v
Search
Advanced Search

Publications > Journals > Journal of Clinical and Translational Hepatology > Article Full Text

  • OPEN ACCESS

Efficacy of Intragastric Balloons in the Markers of Metabolic Dysfunction-associated Fatty Liver Disease: Results from Meta-analyses

  • Zi-Yuan Zou# ,
  • Jing Zeng#,
  • Tian-Yi Ren,
  • Yi-Wen Shi,
  • Rui-Xu Yang and
  • Jian-Gao Fan* 
 Author information
Journal of Clinical and Translational Hepatology   2021;9(3):353-363

doi: 10.14218/JCTH.2020.00183

Abstract

Background and Aims

Nonalcoholic fatty liver disease, now renamed metabolic dysfunction-associated fatty liver disease (MAFLD), is common in obese patients. Intragastric balloon (IGB), an obesity management tool with low complication risk, might be used in MAFLD treatment but there is still unexplained heterogeneity in results across studies.

Methods

We conducted a systematic search of 152 citations published up to September 2020. Meta-analyses, stratified analyses, and meta-regression were performed to evaluate the efficacy of IGB on homeostasis model assessment of insulin resistance (HOMA-IR), alanine aminotransferase (ALT), aspartate aminotransferase (AST), and gamma-glutamyl transpeptidase (GGT), and to identify patients most appropriate for IGB therapy.

Results

Thirteen observational studies and one randomized controlled trial met the inclusion criteria (624 participants in total). In the overall estimate, IGB therapy significantly improved the serum markers change from baseline to follow-up [HOMA-IR: 1.56, 95% confidence interval (CI)=1.16–1.95; ALT: 11.53 U/L, 95% CI=7.10–15.96; AST: 6.79 U/L, 95% CI=1.69–11.90; GGT: 10.54 U/L, 95% CI=6.32–14.75]. In the stratified analysis, there were trends among participants with advanced age having less change in HOMA-IR (1.07 vs. 1.82). The improvement of insulin resistance and liver biochemistries with swallowable IGB therapy was no worse than that with endoscopic IGB. Multivariate meta-regression analyses showed that greater HOMA-IR loss was predicted by younger age (p=0.0107). Furthermore, effectiveness on ALT and GGT was predicted by basal ALT (p=0.0004) and GGT (p=0.0026), respectively.

Conclusions

IGB is effective among the serum markers of MAFLD. Younger patients had a greater decrease of HOMA-IR after IGB therapy.

Keywords

Intragastric balloon, Nonalcoholic fatty liver disease, Insulin resistance, Age groups, Treatment outcome

Introduction

As the prevalence of obesity and insulin resistance continues to rise, nonalcoholic fatty liver disease (NAFLD), now rebranded as metabolic dysfunction-associated fatty liver disease (MAFLD), has emerged as the most prevalent parenchymal liver disease worldwide and explains 9% of deaths from liver cirrhosis.1–3 Currently, there are no approved pharmacotherapies for fatty liver disease.4 Bariatric surgery for fatty liver disease has enjoyed a high profile due to its remarkable capacity for improving liver enzyme, NAFLD activity score, and fibrosis.5,6 However, unexpected rates of liver fibrosis progression in patients who undergo bariatric surgery and excessive risks of postoperative complications limit the acceptance of bariatric surgery.7,8 Additionally, lifestyle modification strategies are difficult to address the disadvantage regarding treatment compliance.9,10 As a result, novel therapeutic applications, which take all efficacy, safety, and treatment compliance into account, are urgently needed for all MAFLD patients.

Recently, the potential role of endoscopic bariatric and metabolic therapies (EBMT) in the management of fatty liver disease has been highlighted.11,12 EBMT are developed to avoid the invasive nature of laparoscopic or open bariatric surgery, in contrast, reproducing similar gastrointestinal physiological alterations and therapeutic effects.13 Among these interventions, intragastric balloon (IGB), as a space-occupying EBMT device with proven efficacy in inducing weight loss, has been used in diminishing liver volume to reduce the risks of subsequent bariatric surgery and has met with success.14,15 Prior study has demonstrated that the change in liver volume was positively correlated with the change in intrahepatic fat,16 which suggested the potential therapeutic effect of using IGB in fatty liver disease. In terms of current evidence, a randomized controlled trial (RCT) evaluated changes in histological scores after 6-month IGB therapy and showed a beneficial effect on the severity of fatty liver disease.17 However, due to the limited sample size of this trial, we still need to combine the existing RCT findings with observational longitudinal studies to present the effectiveness of IGB in larger sample size, before it is widely recommended for the treatment of MAFLD. Therefore, we performed a systematic review with meta-analyses to evaluate the therapeutic effect of IGB on the markers of MAFLD, such as homeostasis model assessment of insulin resistance (HOMA-IR) index, alanine aminotransferase (ALT), aspartate aminotransferase (AST), and gamma-glutamyl transpeptidase (GGT). Furthermore, to identify patients most appropriate for IGB therapy, stratified analyses and meta-regression were both implemented.

Methods

Data sources and search strategy

This systematic review was performed according to the preferred reporting items for systematic reviews and meta-analysis statement (see Table S1).18 The protocol for this review is registered in PROSPERO (no. CRD42020214315).

To collect all full-text articles describing the effect of IGB on the markers of MAFLD, we performed a search of the Medline, Cochrane Library, and Web Of Science with English-language restriction and up to September 2020 using the following strategy: (“Intragastric balloon” OR “Gastric balloon”) AND (“Alanine aminotransferase” OR “Alanine transaminase” OR “ALT” OR “Liver” OR “Nonalcoholic fatty liver disease” OR “Non-alcoholic fatty liver disease” OR “NASH” OR “NAFLD” OR “HOMA-IR” OR “Homeostasis model assessment” OR “Insulin resistance”). The detailed search strategy is summarized in Table S2. Furthermore, the reference lists of each article were manually searched to prevent the omission of any pertinent study.

Study eligibility and selection criteria

Only observational longitudinal studies and RCTs were included. Inclusion criteria of the articles were as follows: (a) population: all patients who are obese or in need of obesity treatment; (b) intervention: liquid-filled IGB procedure; (c) comparator: the participants at baseline before IGB placement; and (d) outcome: the decrease of ALT, AST, GGT, or HOMA-IR index in all the participants treated with IGB. Moreover, the studies which recruited only pediatric patients or utilized the gas-filled IGB as an intervention were excluded to prevent bias.

Data extraction and quality assessment

Data extraction was performed independently by two investigators (ZYZ, JZ). The information and characteristics extracted from the included study were first author, year of publication, study design, country, study size of participants with IGB therapy, IGB type, dwelling time of IGB, filling of IGB, method of IGB implantation, additional nutrition and exercise prescription, description of liver disease in exclusion criteria, percentage of male individuals, prevalence of diabetes, participants’ age and body mass index (BMI) at baseline, and participants’ ALT, AST, GGT and HOMA-IR before and after IGB therapy. When standard deviation was unavailable, it was replaced with a quarter of the range.19 The risk of bias of the selected studies was evaluated using the modified Newcastle-Ottawa scale (NOS) for observational longitudinal studies20 and Cochrane Collaboration’s tool for RCT.21

Data analysis

Using R software version 3.6.3 (R Foundation for Statistical Computing, Vienna, Austria) and Review Manager version 5.3 (Nordic Cochrane Centre, The Cochrane Collaboration, Copenhagen, Denmark), meta-analyses (quantitative synthesis) were performed to evaluate the pooled mean difference (MD) in HOMA-IR, ