactorial, daily, intensive, psychologist coaching program based on cognitive behavioral therapy (CBT) modules. The program was delivered via the digital health care mobile services Noom Coach and InBody. Methods: This was an open-label, active-comparator, randomized controlled trial. A total of 70 female participants with BMI scores above 24 kg/m2 and no clinical problems besides obesity were randomized into experimental and control groups. The experimental (ie, digital CBT) group (n=45) was connected with a therapist intervention using a digital health care service that provided daily feedback and assignments for 8 weeks. The control group (n=25) also used the digital health care service, but practiced self-care without therapist intervention. The main outcomes of this study were measured objectively at baseline, 8 weeks, and 24 weeks and included weight (kg) as well as other body compositions. Differences between groups were evaluated using independent t tests and a per-protocol framework. Results: Mean weight loss at 8 weeks in the digital CBT group was significantly higher than in the control group (-3.1%, SD 4.5, vs -0.7%, SD 3.4, P=.04). Additionally, the proportion of subjects who attained conventional 5% weight loss from baseline in the digital CBT group was significantly higher than in the control group at 8 weeks (32% [12/38] vs 4% [1/21], P=.02) but not at 24 weeks. Mean fat mass reduction in the digital CBT group at 8 weeks was also significantly greater than in the control group (-6.3%, SD 8.8, vs -0.8%, SD 8.1, P=.02). Mean leptin and insulin resistance in the digital CBT group at 8 weeks was significantly reduced compared to the control group (-15.8%, SD 29.9, vs 7.2%, SD 35.9, P=.01; and -7.1%, SD 35.1, vs 14.4%, SD 41.2, P=.04). Emotional eating behavior (ie, mean score) measured by questionnaire (ie, the Dutch Eating Behavior Questionnaire) at 8 weeks was significantly improved compared to the control group (-2.8%, SD 34.4, vs 21.6%, SD 56.9, P=.048). Mean snack calorie intake in the digital CBT group during the intervention period was significantly lower than in the control group (135.9 kcal, SD 86.4, vs 208.2 kcal, SD 166.3, P=.02). Lastly, baseline depression, anxiety, and self-esteem levels significantly predicted long-term clinical outcomes (24 weeks), while baseline motivation significantly predicted both short-term (8 weeks) and long-term clinical outcomes. Conclusions: These findings confirm that technology-based interventions should be multidimensional and are most effective with human feedback and support. This study is innovative in successfully developing and verifying the effects of a new CBT approach with a multidisciplinary team based on digital technologies rather than standalone technology-based interventions. Trial registration: ClinicalTrials.gov NCT03465306; https://clinicaltrials.gov/ct2/show/NCT03465306.
Content may be subject to copyright.
Original Paper
Multidimensional Cognitive Behavioral Therapy for Obesity Applied
by Psychologists Using a Digital Platform: Open-Label
Randomized Controlled Trial
Meelim Kim1,2, MA; Youngin Kim3,4, MD; Yoonjeong Go1; Seokoh Lee1, BS; Myeongjin Na2, BA; Younghee Lee1,
BS; Sungwon Choi2*, PhD; Hyung Jin Choi1*, MD
1Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
2Department of Psychology, Duksung Women’s University, Ssangmun-Dong, Dobong-Gu, Republic of Korea
3Noom Inc, New York City, NY, United States
4Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
*these authors contributed equally
Corresponding Author:
Hyung Jin Choi, MD
Department of Biomedical Sciences
Seoul National University College of Medicine
28 Yungun-Dong, Chongno-Gu
Seoul
Republic of Korea
Phone: 82 2 740 8204
Email: hjchoi@snu.ac.kr
Abstract
Background: Developing effective, widely useful, weight management programs is a priority in health care because obesity is
a major health problem.
Objective: This study developed and investigated a new, comprehensive, multifactorial, daily, intensive, psychologist coaching
program based on cognitive behavioral therapy (CBT) modules. The program was delivered via the digital health care mobile
services Noom Coach and InBody.
Methods: This was an open-label, active-comparator, randomized controlled trial. A total of 70 female participants with BMI
scores above 24 kg/m2and no clinical problems besides obesity were randomized into experimental and control groups. The
experimental (ie, digital CBT) group (n=45) was connected with a therapist intervention using a digital health care service that
provided daily feedback and assignments for 8 weeks. The control group (n=25) also used the digital health care service, but
practiced self-care without therapist intervention. The main outcomes of this study were measured objectively at baseline, 8
weeks, and 24 weeks and included weight (kg) as well as other body compositions. Differences between groups were evaluated
using independent ttests and a per-protocol framework.
Results: Mean weight loss at 8 weeks in the digital CBT group was significantly higher than in the control group (–3.1%, SD
4.5, vs –0.7%, SD 3.4, P=.04). Additionally, the proportion of subjects who attained conventional 5% weight loss from baseline
in the digital CBT group was significantly higher than in the control group at 8 weeks (32% [12/38] vs 4% [1/21], P=.02) but not
at 24 weeks. Mean fat mass reduction in the digital CBT group at 8 weeks was also significantly greater than in the control group
(–6.3%, SD 8.8, vs –0.8%, SD 8.1, P=.02). Mean leptin and insulin resistance in the digital CBT group at 8 weeks was significantly
reduced compared to the control group (–15.8%, SD 29.9, vs 7.2%, SD 35.9, P=.01; and –7.1%, SD 35.1, vs 14.4%, SD 41.2,
P=.04). Emotional eating behavior (ie, mean score) measured by questionnaire (ie, the Dutch Eating Behavior Questionnaire) at
8 weeks was significantly improved compared to the control group (–2.8%, SD 34.4, vs 21.6%, SD 56.9, P=.048). Mean snack
calorie intake in the digital CBT group during the intervention period was significantly lower than in the control group (135.9
kcal, SD 86.4, vs 208.2 kcal, SD 166.3, P=.02). Lastly, baseline depression, anxiety, and self-esteem levels significantly predicted
long-term clinical outcomes (24 weeks), while baseline motivation significantly predicted both short-term (8 weeks) and long-term
clinical outcomes.
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 1http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
Conclusions: These findings confirm that technology-based interventions should be multidimensional and are most effective
with human feedback and support. This study is innovative in successfully developing and verifying the effects of a new CBT
approach with a multidisciplinary team based on digital technologies rather than standalone technology-based interventions.
Trial Registration: ClinicalTrials.gov NCT03465306; https://clinicaltrials.gov/ct2/show/NCT03465306
(JMIR Mhealth Uhealth 2020;8(4):e14817) doi: 10.2196/14817
KEYWORDS
obesity; digital health care; cognitive behavioral therapy; mobile phone
Introduction
One of the major concerns of the health care industry is to find
effective and widely practical solutions for weight management,
given that obesity is one of the dominant public health problems
of the 21st century. It is well known that weight reduction is
highly correlated with reductions in the incidence of type 2
diabetes, as well as other medical weight-related comorbidities
and psychosocial issues, and that it improves the quality of life
[1].
Accordingly, various types of treatments for obesity have been
developed. Several drugs have been proposed as
pharmacotherapy for obesity since the 1990s, but most have
demonstrated a lack of efficacy and unfavorable risks [2].
Bariatric surgery is another obesity treatment that has been used
for over 50 years. Because the prevalence of obesity is rapidly
rising, the number of patients who believe that bariatric surgery
is an effective treatment to cure their obesity is also increasing
[3]. Additionally, patients may believe that surgical intervention
to overcome obesity will ultimately lead to behavioral changes
sustaining weight loss [3], which may increase the risk of weight
regains after the surgery. To date, the most effective standard
obesity treatment is weight-loss lifestyle modification based on
a combination of behavioral and cognitive approaches and
nutrition and physical education.
Clinical psychological treatment approaches are pivotal and
involve engaging patients in lifestyle modification and
motivating them to successfully lose weight with the help of a
multidisciplinary team [4]. Cognitive behavioral therapy (CBT)
for obesity is aimed at not only losing weight but also preventing
weight regain, thereby avoiding the dissatisfactory long-term
results of earlier behavioral treatments. It firmly distinguishes
between weight loss and weight maintenance, allowing patients
to practice effective weight-maintenance strategies (eg, avoiding
unrealistic weight goals and addressing obstacles to weight
maintenance) [5]. One study applied a 12-week CBT program
for obese people, resulting in a 6% reduction in body fat relative
to the control group [6]. Moreover, a 20-week CBT intervention
involving a 10-week main program followed by a 10-week
less-intensive care program significantly improved body
composition and improved soft drink consumption habits
compared to the control group [7].
Although cognitive behavioral programs involving weekly clinic
visits are known to be the most effective treatments for obesity,
they place high demands due to time, cost, distance, status of
endorsement, and difficulties securing child care [8]. A previous
study found that people would prefer cost-effective and
time-saving methods to lose weight [9]. Researchers have thus
explored alternative methods for carrying out weight-loss
programs, such as television, computers, and smartphone apps,
to meet individual needs and to make obesity treatment more
accessible. Among these, self-monitoring via smartphone apps
has shown the greatest potential to make diet tracking easier
and engaging because of its convenience and accessibility [10].
Despite the use of smartphone apps for self-monitoring, a law
of attrition in digital health interventions still holds, whereby
users stop using technology-based components over time.
Because the effectiveness of treatments via digital tools is
closely associated with the user’s extent of engagement [11], a
high attrition rate is a critical issue in the assessment of the
efficacy of digital intervention programs. Therefore, based on
behavioral modification principles, periodic prompts that
encourage healthy behaviors are one method to remind and
motivate people to change their health behaviors. A systematic
review of the use of technology tools to send periodic
notifications about users’ behavior changes found them to be
more effective than nontechnological notifications or no
notifications [12]. However, this review only focused on the
effectiveness of digital interventions for behavior change as a
whole and did not investigate how to enhance engagement with
the intervention.
The goal of this study was to test a novel approach to losing
weight and maintaining the new weight after participation in
an intensive and comprehensive human coaching program based
on CBT modules via digital tools, such as the Noom Coach app
and InBody Dial. The Noom Coach app is one of the most
popular smartphone apps currently available; it has received
higher quality assessment scores than other smartphone apps
[13]. It allows participants to log their food intake, exercise
activities, and weight, and to engage in in-app group activities,
read in-app articles, and interact with a human coach via in-app
messages. In-app group activity lets participants communicate
with other participants and share their experience of healthy
lifestyle trials. In-app articles deliver practical information about
healthy lifestyles written by physicians, nutritionists, and clinical
psychologists. In-app messages enable participants to receive
individualized feedback from human coaches based on their
own records presented on the Web-based dashboard. A
Web-based dashboard is provided to the coaches to monitor
participants’data. InBody Dial is a body composition analyzer
for the home linked to a mobile app, allowing users to
conveniently measure their body composition. Furthermore, we
addressed the self-sustainability of the promoted lifestyle change
after the intervention. We hypothesized that individuals
randomized to the digital CBT group would lose weight and
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 2http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
better maintain their weight loss than individuals in the control
group.
Methods
Participants
A total of 70 female subjects were recruited between September
and October 2017 through both online and offline boards of a
university campus in Seoul, South Korea, and a social network
service. Eligibility criteria included the following: 18-39 years
of age, body mass index of 25-40 kg/m2, smartphone usage,
and scores in the highest 40% (ie, scores above 68 out of 112
total) on the Situational Motivation Scale (SIMS). Participants
were ineligible if they had a history of major medical problems,
such as diabetes, angina, or stroke; a major psychiatric disorder
involving hospitalization or medication in the past; and a current
or planned pregnancy within the next 6 months. The flow of
participants from recruitment to final assessment at 24 weeks
is shown in Figure 1.
Figure 1. Digital cognitive behavioral therapy (CBT) CONSORT (Consolidated Standards of Reporting Trials) flow diagram. SIMS: Situational
Motivation Scale.
The Institutional Review Board of Seoul National University
Hospital approved the study (approval number
H-1707-122-872). All study participants provided written
informed consent. This study was conducted to examine the
clinical efficacy of the obesity digital CBT model and find
factors predicting its efficacy. The study was registered with
ClinicalTrials.gov (NCT03465306).
Study Design
This was an open-label, active-comparator, randomized
controlled trial (RCT). Following initial screening, all
participants were asked to attend an orientation session where
the study was described in more detail. Written informed consent
and baseline measurements were obtained in person. Blood
samples were taken in the morning after overnight fasting to
avoid daily variations in activities. The basics of the tutorial
and log-in procedures for both the Noom app and the InBody
H20B (InBody Co) body composition analyzer were
demonstrated to all participants during the orientation session
of the study. The Noom app was mainly used to keep a food
diary and deliver messages between the therapist and
participants, while the InBody H20B analyzer was used to
monitor and collect the body composition data of the
participants. The randomization was designed to randomly
assign 75 participants in total to a control (app only) group or
a digital CBT (app + human CBT) group at a ratio of 1:2 in
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 3http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
order to deliver a more powerful trial within resource constraints
and to maximize the statistical power of predictor analysis (ie,
within-group analysis) [14]. Randomization was performed by
the project manager by drawing lots. The digital CBT group
was given daily feedback and assignments from a psychologist,
based on the CBT modules, for 8 weeks and could access the
digital tools from the intervention period to the 24-week
follow-up. The control group was instructed to use only a food
diary without therapist intervention until the 24-week follow-up
but was given the same digital tools and instruction as the digital
CBT group. Thus, the control group underwent the same
standard-of-care trial as the digital CBT group, except that it
was asked to practice self-care. All participants were asked to
visit at baseline, 8 weeks, and 24 weeks for objective
measurements and completion of questionnaires, and they were
each paid US $4 for attending each of the appointments. This
study was conducted from September 2017 to April 2018.
Assessment
The primary outcome was change in body weight. Other
measures, such as change in BMI and body fat mass, were
secondary outcomes. Anthropometric measurements were
assessed by the InBody H20B analyzer at baseline, 8 weeks,
and 24 weeks in light street clothing and without socks and
shoes. For secondary outcomes, blood samples were collected
at baseline and 8 weeks after a 10-hour fast. We examined serum
insulin, leptin, glucose concentrations, aspartate
aminotransferase, alanine aminotransferase, gamma-glutamyl
transferase, total cholesterol, and triglyceride levels to assess
the changes in these indices in relation to the change in body
weight. The engagement criteria of the program were completing
actions, such as responding to the daily assessment (responses
per day), logging meals (meals per week), consuming green
foods as defined by Noom [15] (logged per week), performing
exercise (times per week), registering exercise time (minutes
per week), recording steps taken (steps per week), logging
weigh-ins (times per week), reading articles (articles per week),
completing group posts (posts per week), posting group
comments (comments per week), sending messages to the coach
(messages per week), and liking group posts (likes per week).
These criteria were used to assess the use of the app by each
participant with objective measures.
Participants’ situational motivation toward the weight-loss
program was assessed using an adapted version of the SIMS.
The SIMS typically measures four types of motivation—intrinsic
motivation, identified regulation, external regulation, and
amotivation—to engage in a task (ie, the weight-loss program)
at a specific point in time, with four items per subscale. The
SIMS has demonstrated acceptable levels of reliability and
validity in past research. The Body Shape Questionnaire-8C
(BSQ-8C) is a brief version of the Body Shape Questionnaire
(BSQ) consisting of eight items extracted from the full version
measuring the extent of psychopathology of concerns about
body shape. Higher values on the BSQ indicated more body
dissatisfaction. Depression was assessed using the Korean
version of the Beck Depression Inventory-II (K-BDI-II) scoring
system. A total score of 0-9 indicated no depression, 10-15
indicated mild depression, 16-23 indicated moderate depression,
and 24-63 indicated severe depression. Anxiety was measured
using the 20-item Trait Anxiety Inventory (TAI) of the
State-Trait Anxiety Inventory, with higher scores indicating
greater trait anxiety. The Rosenberg Self-Esteem Scale (RSES)
measure of self-esteem was used in this research with a 10-item
scale consisting entirely of negatively worded items. Thus,
higher scores implied lower self-esteem. Eating behavior notions
were measured with the Dutch Eating Behavior Questionnaire
(DEBQ), which identifies three distinct psychologically based
eating behaviors: restrained eating, emotional eating, and
external eating. It contains 33 items, with higher scores
indicating a greater tendency to present subscale behavior. The
frequency of occurrence of automatic negative thoughts
associated with depression was assessed by the Automatic
Thoughts Questionnaire (ATQ-30). The scores ranged from 30
to 150, where higher scores indicated more frequent automatic
negative thoughts. All the psychological questionnaires were
in Korean.
Interventions
The intervention of this study was a multifactorial, daily-based
personalized coaching program implemented by a psychologist
using CBT modules via the digital platform. The digital CBT
contents were based on programs proposed to clinicians [16]
as a guide. We monitored and assessed various factors related
to the behavior, cognition, mood, and motivation of each
participant assigned to the digital CBT group.
The following were assessed every day using responses to
questions and scores from the questionnaires: eating behaviors
(eg, Where did you eat? What type of food did you have? How
fast did you eat? and What time did you eat?), automatic
thoughts (eg, What came to your mind when you were eating
or thinking of food?), mood (eg, Score your mood from 0 to
100 regarding each type of negative mood: irritated, lonely,
anxious, bored, and depressed), and motivation (eg, Score your
status from 0 to 10 based on the following items: willingness
to lose weight, importance of losing weight, assurance of losing
weight, and helpfulness of this program to lose weight). Scores
were used to individually track the daily patterns of the four
factors—eating behaviors, automatic thoughts, mood, and
motivation—and provide individualized interventions. As such,
participants in the digital CBT group received daily self-report
assessments in a Google survey form via text message on their
phone. Participants were also instructed to log their dietary
intake and physical exercise on a daily basis. Additionally, they
were asked to measure their weight, BMI, and fat mass twice a
week with the InBody H20B analyzer as soon as they woke up
in the morning and were instructed to log their meals and
physical activity by self-report on the Noom Coach app on a
weekly basis.
After participants’ responses to the components related to the
four factors were collected, digital mobile tools collected the
data to allow the therapist to securely monitor participants’
progress through a Web-based dashboard. The participants
received at least three individual messages from the coach every
day, except on weekends and holidays, via the Noom Coach
app. Furthermore, the therapist individually sent a daily report,
a weekly report, and a midweek report (ie, Week 4) to the
participants for the purpose of goal setting and to strengthen
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 4http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
their motivation. Weekly group missions were provided to the
digital CBT group based on the expectation that social supports
(eg, communicating needs and building positive support) would
intensify the motivation. When the participants were inactive
for more than 3 consecutive days or asked for thorough
counseling, the therapist phoned them and conducted
motivational interviews. The motivational interviews could be
implemented only once a week per person. The duration of the
phone call did not exceed 15 minutes.
All contents of the coaching messages, group missions, and
articles were managed by a supervisor of the digital health care
coach, who has a master-level degree in clinical psychology.
She has trained as a behavioral therapist using CBT modules,
such as self-monitoring, goal setting, problem solving,
nutritional and physical activity education, stimulus control,
challenging automatic thoughts, thought restructuring, and
relapse prevention. Throughout the intervention, we expected
the participants in the digital CBT group to experience a lifestyle
change by finding a healthy pattern of living that fit each
participant’s context. The diagram of the digital CBT process
and features of the digital platform are presented in Figure 2
and Figure 3, respectively.
Figure 2. Diagram of the digital cognitive behavioral therapy (CBT) process.
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 5http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
Figure 3. Screenshots of the digital platform (ie, mobile apps) for the participants (top) and screenshots of the digital platform (ie, dashboard) for the
therapist (ie, clinical psychologist) (bottom).
Statistical Analysis
The sample size was selected to provide the study with a
statistical power of 80% to detect clinically meaningful mean
differences in weight loss of 5 kg with an SD of 7 kg in
treatment effect, based on previous studies [17]. Assuming an
average attrition rate of 10%, a sample of at least 70 subjects
was selected. For differences in baseline characteristics,
independent-sample ttests were used for continuous variables
and a chi-square test of independence was used for categorical
data assessing the demographic patterns of subjects.
We conducted the analysis following per-protocol principles.
The participants who attended at either 8 or 24 weeks were
included in the analysis of the applicable period without missing
imputations. There were no outliers in the dataset. To investigate
differences in the outcomes between the two groups, changes
in the outcomes of weight, BMI, and fat mass were analyzed
using an independent-sample ttest. To investigate statistical
differences between baseline and postintervention within a
group, a paired ttest was used. To detect statistical differences
of the proportion within the thresholds and engagement rates
between groups, a chi-square test was used. Correlation analysis
using the Pearson correlation coefficient was used to investigate
which variables at the baseline had a predictive role in changes
in anthropometrics at 8 and 24 weeks. Receiver operating
characteristic (ROC) curve analysis was undertaken to identify
the optimum trade-off between sensitivity and specificity for
cutoffs in weight-change distribution. For the ROC analysis in
this study, we set a cutoff of 3% loss of initial body weight as
a good response at 24 weeks for the digital CBT group data.
The Youden index was used for the optimal cutoff. The results
regarding the proportion of people who reached 5% weight-loss
threshold are also reported to permit comparison with other
previous studies. All analyses were conducted using SPSS
Statistics for Windows, version 20 (IBM Corp), and statistical
significance of two-tailed Pvalues were set at .05. For multiple
comparison correction, a threshold of P<.001 was used (ie, the
Pvalue threshold of .05 divided by 42, corresponding to two
different time periods and 21 phenotypes).
Results
Overview
There were no significant differences between the randomization
groups on key demographic characteristics (see Table 1).
However, the DEBQ emotional eating scale (DEBQ-EM)
(P=.001) and the DEBQ external eating scale (DEBQ-EX)
(P=.049) scores of the two groups did differ at baseline. These
differences between the groups were found after lots were drawn
for the randomized control procedure. Participants had a mean
age of 21.8 years (SD 3.3) and a mean BMI of 28.0 kg/m2(SD
3.2).
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 6http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
Table 1. Baseline characteristics of participants in both groups.
Digital CBTa(ie, app + human CBT)
(n=45)
Control (ie, app only)
(n=25)Characteristic
22.3 (3.5)21.0 (2.7)Age (years), mean (SD)
Anthropometric measures, mean (SD)
74.5 (9.0)71.9 (7.7)Weight (kg)
28.2 (3.4)27.7 (2.9)
BMI (kg/m2)
30.2 (6.8)29.3 (6.0)Fat mass (kg)
40.4 (5.4)40.5 (4.8)Fat percent (%)
24.0 (2.6)23.8 (3.3)Lean body mass (kg)
Blood measures, mean (SD)
87.3 (7.4)87.0 (8.1)Fasting glucose (mg/dL)
93.2 (42.6)92.2 (35.9)Triglyceride (mg/dL)
191.1 (30.4)184.7 (24.9)Total cholesterol (mg/dL)
15.3 (11.9)12.7 (6.9)Alanine aminotransferase (U/L)
16.9 (4.8)17.0 (4.7)Aspartate aminotransferase (U/L)
21.3 (32.8)15.3 (8.5)Gamma-glutamyl transpeptidase (U/L)
42.5 (15.3)37.5 (14.7)Leptin (ng/mL)
16.1 (9.1)12.6 (6.1)Fasting insulin (µU/mL)
3.5 (2.1)2.8 (1.5)
Homeostasis Model for Assessment of Insulin Resistanceb, mean (SD)
Scale or questionnaire (score), mean (SD)
76.1 (5.7)77.0 (5.8)Situational Motivation Scale
36.2 (7.5)34.8 (8.9)Body Shape Questionnaire-8C
13.6 (9.0)14.7 (9.6)Beck Depression Inventory-II in Korean
48.0 (10.4)47.8 (11.0)Trait Anxiety Inventory
19.8 (5.6)21.9 (6.4)Rosenberg Self-Esteem Scale
29.9 (6.6)30.6 (7.3)
DEBQcrestrained eating scale
38.0 (10.1)29.1 (11.6)
DEBQ emotional eating scaled
34.9 (4.8)32.0 (7.0)
DEBQ external eating scaled
57.2 (22.3)57.6 (26.0)Automatic Thoughts Questionnaire
3.0 (1.7)2.2 (1.7)Yale Food Addiction Scale
Residence status, n (%)
27 (60)10 (40)Living with family
8 (18)8 (32)Living alone
9 (20)7 (28)Living with roommates
1 (2)0 (0)Others
Number of attempts to lose weight by different methods, n (%)
1 (2)0 (0)None
4 (9)3 (12)Once
15 (33)12 (48)Twice
13 (29)3 (12)Three times
8 (18)4 (16)Four times
4 (9)2 (8)Five times
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 7http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
Digital CBTa(ie, app + human CBT)
(n=45)
Control (ie, app only)
(n=25)Characteristic
0 (0)1 (4)Six times
aCBT: cognitive behavioral therapy.
bInsulin resistance = (insulin [µU/mL] × glucose [mg/dL]) / 405.
cDEBQ: Dutch Eating Behavior Questionnaire.
dThere was a statistical difference between the two groups at baseline.
Primary Outcome of Weight Change and
Anthropometric Outcomes
The primary outcome (ie, weight change) was assessed at two
time points—immediately after lifestyle change with digital
CBT (8 weeks) and at the long-term follow-up without digital
CBT (24 weeks)—to investigate the self-sustaining effect of
lifestyle change induced by 8 weeks of digital CBT. Of the 70
randomized participants, 65 (93%) were assessed for the primary
outcome—body weight—at 24 weeks and 5 (7%) were lost to
follow-up. Figures 4 and 5represents the mean weight change
along with other anthropometric measures—BMI, body fat
mass, and body lean mass—at each study time point. Participants
in the digital CBT group showed significant changes in mean
body weight at 8 weeks compared to the control group (–3.1%,
SD 4.5, vs –0.7%, SD 3.4, P=.04) but not at 24 weeks. The
proportion of subjects who showed good response was 45%
(17/38) in the digital CBT group and 29% (6/21) in the control
group at 8 weeks (P=.22), while at 24 weeks it was 54% (22/41)
in the digital CBT group and 42% (10/24) in the control group
(P=.35). In addition, the number reaching the conventional 5%
weight loss from the baseline in the digital CBT group was
significantly higher than in the control group at 8 weeks (12/38,
32%, vs 1/21, 4%, P=.02) but not at 24 weeks (18/41, 44%, vs
7/24, 29%, P=.24). Changes in mean BMI (–3.1%, SD 4.6, vs
–0.7%, SD 3.5, P=.04) and body fat mass (–6.3%, SD 8.8, vs
–0.8%, SD 8.1, P=.02) of the digital CBT group were also
significant compared to the control group at 8 weeks but not at
24 weeks (see Multimedia Appendix 1, Table MA1-1). Body
lean mass did not significantly differ between the two groups
at both 8 and 24 weeks. Examining within-group changes, only
the digital CBT group achieved significant weight changes, as
well as BMI and body fat mass, at both 8 and 24 weeks; the
digital CBT group achieved significant changes in lean body
mass at 24 weeks but not at 8 weeks (see Multimedia Appendix
1, Tables MA1-2 and MA1-3).
Figure 4. Patterns of changes in mean body weight (A), BMI (B), body fat mass (C), and lean body mass (LBM) (D). CBT: cognitive behavioral
therapy. *P<.05; **P<.01.
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 8http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
Figure 5. Weight change based on individual data from the experimental group at the 8-week follow-up (A), from the experimental group at the 24-week
follow-up (B), from the control group at the 8-week follow-up (C), and from the control group at the 24-week follow-up (D). CBT: cognitive behavioral
therapy.
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 9http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
Figure 6. Changes in meal calories between experimental and control groups during the intervention period, as well as the contrast of mean energy
intake between groups. *P<.05; ** P<.01.
Secondary Outcomes: Metabolic and Psychological
Outcomes
Multimedia Appendix 1, Table MA1-4, shows a comparison of
the metabolic outcomes from baseline to 8 weeks in each group
and by intervention condition. The mean decreases in leptin
(–15.8%, SD 29.9, vs 7.2%, SD 35.9, P=.01), insulin (–4.4%,
SD 35.2, vs 15.4%, SD 35.1, P=.048), and Homeostatic Model
Assessment for Insulin Resistance (HOMA-IR) (–7.1%, SD
35.1, vs 14.4%, SD 41.2, P=.04) were significantly greater in
the digital CBT group than in the control group. For
within-group analysis, the changes in glucose (–2.91%, P=.04)
and leptin (–15.82%, P=.003) of the digital CBT group were
significant. No significant outcome changes were found in the
control group. The mean percentage changes in psychological
outcomes are shown in Multimedia Appendix 1, Table MA1-5,
by intervention condition. There was no significant difference
between the groups regarding the number of changes in
psychological outcomes except for the change in the DEBQ-EM
from baseline to 8 weeks (P=.048). Paired ttest analysis showed
significant changes in the BSQ-8C and DEBQ-EX scores at 8
and 24 weeks in both groups. However, the changes in the scores
of the DEBQ restrained eating scale (DEBQ-RE) (P<.001) at
8 weeks, and those of the K-BDI-II (P=.001), TAI (P=.04),
RSES (P=.03), and ATQ-30 (P=.02) at 24 weeks, appeared to
be significant only in the digital CBT group (see Multimedia
Appendix 1, Tables MA1-6 and MA1-7). Behavioral outcomes,
measured via the Noom app, are represented as the amount of
calorie intake and the pattern of weekly changes between the
groups and the average energy intake of each group, as presented
in Figure 3. Mean snack calories (P=.02) significantly differed
between the two groups, and total calories (P=.06) had a
tendency toward critical difference by intervention condition
(see Figure 6 and Multimedia Appendix 1, Table MA1-8).
Lastly, the digital CBT group had a higher engagement rate
when using digital tools than the control group, though it
declined over time in both groups (see Figure 7and Multimedia
Appendix 1, Table MA1-9).
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 10http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
Figure 7. Patterns of changes in engagement rate of the experimental and control groups during the intervention period. *P<.05.
Predictors of the Primary Outcome, Weight Change
Correlations Between the Primary Outcome and the
Baseline Characteristics
The baseline motivation, as measured by the SIMS, was
significantly correlated with weight change at 8 weeks (P=.009)
and 24 weeks (P=.003). Depression, as measured by the
K-BDI-II (P=.03); anxiety, as measured by the TAI (P=.008);
and self-esteem, as measured by the RSES (P=.002) at baseline
also showed a significant correlation with weight change at 24
weeks but not at 8 weeks. Depression, anxiety, self-esteem,
restrained eating behavior, external eating behavior, and
automatic thoughts at baseline were significantly correlated
with BMI change at 24 weeks. Lastly, lean body mass, anxiety,
and self-esteem at baseline were significantly correlated with
change in body fat mass at 24 weeks. Figure 8 illustrates the
significant correlations between the predictive markers and the
change of the anthropometric measures at 24 weeks. All the
results of the correlation analysis are presented in detail in
Multimedia Appendix 1, Table MA1-10. Multimedia Appendix
1, Figure MA1-1, also illustrates the correlations between
predictive markers and the change of BMI.
Figure 8. The correlation between weight change at the long-term follow-up period (24 weeks) and the level of motivation, self-esteem, depression,
and anxiety at baseline. Also shown are the correlation between BMI change at the long-term follow-up and the level of motivation at baseline, and the
correlation between fat mass change at the long-term follow-up and lean body mass at baseline. K-BDI: Korean version of the Beck Depression Inventory;
RSES: Rosenberg Self-Esteem Scale; SIMS: Situational Motivation Scale; TAI: Trait Anxiety Inventory.
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 11http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
Receiver Operating Characteristic Analysis Determining
the Optimal Cutoff Scores of the Predictive Markers of
Success in Weight Loss by Digital Cognitive Behavioral
Therapy
Multimedia Appendix 1, Table MA1-11, shows the sensitivity
and specificity of the baseline psychological characteristics
showing significant correlations with weight change, the primary
outcome. The definition of optimal statistical prediction
threshold is weight loss of more than 3% of the initial body
weight. This is an important threshold because our treatment
was CBT as a lifestyle modification without any biological
intervention. Both motivation and self-esteem had the greatest
area under the curve (AUC) (0.63). The AUCs of depression
and anxiety were 0.61 and 0.62, respectively. To predict a good
response, the cutoff for motivation (SIMS score=76.5) provided
a good trade-off between sensitivity (59%) and specificity
(74%). Additionally, the cutoff for depression (K-BDI-II
score=7.5), anxiety (TAI score=41.5), and self-esteem (RSES
score=24.5) provided optimal sensitivity and specificity to
predict a good response. Overall, motivation showed the best
predictive performance.
Clinical Efficacy of Digital Cognitive Behavioral
Therapy Based on the Optimal Cutoff Scores of the
Predictive Markers in the Clinical Setting
The high-motivation subgroup (SIMS scores >76.5) showed a
65% (13/20) probability of successful 3% weight loss, whereas
the low-motivation subgroup (SIMS scores <76.5) showed a
36% (9/25) probability of successful 3% weight loss. Optimal
predictive performance was achieved by combining both
motivation and depression scores. The high-motivation plus
low-depression subgroup (SIMS scores >76.5 and K-BDI-II
scores <7.5) showed a 100% (6/6) probability of successful 3%
weight loss. Other subgroups showed a lower probability of
successful 3% weight loss: 55% (5/9) of the low-motivation
and low-depression subgroup, 50% (7/14) of the high-motivation
and high-depression subgroup, and 25% (4/16) of the
low-motivation and high-depression subgroup (see Figure 9).
Figure 9. The clinical efficacy of digital cognitive behavioral therapy (CBT) by applying the optimal cutoff scores of the predictive markers in the
clinical setting. The pink line represents the threshold for successful weight loss.
Even when the strict statistical threshold for multiple comparison
corrections was applied, changes in weight, BMI, and fat mass
from baseline to 8 weeks in the digital CBT group were
considered significant (P<.001). The changes in the scores of
the DEBQ-RE from baseline to 8 weeks, and in the K-BDI-II
and DEBQ-EX scores from baseline to 24 weeks, in the digital
CBT group were also significant after multiple corrections
(P<.001). Furthermore, the changes in the scores of the BSQ-8C
from baseline to 8 weeks and 24 weeks in both the digital CBT
and control groups were considered significant after multiple
corrections (P<.001).
Discussion
Principal Findings
This study successfully examined the efficacy of a newly
developed, multifactorial, and daily-based personalized CBT
model conducted by a psychologist via a digital platform for
managing body weight, BMI, and body fat mass and showed a
legacy effect even after the intervention terminated. This was
performed by comparing this group to the active comparators
using only the app as the control group. Furthermore, this study
successfully explored the predictors for the efficacy of digital
CBT from the baseline characteristics and recommended them
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 12http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
as precision medicine biomarkers, namely, depression, anxiety,
self-esteem, and motivation.
Among mobile health (mHealth) RCTs for obesity, this study
has unique implications regarding the application of CBT
strategies by a human coach in the intervention. This study,
therefore, contributes to the broader literature on weight-loss
treatments that involve human factors. There have been
widespread studies of mHealth approaches to weight-loss
programs [18-33]. There are several studies of obesity treatments
that did not investigate CBT settings; these include studies of
human-based mHealth RCTs [18,21,24,29,34,35], human-based
mHealth but without RCT design [17,26,30], and mHealth not
based on human factors but with RCT design
[19,20,22,25,27,28,32,33]. There are also several studies of
human-based RCT designs, including CBT settings for obesity
but not mHealth procedures (ie, telephone, website, face-to-face,
and others) [16,36-40].
This study is comparable to other mHealth RCTs. The mean
percentage weight loss of our study was 4% of initial body
weight, and previous mHealth RCTs reported a mean percentage
weight loss ranging from 1% to 3% [24,29,32,34]. Moreover,
this study successfully showed weight maintenance. Most
interventions for obesity have shown a tendency to regain weight
after discontinuing the treatment [11,16,24,29,32,41-43], but
our digital CBT intervention showed a sustained trend of further
decrease even up to 16 weeks after cessation of the 8-week
intervention. This affords solid support for the assumption that
digital CBT promotes an overall healthy lifestyle. However,
because we do not have data beyond a 1-year period, a direct
comparison with previous studies is not feasible. Preventing
weight regain at 24 weeks is closely related with a decrease in
body fat mass and an increase in lean body mass at 8 weeks,
which are relevant to physical activity rate and nutrition status
[44,45]. Indeed, an improvement in both physical activity and
diet, representing changes in lifestyle, leads to healthy body
composition. Therefore, the patterns of changes in not only
body weight but also body fat mass and lean body mass may
imply that the participants in the digital CBT group experienced
self-sustainable transitions in daily decision making for a healthy
life.
With regard to the appropriate threshold, previous behavioral
weight-loss studies often reported 5% weight loss in the majority
of participants [16,26,30]. Conventionally, several studies
adopted a 5% threshold as a clinically significant threshold
[16,19,26,29]. However, in contrast to the conventional 5%
threshold, we adopted a tempered 3% weight-loss threshold as
the good response threshold for two main reasons. First, the
duration of the active intervention period in this study was
shorter than in other studies and only persisted transiently for
the initial 2 months. The majority of previous behavioral studies
had a full 6-month active intervention design [16,36,37].
However, the duration of the active intervention period in our
study was only 8 weeks (2 months). There was no intervention
delivered after 8 weeks (2 months) until the 6-month time point.
Thus, the subjects did not receive the intervention during the
remaining 4 months after the initial 2-month active intervention.
Second, the components of the intervention in this study did
not include extreme restrictions or requirements in either diet
or exercise. The main goal of our intervention was to implement
sustainable weight management skills by learning an appropriate
behavioral process as well as establishing new cognitive
processes. Therefore, the weight loss per se could be weaker
than with the stringent diet restrictions and exercise requirements
of a behavioral program during the intervention. In addition to
the 3% threshold, we also reported the results based on the
conventional 5% threshold to allow a direct comparison of
clinical efficacy between studies.
Regarding personalization, our digital CBT was fully tailored
to each participant’s characteristics in multifactorial domains:
the behavioral, cognitive, emotional, motivational, and physical
domains. The therapist in our study altered the feedback styles
based on data from five types of domains for every participant
and conducted intensive daily monitoring. Most of the previous
RCTs on mHealth interventions for obesity—those not based
on human factors—considered one or two factors of individual
symptoms that led to the implementation of homogeneous
interventions [18,19,22,31]. Although there are some
interventions that use custom algorithms to provide
individualized feedback, they only focus on diet, physical
activity, weight loss, or any two of these [27,28,32,46].
Furthermore, some earlier mHealth RCTs for obesity based on
human factors only dealt with diet and physical activities
[21,24]. One study managed three domains for the intervention:
diet, physical activities, and eating behaviors [29]. However,
instructions on behavior change strategies were not delivered
by smartphone but by attending weekly group sessions for the
first phase of the intervention. The study was deficient in other
principal factors, such as emotional, cognitive, and motivational
domains, implying insufficient potentiality for long-term
lifestyle change. Because cognitive conceptualization and
emotional regulation process are naturally associated with
behavioral patterns, consideration of all these components can
allow changes in one’s lifestyle and ultimately solve problems
related to obesity [47]. Therefore, it is important to address the
respective multifactorial domains so as to conduct tailored
treatment for individuals with fully integrated techniques. Our
digital CBT strategies operate in a fully comprehensive system
that deals with behavioral, cognitive, emotional, motivational,
and physical factors and allows integrated mediation to
successfully manage obesity.
After examining aspects of temporal strategies for intervention,
we arranged three different time points (ie, daily, weekly, and
monthly points) and initiated a daily human-agent intervention
in an mHealth RCT for obesity. All previous face-to-face,
electronic health (eHealth), and mHealth RCTs for obesity
treatment have been either weekly- or monthly-based
interventions delivered by therapists [7,16,21,24,25,29,36-38].
Temporal strategies can influence the engagement rate, which
is closely related to treatment outcomes [48]. Unfortunately,
according to a systematic review, mHealth RCTs related to
weight-loss programs suffer from a high attrition rate of more
than 30% [49]. Our digital CBT trial, however, showed high
in-app activity rates as well as engagement in the intervention
program. Only one participant in the digital CBT group had to
withdraw for personal reasons; 80% of the participants were
active until the end of the treatment session. One possible reason
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 13http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
for these outcomes is that our digital CBT intervention
effectively managed participants’ motivation to lose weight as
well as participate in the program. Our intervention did this by
delivering individualized messages every day, based on data
from the in-app database and daily assessment of various
psychological factors using CBT modules, as well as facilitating
real-time access to the therapist. Additionally, the midreport,
employed as a monthly intervention, allowed personalized
precision treatment based on initial psychological conditions
to keep participants motivated. Thus, the engagement rate of
the digital CBT group improved from 91% to 93%, whereas
the engagement rate in the control group dropped from 84% to
76% between Week 4 and Week 5.
Through our digital CBT, changes in biological indexes, leptin,
insulin, and HOMA-IR indicated that factors related to physical
health can be successfully improved. Moreover, we also
successfully managed motivation, emotion, cognition, and
behavior. The level of self-body-image satisfaction and external
eating behaviors was improved in both groups. This indicates
that simply including the standard mHealth treatment in the
control group in our study was practical for improving body
image perception and external eating habits. Digital CBT
improved the level of depression, anxiety, self-esteem, and
automatic thoughts related to depression. In fact, the DEBQ-EM
and DEBQ-EX scores showed a significant difference between
the two groups at baseline but were not notably correlated with
the primary measures at baseline. This may be considered a
random circumstance of randomization. Therefore, these
differences can be interpreted as not affecting the main outcomes
of our study. Furthermore, a significant difference in reported
snack calorie intake between the two groups suggests that our
digital CBT intervention had an impact on managing snack
calories compared to other meals. Stress is highly correlated
with the frequency of snacks [50]. Thus, it is possible that our
digital CBT intervention affected snack calorie intake by finding
individualized stress coping strategies, restructuring cognitive
structures of automatic negative eating or weight-related
thoughts, and developing regular and balanced eating behaviors.
Therefore, this provides evidence that the participants in the
digital CBT group changed their lifestyle to constantly manage
their weight.
This study can be considered a practical one because it explored
clinical markers that predict the effect of digital CBT and
suggested plausible criteria that can be applied to clinical
settings. The follow-up results at 24 weeks in this study showed
that the levels of motivation, depression, anxiety, and
self-esteem were the predictive markers of weight loss based
on the digital CBT intervention. Some of our results regarding
the predictors of weight control conflict with the findings of
previous research [51], but they are consistent with recent
findings that the level of motivation is the strongest predictive
trait for weight control [52,53]. We defined people who lost
less than 3% of their baseline weight as poor responders to the
treatment. Thus, people with a SIMS score lower than 76.5 are
recommended to find and pursue their own way of enhancing
their motivation to lose weight before they undertake digital
CBT. Furthermore, a person whose score is higher than 7.5 on
the K-BDI-II, 41.5 on the TAI, or 24.5 on the RSES is
encouraged to handle the relevant issue before, or at the same
time as, digital CBT. This will prevent further distress from
repeated failure to control weight, save limited resources, and
allow better concentration in individuals with a higher chance
of success in weight control.
Considering the comparator of this study as the best active
comparator without human coaching, digital CBT is a competent
intervention for obesity in the current situation in the digital
health care industry. We provided education on how to log
meals and exercise as well as how to use InBody Dial and the
mobile app, not only to the digital CBT group but also to the
control group during the orientation. Thus, the control group in
this study can be defined as an active group as in previous
studies [19,22,31]. As expected, the control group in this study
showed favorable results. Therefore, the results of this study
are superior and significant compared to those of previous
studies of digital health care interventions.
Limitations
While the results are highly promising, the study is not without
limitations. First of all, the participants were limited to those in
their 20s and 30s, resulting in limited generalizability. Second,
since this is not a blinded study, an observer bias could have
been generated. Thus, an implication of this study that should
be noted is that it tested the digital CBT and did not validate it.
Third, the sample size was relatively small (N=70). Therefore,
most of the results did not pass the strict
multiple-comparison-corrected Pthreshold. Fourth, the
follow-up period needs to be extended to increase the reliability
and validity of our results. Accordingly, we recommend that
future studies examine more information on personal
characteristics, such as single nucleotide polymorphisms (SNPs)
and daily patterns of digital phenotypes for individuals within
in-app data, in order to enhance the interpretation of the efficacy
of digital-based interventions. Fifth, the total amount of food
calories in the app might have been underestimated because the
amount per serving for diverse types of food was not precise
and people may have miscalculated their food intake. The
primary reason for errors in food records is that most people
have difficulties in estimating food portions [54]. The
discrepancy in food choice between the food diary and actual
meals (ie, recording similar but not exact menus, skipping
reports of foods eaten, or logging foods not offered) could
explain the remainder of the total miscalculation [55]. Thus, we
suggest that a direct assessment of food choice and intake, such
as buffet tests, should be performed in parallel with logging
intake in the food diary on the app for future research. In
addition, it should be noted that it is necessary to involve
dietitians on multidisciplinary health care teams for obesity
CBT, as their evaluations of dietary assessment and nutritional
advice would greatly strengthen the efficacy of the intervention.
Lastly, there is a feasibility issue regarding the digital CBT of
this study since it is intensive and costly, requiring daily
intervention by therapists trained in both physical and mental
health care. Therefore, more research involving human factors
in technology-based treatments should be conducted to collect
enough data to create automatic functions, thereby decreasing
the burdens of therapists in the future.
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 14http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
Conclusions
For the first time, we discovered that human-based digital CBT
is capable of treating obesity using digital tools. Anthropometric
measures, such as body weight and body compositions, were
comparably improved by the digital CBT model as well as
physiological indices and obesity-related psychological factors.
There was no relapse in weight change after the end of the
intervention. We also found predictable psychological markers
to estimate the efficacy of the digital CBT treatment for obesity.
This will open up new aspects of digital precision remedies for
obesity in the digital health care industry.
Acknowledgments
We would like to thank Hyunjae Kim, the chief executive manager of LookinBody, for supporting this study by sharing his
company’s advanced digital technologies. Also, I would like to thank Ms Younghyun Yun from the Department of Anatomy and
Cell Biology, Seoul National University College of Medicine, for the preparation of the excellent illustrations and graphic design.
This study was supported by a grant from the National Research Foundation of Korea (NRF), funded by the Korean Government,
Ministry of Science and ICT (MSIT) (No. NRF-2018R1A5A2025964), and was supported by the Creative-Pioneering Researchers
Program through Seoul National University (SNU).
Noom provided the funding to conduct this research and InBody provided body composition analyzer devices for this research.
Representatives of InBody had no role in the management, analysis, and interpretation of the data; preparation, review, or approval
of the manuscript; and decision to submit the manuscript for publication. YK, an employee of Noom, participated in the generation
of the study design and in data collection.
Authors' Contributions
MK conceptualized and designed the clinical infrastructure for the digital CBT intervention during the implementation phase.
HJC and SC gave valuable research insights when designing the digital CBT intervention. MK, HJC, YK, YG, MN, SL, and YL
contributed to the study design and data collection. MK, HJC, YG, and SC analyzed and interpreted the data. MK wrote the
manuscript and edited the contents of the manuscript. HJC and SC reviewed the manuscript. All authors approved the final version
of the manuscript for submission.
Conflicts of Interest
YK is an employee of Noom.
Multimedia Appendix 1
Changes in outcomes from baseline, correlations, and predicting efficacy of digital cognitive behavioral therapy.
[DOCX File , 200 KB-Multimedia Appendix 1]
Multimedia Appendix 2
CONSORT-eHEALTH checklist (V 1.6.1).
[PDF File (Adobe PDF File), 1466 KB-Multimedia Appendix 2]
References
1. Gardner CD, Trepanowski JF, Del Gobbo LC, Hauser ME, Rigdon J, Ioannidis JP, et al. Effect of low-fat vs low-carbohydrate
diet on 12-month weight loss in overweight adults and the association with genotype pattern or insulin secretion: The
DIETFITS randomized clinical trial. JAMA 2018 Feb 20;319(7):667-679 [FREE Full text] [doi: 10.1001/jama.2018.0245]
[Medline: 29466592]
2. Wyatt HR. Update on treatment strategies for obesity. J Clin Endocrinol Metab 2013 Apr;98(4):1299-1306 [FREE Full
text] [doi: 10.1210/jc.2012-3115] [Medline: 23443815]
3. Sjöström L, Narbro K, Sjöström CD, Karason K, Larsson B, Wedel H, Swedish Obese Subjects Study. Effects of bariatric
surgery on mortality in Swedish obese subjects. N Engl J Med 2007 Aug 23;357(8):741-752. [doi: 10.1056/NEJMoa066254]
[Medline: 17715408]
4. Castelnuovo G, Pietrabissa G, Manzoni GM, Cattivelli R, Rossi A, Novelli M, et al. Cognitive behavioral therapy to aid
weight loss in obese patients: Current perspectives. Psychol Res Behav Manag 2017;10:165-173 [FREE Full text] [doi:
10.2147/PRBM.S113278] [Medline: 28652832]
5. Cooper MJ, Wells A, Todd G. A cognitive model of bulimia nervosa. Br J Clin Psychol 2004 Mar;43(Pt 1):1-16. [doi:
10.1348/014466504772812931] [Medline: 15005903]
6. Brennan L. Cognitive Behavioural Evaluation and Treatment of Adolescent Overweight and Obesity [doctoral thesis].
Melbourne, Australia: RMIT University; 2006 Aug 31. URL: http://researchbank.rmit.edu.au/eserv/rmit:6333/Brennan.pdf
[accessed 2006-08-31]
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 15http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
7. Tsiros MD, Sinn N, Brennan L, Coates AM, Walkley JW, Petkov J, et al. Cognitive behavioral therapy improves diet and
body composition in overweight and obese adolescents. Am J Clin Nutr 2008 May;87(5):1134-1140. [doi:
10.1093/ajcn/87.5.1134] [Medline: 18469231]
8. Steele RM, Mummery WK, Dwyer T. A comparison of face-to-face or internet-delivered physical activity intervention on
targeted determinants. Health Educ Behav 2009 Dec;36(6):1051-1064. [doi: 10.1177/1090198109335802] [Medline:
19502534]
9. Sherwood NE, Morton N, Jeffery RW, French SA, Neumark-Sztainer D, Falkner NH. Consumer preferences in format and
type of community-based weight control programs. Am J Health Promot 1998;13(1):12-18. [doi: 10.4278/0890-1171-13.1.12]
[Medline: 10186930]
10. Klasnja P, Pratt W. Healthcare in the pocket: Mapping the space of mobile-phone health interventions. J Biomed Inform
2012 Feb;45(1):184-198 [FREE Full text] [doi: 10.1016/j.jbi.2011.08.017] [Medline: 21925288]
11. Yon BA, Johnson RK, Harvey-Berino J, Gold BC. The use of a personal digital assistant for dietary self-monitoring does
not improve the validity of self-reports of energy intake. J Am Diet Assoc 2006 Aug;106(8):1256-1259. [doi:
10.1016/j.jada.2006.05.004] [Medline: 16863723]
12. Fry JP, Neff RA. Periodic prompts and reminders in health promotion and health behavior interventions: Systematic review.
J Med Internet Res 2009 May 14;11(2):e16 [FREE Full text] [doi: 10.2196/jmir.1138] [Medline: 19632970]
13. Chen J, Cade JE, Allman-Farinelli M. The most popular smartphone apps for weight loss: A quality assessment. JMIR
Mhealth Uhealth 2015 Dec 16;3(4):e104 [FREE Full text] [doi: 10.2196/mhealth.4334] [Medline: 26678569]
14. Hey SP, Kimmelman J. The questionable use of unequal allocation in confirmatory trials. Neurology 2014 Jan 07;82(1):77-79
[FREE Full text] [doi: 10.1212/01.wnl.0000438226.10353.1c] [Medline: 24306005]
15. Michaelides A, Major J, Pienkosz E, Wood M, Kim Y, Toro-Ramos T. Usefulness of a novel mobile diabetes prevention
program delivery platform with human coaching: 65-week observational follow-up. JMIR Mhealth Uhealth 2018 May
03;6(5):e93 [FREE Full text] [doi: 10.2196/mhealth.9161] [Medline: 29724709]
16. Cooper Z, Doll HA, Hawker DM, Byrne S, Bonner G, Eeley E, et al. Testing a new cognitive behavioural treatment for
obesity: A randomized controlled trial with three-year follow-up. Behav Res Ther 2010 Aug;48(8):706-713 [FREE Full
text] [doi: 10.1016/j.brat.2010.03.008] [Medline: 20691328]
17. Mao AY, Chen C, Magana C, Caballero Barajas K, Olayiwola JN. A mobile phone-based health coaching intervention for
weight loss and blood pressure reduction in a national payer population: A retrospective study. JMIR Mhealth Uhealth
2017 Jun 08;5(6):e80 [FREE Full text] [doi: 10.2196/mhealth.7591] [Medline: 28596147]
18. Turner-McGrievy GM, Beets MW, Moore JB, Kaczynski AT, Barr-Anderson DJ, Tate DF. Comparison of traditional
versus mobile app self-monitoring of physical activity and dietary intake among overweight adults participating in an
mHealth weight loss program. J Am Med Inform Assoc 2013 May 01;20(3):513-518 [FREE Full text] [doi:
10.1136/amiajnl-2012-001510] [Medline: 23429637]
19. Carter MC, Burley VJ, Nykjaer C, Cade JE. Adherence to a smartphone application for weight loss compared to website
and paper diary: Pilot randomized controlled trial. J Med Internet Res 2013 Apr 15;15(4):e32 [FREE Full text] [doi:
10.2196/jmir.2283] [Medline: 23587561]
20. Spring B, Duncan JM, Janke EA, Kozak AT, McFadden HG, DeMott A, et al. Integrating technology into standard weight
loss treatment: A randomized controlled trial. JAMA Intern Med 2013 Jan 28;173(2):105-111 [FREE Full text] [doi:
10.1001/jamainternmed.2013.1221] [Medline: 23229890]
21. Allen JK, Stephens J, Dennison Himmelfarb CR, Stewart KJ, Hauck S. Randomized controlled pilot study testing use of
smartphone technology for obesity treatment. J Obes 2013;2013:151597 [FREE Full text] [doi: 10.1155/2013/151597]
[Medline: 24392223]
22. Laing BY, Mangione CM, Tseng C, Leng M, Vaisberg E, Mahida M, et al. Effectiveness of a smartphone application for
weight loss compared with usual care in overweight primary care patients: A randomized, controlled trial. Ann Intern Med
2014 Nov 18;161(10 Suppl):S5-S12 [FREE Full text] [doi: 10.7326/M13-3005] [Medline: 25402403]
23. Wharton CM, Johnston CS, Cunningham BK, Sterner D. Dietary self-monitoring, but not dietary quality, improves with
use of smartphone app technology in an 8-week weight loss trial. J Nutr Educ Behav 2014;46(5):440-444. [doi:
10.1016/j.jneb.2014.04.291] [Medline: 25220777]
24. Godino JG, Merchant G, Norman GJ, Donohue MC, Marshall SJ, Fowler JH, et al. Using social and mobile tools for weight
loss in overweight and obese young adults (Project SMART): A 2 year, parallel-group, randomised, controlled trial. Lancet
Diabetes Endocrinol 2016 Sep;4(9):747-755 [FREE Full text] [doi: 10.1016/S2213-8587(16)30105-X] [Medline: 27426247]
25. Jun MK, Ha JY. Effect of smartphone apps applying BodyThink program on obesity in adolescent girls [Article in Korean].
J Korean Acad Nurs 2016 Jun;46(3):390-399. [doi: 10.4040/jkan.2016.46.3.390] [Medline: 27411766]
26. Michaelides A, Raby C, Wood M, Farr K, Toro-Ramos T. Weight loss efficacy of a novel mobile diabetes prevention
program delivery platform with human coaching. BMJ Open Diabetes Res Care 2016;4(1):e000264 [FREE Full text] [doi:
10.1136/bmjdrc-2016-000264] [Medline: 27651911]
27. Mummah SA, Mathur M, King AC, Gardner CD, Sutton S. Mobile technology for vegetable consumption: A randomized
controlled pilot study in overweight adults. JMIR Mhealth Uhealth 2016 May 18;4(2):e51 [FREE Full text] [doi:
10.2196/mhealth.5146] [Medline: 27193036]
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 16http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
28. Turner-McGrievy GM, Wilcox S, Boutté A, Hutto BE, Singletary C, Muth ER, et al. The Dietary Intervention to Enhance
Tracking with Mobile Devices (DIET Mobile) Study: A 6-month randomized weight loss trial. Obesity (Silver Spring)
2017 Aug;25(8):1336-1342 [FREE Full text] [doi: 10.1002/oby.21889] [Medline: 28600833]
29. Spring B, Pellegrini CA, Pfammatter A, Duncan JM, Pictor A, McFadden HG, et al. Effects of an abbreviated obesity
intervention supported by mobile technology: The ENGAGED randomized clinical trial. Obesity (Silver Spring) 2017
Jul;25(7):1191-1198 [FREE Full text] [doi: 10.1002/oby.21842] [Medline: 28494136]
30. Toro-Ramos T, Lee D, Kim Y, Michaelides A, Oh TJ, Kim KM, et al. Effectiveness of a smartphone application for the
management of metabolic syndrome components focusing on weight loss: A preliminary study. Metab Syndr Relat Disord
2017 Nov;15(9):465-473. [doi: 10.1089/met.2017.0062] [Medline: 29035677]
31. Turner-McGrievy G, Tate D. Tweets, apps, and pods: Results of the 6-month Mobile Pounds Off Digitally (Mobile POD)
randomized weight-loss intervention among adults. J Med Internet Res 2011 Dec 20;13(4):e120 [FREE Full text] [doi:
10.2196/jmir.1841] [Medline: 22186428]
32. Burke LE, Styn MA, Sereika SM, Conroy MB, Ye L, Glanz K, et al. Using mHealth technology to enhance self-monitoring
for weight loss: A randomized trial. Am J Prev Med 2012 Jul;43(1):20-26 [FREE Full text] [doi:
10.1016/j.amepre.2012.03.016] [Medline: 22704741]
33. Shaw RJ, Bosworth HB, Silva SS, Lipkus IM, Davis LL, Sha RS, et al. Mobile health messages help sustain recent weight
loss. Am J Med 2013 Nov;126(11):1002-1009 [FREE Full text] [doi: 10.1016/j.amjmed.2013.07.001] [Medline: 24050486]
34. Nezami BT, Ward DS, Lytle LA, Ennett ST, Tate DF. A mHealth randomized controlled trial to reduce sugar-sweetened
beverage intake in preschool-aged children. Pediatr Obes 2018 Nov;13(11):668-676. [doi: 10.1111/ijpo.12258] [Medline:
29119719]
35. Spring B, Pellegrini C, McFadden HG, Pfammatter AF, Stump TK, Siddique J, et al. Multicomponent mHealth intervention
for large, sustained change in multiple diet and activity risk behaviors: The Make Better Choices 2 randomized controlled
trial. J Med Internet Res 2018 Jun 19;20(6):e10528 [FREE Full text] [doi: 10.2196/10528] [Medline: 29921561]
36. Tate DF, Wing RR, Winett RA. Using internet technology to deliver a behavioral weight loss program. JAMA 2001 Mar
07;285(9):1172-1177. [doi: 10.1001/jama.285.9.1172] [Medline: 11231746]
37. Harvey-Berino J, Pintauro S, Buzzell P, Gold EC. Effect of internet support on the long-term maintenance of weight loss.
Obes Res 2004 Feb;12(2):320-329 [FREE Full text] [doi: 10.1038/oby.2004.40] [Medline: 14981225]
38. Tate DF, Jackvony EH, Wing RR. A randomized trial comparing human e-mail counseling, computer-automated tailored
counseling, and no counseling in an internet weight loss program. Arch Intern Med 2006;166(15):1620-1625. [doi:
10.1001/archinte.166.15.1620] [Medline: 16908795]
39. Stapleton P, Bannatyne A, Chatwin H, Urzi K, Porter B, Sheldon T. Secondary psychological outcomes in a controlled
trial of Emotional Freedom Techniques and cognitive behaviour therapy in the treatment of food cravings. Complement
Ther Clin Pract 2017 Aug;28:136-145. [doi: 10.1016/j.ctcp.2017.06.004] [Medline: 28779921]
40. Zerwas SC, Watson HJ, Hofmeier SM, Levine MD, Hamer RM, Crosby RD, et al. CBT4BN: A randomized controlled
trial of online chat and face-to-face group therapy for bulimia nervosa. Psychother Psychosom 2017;86(1):47-53 [FREE
Full text] [doi: 10.1159/000449025] [Medline: 27883997]
41. Wadden TA, Hollander P, Klein S, Niswender K, Woo V, Hale PM, NN8022-1923 Investigators. Weight maintenance and
additional weight loss with liraglutide after low-calorie-diet-induced weight loss: The SCALE Maintenance randomized
study. Int J Obes (Lond) 2013 Nov;37(11):1443-1451. [doi: 10.1038/ijo.2013.120] [Medline: 23812094]
42. Srivastava G, Buffington C. A specialized medical management program to address post-operative weight regain in bariatric
patients. Obes Surg 2018 Aug;28(8):2241-2246. [doi: 10.1007/s11695-018-3141-z] [Medline: 29464536]
43. Pareek M, Schauer PR, Kaplan LM, Leiter LA, Rubino F, Bhatt DL. Metabolic surgery: Weight loss, diabetes, and beyond.
J Am Coll Cardiol 2018 Feb 13;71(6):670-687 [FREE Full text] [doi: 10.1016/j.jacc.2017.12.014] [Medline: 29420964]
44. Vehrs PR, Hager R. Assessment and interpretation of body composition in physical education. J Phys Educ Recreat Dance
2013 Jan 26;77(7):46-51 [FREE Full text] [doi: 10.1080/07303084.2006.10597907]
45. Kyle UG, Schutz Y, Dupertuis YM, Pichard C. Body composition interpretation. Contributions of the fat-free mass index
and the body fat mass index. Nutrition 2003;19(7-8):597-604. [doi: 10.1016/s0899-9007(03)00061-3] [Medline: 12831945]
46. Kim EK, Kwak SH, Jung HS, Koo BK, Moon MK, Lim S, et al. The effect of a smartphone-based, patient-centered diabetes
care system in patients with type 2 diabetes: A randomized, controlled trial for 24 weeks. Diabetes Care 2019 Jan;42(1):3-9.
[doi: 10.2337/dc17-2197] [Medline: 30377185]
47. Fabricatore AN. Behavior therapy and cognitive-behavioral therapy of obesity: Is there a difference? J Am Diet Assoc 2007
Jan;107(1):92-99. [doi: 10.1016/j.jada.2006.10.005] [Medline: 17197276]
48. Alkhaldi G, Hamilton FL, Lau R, Webster R, Michie S, Murray E. The effectiveness of prompts to promote engagement
with digital interventions: A systematic review. J Med Internet Res 2016 Jan 08;18(1):e6 [FREE Full text] [doi:
10.2196/jmir.4790] [Medline: 26747176]
49. Byambasuren O, Sanders S, Beller E, Glasziou P. Prescribable mHealth apps identified from an overview of systematic
reviews. NPJ Digit Med 2018;1:12 [FREE Full text] [doi: 10.1038/s41746-018-0021-9] [Medline: 31304297]
50. Jeong EK, Kim KN. Influence of stress on snack consumption in middle school girls. Nutr Res Pract 2007;1(4):349-355
[FREE Full text] [doi: 10.4162/nrp.2007.1.4.349] [Medline: 20368961]
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 17http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
51. Teixeira PJ, Going SB, Sardinha LB, Lohman TG. A review of psychosocial pre-treatment predictors of weight control.
Obes Rev 2005 Feb;6(1):43-65. [doi: 10.1111/j.1467-789X.2005.00166.x] [Medline: 15655038]
52. Teixeira PJ, Going SB, Houtkooper LB, Cussler EC, Martin CJ, Metcalfe LL, et al. Weight loss readiness in middle-aged
women: Psychosocial predictors of success for behavioral weight reduction. J Behav Med 2002 Dec;25(6):499-523. [doi:
10.1023/a:1020687832448] [Medline: 12462956]
53. Teixeira PJ, Going SB, Houtkooper LB, Cussler EC, Metcalfe LL, Blew RM, et al. Pretreatment predictors of attrition and
successful weight management in women. Int J Obes Relat Metab Disord 2004 Sep;28(9):1124-1133. [doi:
10.1038/sj.ijo.0802727] [Medline: 15263921]
54. Teixeira V, Voci SM, Mendes-Netto RS, da Silva DG. The relative validity of a food record using the smartphone application
MyFitnessPal. Nutr Diet 2018 Apr;75(2):219-225. [doi: 10.1111/1747-0080.12401] [Medline: 29280547]
55. Beasley J, Riley WT, Jean-Mary J. Accuracy of a PDA-based dietary assessment program. Nutrition 2005 Jun;21(6):672-677.
[doi: 10.1016/j.nut.2004.11.006] [Medline: 15925290]
Abbreviations
ATQ-30: Automatic Thoughts Questionnaire
AUC: area under the curve
BSQ: Body Shape Questionnaire
BSQ-8C: Body Shape Questionnaire-8C
CBT: cognitive behavioral therapy
DEBQ: Dutch Eating Behavior Questionnaire
DEBQ-EM: Dutch Eating Behavior Questionnaire emotional eating scale
DEBQ-EX: Dutch Eating Behavior Questionnaire external eating scale
DEBQ-RE: Dutch Eating Behavior Questionnaire restrained eating scale
eHealth: electronic health
HOMA-IR: Homeostatic Model for Assessment of Insulin Resistance
K-BDI-II: Korean version of the Beck Depression Inventory-II
mHealth: mobile health
NRF: National Research Foundation of Korea
RCT: randomized controlled trial
ROC: receiver operating characteristic
RSES: Rosenberg Self-Esteem Scale
SIMS: Situational Motivation Scale
SNP: single nucleotide polymorphism
SNU: Seoul National University
TAI: Trait Anxiety Inventory
Edited by G Eysenbach; submitted 25.05.19; peer-reviewed by S Smith, J Chen, A Pfammatter, M Gaman; comments to author 30.09.19;
revised version received 13.10.19; accepted 09.02.20; published 30.04.20
Please cite as:
Kim M, Kim Y, Go Y, Lee S, Na M, Lee Y, Choi S, Choi HJ
Multidimensional Cognitive Behavioral Therapy for Obesity Applied by Psychologists Using a Digital Platform: Open-Label Randomized
Controlled Trial
JMIR Mhealth Uhealth 2020;8(4):e14817
URL: http://mhealth.jmir.org/2020/4/e14817/
doi: 10.2196/14817
PMID: 32352391
©Meelim Kim, Youngin Kim, Yoonjeong Go, Seokoh Lee, Myeongjin Na, Younghee Lee, Sungwon Choi, Hyung Jin Choi.
Originally published in JMIR mHealth and uHealth (http://mhealth.jmir.org), 30.04.2020. This is an open-access article distributed
under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/), which permits
unrestricted use, distribution, and reproduction in any medium, provided the original work, first published in JMIR mHealth and
uHealth, is properly cited. The complete bibliographic information, a link to the original publication on http://mhealth.jmir.org/,
as well as this copyright and license information must be included.
JMIR Mhealth Uhealth 2020 | vol. 8 | iss. 4 | e14817 | p. 18http://mhealth.jmir.org/2020/4/e14817/ (page number not for citation purposes)
Kim et alJMIR MHEALTH AND UHEALTH
XSL
FO
RenderX
... The included studies represent 2478 participants with a mean age of 39.1 years and a mean BMI of 31.8 kg/m 2 . Most of the included articles were from the United States (10/14, 71.4%) [20][21][22][23][24][25][26][27][28][29], and one each from Australia (1/14, 7.1%) [30], Belgium (1/14, 7.1%) [31], Korea (1/14, 7.1%) [32], and Japan (1/14, 7.1%) [33]. Half of the articles (7/14, 50%) reported the use of theoretical frameworks including sociocognitive theory [21,25,28], control theory [25,30], transtheoretical model [30,33], learning theory [23], operant conditioning, ecological theory, social network theory [25], cognitive behavioral therapy [32], and self-efficacy theory [28]. ...
... Most of the included articles were from the United States (10/14, 71.4%) [20][21][22][23][24][25][26][27][28][29], and one each from Australia (1/14, 7.1%) [30], Belgium (1/14, 7.1%) [31], Korea (1/14, 7.1%) [32], and Japan (1/14, 7.1%) [33]. Half of the articles (7/14, 50%) reported the use of theoretical frameworks including sociocognitive theory [21,25,28], control theory [25,30], transtheoretical model [30,33], learning theory [23], operant conditioning, ecological theory, social network theory [25], cognitive behavioral therapy [32], and self-efficacy theory [28]. The attrition rate ranged from 4.8% [28] to 36.8% [21], and the follow-up time points ranged from 3 [21,28] to 24 months [25]. ...
... articles [26,27,33] [20,23,29], health coaches (2/14, 14.3%) [25,28], physical activity coaches or physiologists (2/14, 14.3%) [27,31], nutritionists [21], physicians [33], pharmacists [33], and nurses [33]. Five (35.7%) articles mentioned some form of certification in nutrition, fitness, and lifestyle coaching [20,29,[31][32][33]. Seven studies [20,21,24,26,31,32] reported control groups receiving apps, whereas 8 studies [22,23,25,27,[29][30][31]33] reported control groups receiving usual care (without app or health coaching). ...
Article
Full-text available
Background Self-monitoring smartphone apps and health coaching have both individually been shown to improve weight-related outcomes, but their combined effects remain unclear. Objective This study aims to examine the effectiveness of combining self-monitoring apps with health coaching on anthropometric, cardiometabolic, and lifestyle outcomes in people with overweight and obesity. Methods Relevant articles published from inception till June 9, 2022, were searched through 8 databases (Embase, CINAHL, PubMed, PsycINFO, Scopus, The Cochrane Library, and Web of Science). Effect sizes were pooled using random-effects models. Behavioral strategies used were coded using the behavior change techniques taxonomy V1. Results A total of 14 articles were included, representing 2478 participants with a mean age of 39.1 years and a BMI of 31.8 kg/m2. Using combined intervention significantly improved weight loss by 2.15 kg (95% CI −3.17 kg to −1.12 kg; P<.001; I2=60.3%), waist circumference by 2.48 cm (95% CI −3.51 cm to −1.44 cm; P<.001; I2=29%), triglyceride by 0.22 mg/dL (95% CI −0.33 mg/dL to 0.11 mg/dL; P=.008; I2=0%), glycated hemoglobin by 0.12% (95% CI −0.21 to −0.02; P=.03; I2=0%), and total calorie consumption per day by 128.30 kcal (95% CI −182.67 kcal to −73.94 kcal; P=.003; I2=0%) kcal, but not BMI, blood pressure, body fat percentage, cholesterol, and physical activity. Combined interventional effectiveness was superior to receiving usual care and apps for waist circumference but only superior to usual care for weight loss. Conclusions Combined intervention could improve weight-related outcomes, but more research is needed to examine its added benefits to using an app. Trial Registration PROSPERO CRD42022345133; https://tinyurl.com/2zxfdpay
... The included studies were published from 2010 to 2021, representing a total of 1203 participants with mean ages ranging from 21.8 to 57.3 years old (Afari et al., 2019;Cesa et al., 2013;Czepczor-Bernat et al., 2020;Forman et al., 2013;Gade et al., 2013;Goldbacher et al., 2016;Hjelmesaeth et al., 2019;Jarvela-Reijonen et al., 2018;Kim et al., 2020;Kristeller et al., 2014;Kullgren et al., 2013;Manzoni et al., 2016;Mason et al., 2019;Meekums et al., 2012;Nourizadeh et al., 2020;Nurkkala et al., 2015;Paul et al., 2021;Simos et al., 2019;Stapleton et al., 2016Stapleton et al., , 2020Teixeira et al., 2010;Weineland et al., 2012;Yancy et al., 2019;Supp. 3). ...
... Intervention type CBT (Cesa et al., 2013;Gade et al., 2013;Goldbacher et al., 2016;Hjelmesaeth et al., 2019;Kim et al., 2020;Kristeller et al., 2014;Manzoni et al., 2016;Paul et al., 2021;Stapleton et al., 2016Stapleton et al., , 2020 12 CBT + mindfulness ( Table 2. ...
... Individual + group (Cesa et al., 2013;Czepczor-Bernat et al., 2020;Forman et al., 2013;Hjelmesaeth et al., 2019;Jarvela-Reijonen et al., 2018;Kim et al., 2020;Kristeller et al., 2014;Kullgren et al., 2013;Manzoni et al., 2016;Mason et al., 2019;Nurkkala et al., 2015;Paul et al., 2021) 17 ...
Article
Full-text available
Objectives: To evaluate the effectiveness of weight-loss interventions on emotional eating among adults with high body mass index (BMI). Methods: A systematic review, meta-analysis and meta-regression were performed on randomized controlled trials published from inception until 19 March 2021. Results: Thirty-one studies were included, representing 1203 participants with mean ages ranging from 21.8 to 57.3 years old and BMI 27.2-43.5 kg/m2 . We found small-to-medium interventional effects on emotional eating (n = 18; Hedges' g = 0.22; p = 0.01, I2 = 61.7%), uncontrolled eating (n = 16; Hedges' g = 0.46; p < 0.001, I2 = 71.6%) and cognitive restraint (n = 18; Hedges' g = 0.42; p < 0.001, I2 = 75.8%). Small-to-medium interventional effects were only found for emotional eating (n = 8; Hedges' g = 0.45; p = 0.02, I2 = 74.3%) 3-month post-intervention, and on BMI (n = 4; Hedges' g = 0.43; p < 0.05, I2 = 33.4%) and weight (n = 6; Hedges' g = 0.36; p < 0.01, I2 < 10.4%) 12-month post-intervention. Age, male proportion, baseline BMI, attrition rate and intervention length were not significant moderators of the heterogeneity between studies. Conclusion: Interventions improved emotional eating and weight loss along a year-long trajectory.
... Noom Weight has previously been shown to be an effective treatment for obesity, frequently producing weight loss exceeding 5% of initial body weight [23,27,34,35] in as little as 8 weeks [36] and persisting for up to 52 weeks [25]. However, Noom Weight's impact and the impact of mHealth technologies generally on HRU and costs among users with overweight or obesity compared with a demographically similar control group have not been previously reported. ...
Article
Full-text available
Background The Noom Weight program is a smartphone-based weight management program that uses cognitive behavioral therapy techniques to motivate users to achieve weight loss through a comprehensive lifestyle intervention. Objective This retrospective database analysis aimed to evaluate the impact of Noom Weight use on health care resource utilization (HRU) and health care costs among individuals with overweight and obesity. Methods Electronic health record data, insurance claims data, and Noom Weight program data were used to conduct the analysis. The study included 43,047 Noom Weight users and 14,555 non–Noom Weight users aged between 18 and 80 years with a BMI of ≥25 kg/m² and residing in the United States. The index date was defined as the first day of a 3-month treatment window during which Noom Weight was used at least once per week on average. Inverse probability treatment weighting was used to balance sociodemographic covariates between the 2 cohorts. HRU and costs for inpatient visits, outpatient visits, telehealth visits, surgeries, and prescriptions were analyzed. ResultsWithin 12 months after the index date, Noom Weight users had less inpatient costs (mean difference [MD] −US $20.10, 95% CI −US $30.08 to −US $10.12), less outpatient costs (MD −US $124.33, 95% CI −US $159.76 to −US $88.89), less overall prescription costs (MD −US $313.82, 95% CI −US $565.42 to −US $62.21), and less overall health care costs (MD −US $450.39, 95% CI −US $706.28 to −US $194.50) per user than non–Noom Weight users. In terms of HRU, Noom Weight users had fewer inpatient visits (MD −0.03, 95% CI −0.04 to −0.03), fewer outpatient visits (MD −0.78, 95% CI −0.93 to −0.62), fewer surgeries (MD −0.01, 95% CI −0.01 to 0.00), and fewer prescriptions (MD −1.39, 95% CI −1.76 to −1.03) per user than non–Noom Weight users. Among a subset of individuals with 24-month follow-up data, Noom Weight users incurred lower overall prescription costs (MD −US $1139.52, 95% CI −US $1972.21 to −US $306.83) and lower overall health care costs (MD −US $1219.06, 95% CI −US $2061.56 to −US $376.55) per user than non–Noom Weight users. The key differences were associated with reduced prescription use. Conclusions Noom Weight use is associated with lower HRU and costs than non–Noom Weight use, with potential cost savings of up to US $1219.06 per user at 24 months after the index date. These findings suggest that Noom Weight could be a cost-effective weight management program for individuals with overweight and obesity. This study provides valuable evidence for health care providers and payers in evaluating the potential benefits of digital weight loss interventions such as Noom Weight.
... This reiterates the need for a holistic approach to e-mental health interventions that is not limited to weight loss. In addition, the results of a recent study demonstrated that participants in a digital CBT study lost significantly more weight, indicating the importance of a multidimensional approach [68]. However, to date, we are not aware of any study that has developed and examined an e-mental health intervention with the main goal of reducing mental distress such as depressive symptomatology based on a user-centred design approach. ...
Article
Full-text available
Introduction: Several studies indicate an association between mental disorders and overweight or obesity. E-mental-health interventions offer an effective way to overcome barriers to health care access for individuals with overweight and obesity. The objective of this study was to examine the needs and demands for e-mental health interventions among individuals with overweight and obesity. Methods: A cross-sectional study was conducted from 2020 to 2021 in Germany. A total of 643 participants were recruited through specialized social media platforms and the Alfried-Krupp hospital in Essen, Germany. Sociodemographic and medical data were analyzed, as well as data on depressive symptoms and on the needs and demands for e-mental health interventions. Results: Contact with and recommendation by experts appear to be key aspects in the acceptance and use of e-mental health interventions. In summary, most participants preferred a 20- to 30-minute weekly session via smartphone over a four-month period. The highest preference in terms of features included practicing coping skills and being provided with information; in regard to desired topics, nutrition consultation, quality of life and adapting to new life situations were considered most important. Discussion: E-mental health interventions can be highly beneficial for individuals, especially when developed through a user-centered design approach. The results of the study indicate which content and design are preferred and, thereby, provide valuable information for consideration when developing a tailored e-mental health intervention.
... between-person randomized controlled trials (RCTs) 75,76 including remote RCTs 77,78 , cluster RCTs 79,80 , or stepped wedge trial. 73,74 If SMART benchmarks are not met or there was not a clinically significant difference observed between the DTx and comparator, then returning to Phase I or Phase II activities is appropriate. ...
Preprint
Full-text available
Digital Therapeutics (DTx) are increasingly seen as a promising way to provide safe, effective, accessible, sustainable, scalable, and equitable approaches to advance individual and population health. Developing DTx is inherently complex in that DTx may include multiple interacting components, such as tools to support activities like medication adherence, health behavior goal-setting or self-monitoring, and algorithms that adapt the provision of these according to individual needs. As recommended in a recent worldwide review of regulatory approaches for DTx, a way to address this complexity is to base the design, development, testing, and iterative improvements of DTx on real-world evidence (RWE). Notably, that review highlights the need for improved guidance on when and how to use RWE for DTx. In this paper, we propose the DTx RWE Framework to provide a pragmatic, iterative, milestone-driven approach for producing RWE for DTx regulation. The DTx RWE Framework is based on best practices from human-centered design, optimization trials for behavioral interventions, behavioral health research, and implementation science. It maps these onto the traditional four phase development model for pharmaceuticals but includes key adaptations relevant to RWE production for DTx, such as including early and ongoing partnerships between developers of DTx and their intended users in clinical and/or community settings. Adoption of our DTx RWE Framework may help address known problems with current DTx, including questionable marketing claims, lack of evidence-based guidance for patients, providers, and health care leaders and the need to improve economic incentives that advance the adoption and use of DTx.
... The main purpose of e-government is to facilitate government-to-citizen engagement by providing citizens with efficient, all-encompassing access. This can be accomplished by including citizens in the political process and through citizen involvement as a form of collaboration to promote democratic values [28,29]. E-participation refers to e-government based on citizen involvement and participation. ...
Article
Full-text available
This study aims to assess the impact of public value on e-government ambidexterity. Furthermore, current technology variables, public value variables, and e-government ambidexterity are incorporated into a framework for measuring e-government service performance. The partial least square structural equation modeling (SEM-PLS) approach was used in this study as the quantitative method. Four hundred twenty-five final data points were used as a sample, and all seven proposed hypotheses are accepted, have positive values, and have a significant impact. This study can be used as a guide for e-government organizers at the national and regional levels, as well as a resource for other studies examining e-government ambidexterity.
... In Korea, when patients were treated for obesity using a mobile app, the average weight loss was -2.73 kg, 27 and a combined approach using both an ICT-based intervention and conventional cognitive behavioral therapy showed a more effective weight loss effect than the existing face-toface treatment (-3.4% vs. -0.7%). 28 Overseas, an intensive contact web-based program showed a remarkable effect on weight loss (mean, -4.31 kg; 95% CI, -5.22 to -3.41). 29 These results suggest that ICT-based interventions can be most effective when provided multi-dimensionally and accompanied by therapist feedback and support. ...
Article
Full-text available
The rapid increase in the obese population is a problem indicates the need for measures to prevent and treat obesity. Since the early 2000s, worldwide digital health has focused on obesity management. Information and communication technology (ICT)-based obesity intervention can be an efficient method for treating obesity/metabolic syndrome, has no time limitations, and is an inexpensive and easily accessible treatment modality for both physicians and patients. Previous studies have confirmed the effects of ICT-based interventions for obesity/metabolic syndrome management for behavioral improvement in lifestyle modification. In addition, ICT-based interventions in obese/metabolic syndrome patients are recommended as part of a comprehensive strategy for weight loss and maintenance. The Committee of IT-convergence Treatment of Metabolic Syndrome was established by the Korean Society for the Study of Obesity in 2021, and had been gathering theoretical and clinical evidence in digital therapeutics fields and developing new methods for managing obesity/metabolic syndrome. As part of this effort, if the "obesity management prototype" is commercialized, it will be available for convenient treatment of individuals with obesity/metabolic syndrome.
Article
Full-text available
Introduction Internet-based and mobile-based interventions (IMIs) provide innovative low-threshold and cost-effective prevention and self-management options for mental health problems complementary to standard treatment. The objective of this systematic review is to summarise the effectiveness and to critically evaluate studies on IMIs addressing comorbid depressive symptoms in adults with overweight or obesity. Methods and analysis The study authors will systematically search the databases MEDLINE, Cochrane Library, PsycINFO, Web of Science, Embase and Google Scholar (for grey literature) for randomised controlled trials (RCTs) of IMIs for individuals with overweight or obesity and comorbid depressive symptoms without restrictions on publication date (planned inception 1 June 2023 to 1 December 2023). Two reviewers will independently extract and evaluate data from studies eligible for inclusion by assessing quality of evidence and qualitatively synthesising results. Preferred Reporting Items for Systematic reviews Meta-Analyses (PRISMA) standards and the revised Cochrane Risk of Bias tool in RCTs (RoB 2) will be applied. Ethics and dissemination Ethical approval is not required as no primary data will be collected. Study results will be disseminated through publication in a peer-reviewed journal and presentations on conferences. PROSPERO registration number CRD42023361771.
Article
Full-text available
The goal of the 8th edition of the Clinical Practice Guidelines for Obesity is to help primary care physician provide safe, effective care to patients with obesity by offering evidence-based recommendations to improve the quality of treatment. The Committee for Clinical Practice Guidelines comprised individuals with multidisciplinary expertise in obesity management. A steering board of seven experts oversaw the entire project. Recommendations were developed based on answers to key questions formulated in patient/problem, intervention, comparison, outcomes (PICO) format. Guidelines underwent multi-level review and cross-checking and received endorsement from relevant scientific societies. This edition of the guidelines includes criteria for diagnosing obesity, abdominal obesity, and metabolic syndrome; evaluation of obesity and its complications; weight loss goals; and treatment options such as diet, exercise, behavioral therapy, pharmacotherapy, and bariatric/metabolic surgery for Korean people with obesity. Compared to previous editions of the guidelines, the current edition includes five new topics: diagnosis of obesity, obesity in women, obesity in patients with mental illness, weight maintenance after weight loss, and the use of information and communication technology-based interventions for obesity treatment. This edition of the guidelines features has improved organization, more clearly linking key questions in PICO format to recommendations and key references. We are confident that these new Clinical Practice Guidelines for Obesity will be a valuable resource for all healthcare professionals as they describe the most current and evidence-based treatment options for obesity in a well-organized format.
Article
Full-text available
Background The rising incidence of chronic diseases among the population, further exacerbated by the phenomenon of aging, is a primary concern and a serious challenge for the healthcare systems worldwide. Among the wide realm of health digital technologies, the rise of Digital Therapeutics (DTx), which are medical devices able to deliver evidence-based treatments to manage and treat diseases, opens new opportunities. However, their diffusion and usage are still fragmented among countries. As the diffusion results from the adoption of technology from a social system and individual acceptance, this study aims to design and test a theoretical model that investigates the intention to use DTx, with a particular focus on the treatment of obesity, as a widespread and burdensome chronic condition. Methods This research is built on 336 answers coming from a survey to test the proposed model, which consists of a combination of organizational mechanisms, derived from Institutional Theory, and rational factors, derived from the Technology Acceptance Model (TAM). The survey has been delivered to patients and former patients of Istituto Auxologico Italiano, a hospital with several locations in northern Italy, recognized as a center of excellence for the treatment of obesity. Results The analyses of the answers, performed through the Structural Equation Modelling (SEM) technique, confirmed the influence of the Perceived Usefulness on Intention To Use, and of the Perceived Ease Of Use on the Perceived Usefulness, confirming the validity of the assumptions derived from the TAM. On the other hand, institutional factors were introduced as antecedents of the Perceived Usefulness, and the Perceived Ease Of Use. Results show that the Regulative Pillar influences both the TAM constructs, the Normative Pillar (peer influence) has a positive effect only on the Perceived Usefulness, and finally, the Cultural Pillar impacts the Perceived Ease Of Use. Conclusion This study allows filling the knowledge gap regarding the usage of the Institutional as a means to predict individuals’ intentions. Moreover, managerial contributions are available as the results have been operationalized into practical advice to managers and healthcare professionals to foster the adoption, and thus the diffusion, of Digital Therapeutics.
Article
Full-text available
OBJECTIVE This study evaluated the efficacy of a smartphone-based, patient-centered diabetes care system (mDiabetes) for type 2 diabetes that contains comprehensive modules for glucose monitoring, diet, physical activity, and a clinical decision support system. RESEARCH DESIGN AND METHODS We conducted a 24-week, multicenter, randomized controlled trial with adult patients with inadequately controlled type 2 diabetes. The patients were randomly assigned to the mDiabetes group or the paper logbook (pLogbook) group. The primary end point was the difference of the change in HbA1c from baseline between the two groups. RESULTS HbA1c reduction from baseline was greater in the mDiabetes group (−0.40 ± 0.09%, n = 90) than in the pLogbook group (−0.06 ± 0.10%, n = 82). The difference of adjusted mean changes was 0.35% (95% CI 0.14–0.55, P = 0.001). The proportion of patients whose HbA1c fell below 7.0% (53 mmol/mol) was 41.1% for the mDiabetes group and 20.7% for the pLogbook group (odds ratio [OR] 2.01, 95% CI 1.24–3.25, P = 0.003). The percentage of patients who attained HbA1c levels below 7.0% (53 mmol/mol) without hypoglycemia was 31.1% in the mDiabetes group and 17.1% in the pLogbook group (OR 1.82, 95% CI 1.03–3.21, P = 0.024). There was no difference in the event numbers of severe hyperglycemia and hypoglycemia between the two groups. CONCLUSIONS The implementation of the mDiabetes for patients with inadequately controlled type 2 diabetes resulted in a significant reduction in HbA1c levels, with tolerable safety profiles.
Article
Full-text available
Mobile health apps aimed towards patients are an emerging field of mHealth. Their potential for improving self-management of chronic conditions is significant. Here, we propose a concept of “prescribable” mHealth apps, defined as apps that are currently available, proven effective, and preferably stand-alone, i.e., that do not require dedicated central servers and continuous monitoring by medical professionals. Our objectives were to conduct an overview of systematic reviews to identify such apps, assess the evidence of their effectiveness, and to determine the gaps and limitations in mHealth app research. We searched four databases from 2008 onwards and the Journal of Medical Internet Research for systematic reviews of randomized controlled trials (RCTs) of stand-alone health apps. We identified 6 systematic reviews including 23 RCTs evaluating 22 available apps that mostly addressed diabetes, mental health and obesity. Most trials were pilots with small sample size and of short duration. Risk of bias of the included reviews and trials was high. Eleven of the 23 trials showed a meaningful effect on health or surrogate outcomes attributable to apps. In conclusion, we identified only a small number of currently available stand-alone apps that have been evaluated in RCTs. The overall low quality of the evidence of effectiveness greatly limits the prescribability of health apps. mHealth apps need to be evaluated by more robust RCTs that report between-group differences before becoming prescribable. Systematic reviews should incorporate sensitivity analysis of trials with high risk of bias to better summarize the evidence, and should adhere to the relevant reporting guideline.
Article
Full-text available
Background: Prevalent co-occurring poor diet and physical inactivity convey chronic disease risk to the population. Large magnitude behavior change can improve behaviors to recommended levels, but multiple behavior change interventions produce small, poorly maintained effects. Objective: The Make Better Choices 2 trial tested whether a multicomponent intervention integrating mHealth, modest incentives, and remote coaching could sustainably improve diet and activity. Methods: Between 2012 and 2014, the 9-month randomized controlled trial enrolled 212 Chicago area adults with low fruit and vegetable and high saturated fat intakes, low moderate to vigorous physical activity (MVPA) and high sedentary leisure screen time. Participants were recruited by advertisements to an open-access website, screened, and randomly assigned to either of two active interventions targeting MVPA simultaneously with, or sequentially after other diet and activity targets (N=84 per intervention) or a stress and sleep contact control intervention (N=44). They used a smartphone app and accelerometer to track targeted behaviors and received personalized remote coaching from trained paraprofessionals. Perfect behavioral adherence was rewarded with an incentive of US $5 per week for 12 weeks. Diet and activity behaviors were measured at baseline, 3, 6, and 9 months; primary outcome was 9-month diet and activity composite improvement. Results: Both simultaneous and sequential interventions produced large, sustained improvements exceeding control (P<.001), and brought all diet and activity behaviors to guideline levels. At 9 months, the interventions increased fruits and vegetables by 6.5 servings per day (95% CI 6.1-6.8), increased MVPA by 24.7 minutes per day (95% CI 20.0-29.5), decreased sedentary leisure by 170.5 minutes per day (95% CI -183.5 to -157.5), and decreased saturated fat intake by 3.6% (95% CI -4.1 to -3.1). Retention through 9-month follow-up was 82.1%. Self-monitoring decreased from 96.3% of days at baseline to 72.3% at 3 months, 63.5% at 6 months, and 54.6% at 9 months (P<.001). Neither attrition nor decline in self-monitoring differed across intervention groups. Conclusions: Multicomponent mHealth diet and activity intervention involving connected coaching and modest initial performance incentives holds potential to reduce chronic disease risk. Trial registration: ClinicalTrials.gov NCT01249989; https://clinicaltrials.gov/ct2/show/NCT01249989 (Archived by WebCite at https://clinicaltrials.gov/ct2/show/NCT01249989).
Article
Full-text available
Background It is widely recognized that the prevalence of obesity and comorbidities including prediabetes and type 2 diabetes continue to increase worldwide. Results from a 24-week Diabetes Prevention Program (DPP) fully mobile pilot intervention were previously published showing promising evidence of the usefulness of DPP-based eHealth interventions on weight loss. Objective This pilot study extends previous findings to evaluate weight loss results of core (up to week 16) and maintenance (postcore weeks) DPP interventions at 65 weeks from baseline. Methods Originally, 140 participants were invited and 43 overweight or obese adult participants with a diagnosis of prediabetes signed up to receive a 24-week virtual DPP with human coaching through a mobile platform. At 65 weeks, this pilot study evaluates weight loss and engagement in maintenance participants by means of repeated measures analysis of variances and backward multiple linear regression to examine predictors of weight loss. Last observation carried forward was used for endpoint measurements. ResultsAt 65 weeks, mean weight loss was 6.15% in starters who read 1 or more lessons per week on 4 or more core weeks, 7.36% in completers who read 9 or more lessons per week on core weeks, and 8.98% in maintenance completers who did any action in postcore weeks (all P
Article
Full-text available
Introduction: Although bariatric surgery results in massive weight loss, weight regain over time up to as much as 25% is not uncommon. Weight regain in this population often leads to long-term weight loss failure and non-compliance in clinical follow-up and program recommendations. Methods: We analyzed early weight outcomes at 3 and 6 months of 48 bariatric patients referred to an individualized, multidisciplinary medical management program at the Center for Obesity Medicine (COM) to address weight regain in 2015 and compared to a group of matched non-bariatric patients. The medical management center, under the direction of a medical obesity specialist and complementary to the surgical program and multidisciplinary team, addressed weight regain with intensive lifestyle (diet, activity, anti-stress therapy, behavioral counseling, sleep) and with medical intervention (one or more anti-obesity medications). Results: According to early findings, the average percentage post-operative weight regain of patients entering the weight management program was 20% above nadir and time since surgery averaged 6 years (range = 1 to 20 years) with a mean weight loss of - 2.3 kg after 3 months and - 4.4 kg at 6 months into the program. Individuals most successful with weight loss were those treated with anorexigenic pharmaceuticals. Weight and percent weight loss were significantly greater for the non-surgical than the surgical patients at 3 and 6 months (p < 0.05). Conclusions: A medically supervised weight management program complementary to surgery is beneficial for the treatment of weight regain and may prove important in assisting the surgical patient achieve long-term weight loss success.
Article
Full-text available
Aim: The present study aimed to validate food records on the application MyFitnessPal (MFP), comparing them with paper-based food records (P-FR). Methods: Thirty university students, including males and females, volunteered and recorded dietary intakes on P-FR and MFP food records (MFP-FR). The values of energy, macronutrients and fibre from MFP-FR were compared with data from P-FR, calculated using Brazilian food composition tables. Adjustments for in-person variability and energy intake were performed, and comparisons were made between each data set, using the Wilcoxon signed-rank test, Spearman's correlation and Bland-Altman agreement plots. Results: Positive moderate correlations between P-FR and MFP-FR for all variables, and non-significant associations for energy and fibre were found. The Bland-Altman plots showed tendency to underestimation and relatively narrow limits of agreement. Carbohydrate and lipids show trends of increasing the degree of overestimation with increased intake, even after data normalisation. Conclusions: MFP tends to underestimate ingestion of nutrients probably due to inadequacies in the MFP database. However, MFP showed good relative validity, especially for energy and fibre. Its use, as well as other similar applications, should be encouraged, due to ease of assessing dietary information, although careful usage is recommended because of database gaps.
Article
Full-text available
Background: There are inconsistent results for the effectiveness of using smartphone applications (apps) or websites on weight loss. We investigated the efficacy of a smartphone intervention using a designated app that utilizes a lifestyle intervention-focused approach, including a human coaching element, toward weight loss in overweight or obese Korean adults. Methods: One hundred four adults aged 20-60 years with a body mass index ≥23 kg/m(2), who signed up for a smartphone program for weight loss (using the Noom app), were recruited. Participants received an in-person orientation about the study and app use, and a baseline blood sample was obtained. The in-app intervention with daily behavior and nutrition education content and coaching lasted 15 weeks. The primary endpoint of the study was a change in weight. The secondary endpoints were changes in metabolic risk factors such as blood pressure, waist circumference, and glucose and lipid profiles. Body composition changes were also assessed, and body weight at 52 weeks was measured to ascertain long-term effects. Results: Participants showed a clinically significant weight loss effect of -7.5% at the end of the 15-week program (P < 0.001), and at a 52-week follow-up, a weight loss effect of -5.2% was maintained. At 15 weeks, percent body fat and visceral fat decreased by -6.0 ± 5.4% and -3.4 ± 2.7 kg, respectively (both P < 0.001). Fasting glucose level also decreased significantly by -5.7 ± 14.6 mg/dL at 15 weeks. Lipid parameters showed significant improvements, except for high-density lipoprotein cholesterol. The frequency of logging meals and exercise was associated with body fat loss. Conclusions: This advanced smartphone app was a useful tool to maintain weight loss in overweight or obese people.
Article
Importance Dietary modification remains key to successful weight loss. Yet, no one dietary strategy is consistently superior to others for the general population. Previous research suggests genotype or insulin-glucose dynamics may modify the effects of diets. Objective To determine the effect of a healthy low-fat (HLF) diet vs a healthy low-carbohydrate (HLC) diet on weight change and if genotype pattern or insulin secretion are related to the dietary effects on weight loss. Design, Setting, and Participants The Diet Intervention Examining The Factors Interacting with Treatment Success (DIETFITS) randomized clinical trial included 609 adults aged 18 to 50 years without diabetes with a body mass index between 28 and 40. The trial enrollment was from January 29, 2013, through April 14, 2015; the date of final follow-up was May 16, 2016. Participants were randomized to the 12-month HLF or HLC diet. The study also tested whether 3 single-nucleotide polymorphism multilocus genotype responsiveness patterns or insulin secretion (INS-30; blood concentration of insulin 30 minutes after a glucose challenge) were associated with weight loss. Interventions Health educators delivered the behavior modification intervention to HLF (n = 305) and HLC (n = 304) participants via 22 diet-specific small group sessions administered over 12 months. The sessions focused on ways to achieve the lowest fat or carbohydrate intake that could be maintained long-term and emphasized diet quality. Main Outcomes and Measures Primary outcome was 12-month weight change and determination of whether there were significant interactions among diet type and genotype pattern, diet and insulin secretion, and diet and weight loss. Results Among 609 participants randomized (mean age, 40 [SD, 7] years; 57% women; mean body mass index, 33 [SD, 3]; 244 [40%] had a low-fat genotype; 180 [30%] had a low-carbohydrate genotype; mean baseline INS-30, 93 μIU/mL), 481 (79%) completed the trial. In the HLF vs HLC diets, respectively, the mean 12-month macronutrient distributions were 48% vs 30% for carbohydrates, 29% vs 45% for fat, and 21% vs 23% for protein. Weight change at 12 months was −5.3 kg for the HLF diet vs −6.0 kg for the HLC diet (mean between-group difference, 0.7 kg [95% CI, −0.2 to 1.6 kg]). There was no significant diet-genotype pattern interaction (P = .20) or diet-insulin secretion (INS-30) interaction (P = .47) with 12-month weight loss. There were 18 adverse events or serious adverse events that were evenly distributed across the 2 diet groups. Conclusions and Relevance In this 12-month weight loss diet study, there was no significant difference in weight change between a healthy low-fat diet vs a healthy low-carbohydrate diet, and neither genotype pattern nor baseline insulin secretion was associated with the dietary effects on weight loss. In the context of these 2 common weight loss diet approaches, neither of the 2 hypothesized predisposing factors was helpful in identifying which diet was better for whom. Trial Registration clinicaltrials.gov Identifier: NCT01826591
Article
The alarming rise in the worldwide prevalence of obesity is paralleled by an increasing burden of type 2 diabetes mellitus. Metabolic surgery is the most effective means of obtaining substantial and durable weight loss in individuals with obesity. Randomized trials have recently shown the superiority of surgery over medical treatment alone in achieving improved glycemic control, as well as a reduction in cardiovascular risk factors. The mechanisms seem to extend beyond the magnitude of weight loss alone and include improvements in incretin profiles, insulin secretion, and insulin sensitivity. Moreover, observational data suggest that the reduction in cardiovascular risk factors translates to better patient outcomes. This review describes commonly used metabolic surgical procedures and their current indications and summarizes the evidence related to weight loss and glycemic outcomes. It further examines their potential effects on cardiovascular outcomes and mortality and discusses future perspectives.
Article
Background: Sugar-sweetened beverages and maternal weight are strong drivers of child obesity, but few studies have targeted these risk factors as an obesity prevention strategy in children. Objective: The objective of this study was to test the efficacy of a smartphone-delivered intervention to reduce parent-provided sugar-sweetened beverage and juice (SSB/juice) consumption among children ages 3-5 and maternal weight. Methods: Mothers with overweight or obesity, who had a child ages 3-5 that consumed at least 12 fl. oz./day of SSB/juice (N = 51 dyads) were randomized to the Smart Moms group that received one group session, lessons on a mobile website, and text messages, or to a waitlist control group. Mothers self-monitored their children's beverages in addition to their own beverages, high-calorie foods, and weight. Assessments at baseline, 3, and 6 months included dietary recalls to measure SSB/juice intake and objectively measured maternal weight. Results: Using linear mixed models controlling for baseline values, child age and race, there was a greater reduction in child SSB/juice in Smart Moms compared with control at 6 months (-9.7 oz./day vs. 1.7 oz./day, p < .01). Mothers in Smart Moms lost 2.4 kg at 6 months compared with a 0.9-kg gain in the control group (p < .01). Conclusions: An intervention delivered using mHealth technologies can target mothers to change child dietary behaviours and improve maternal weight, which suggests a novel approach to family-based obesity prevention.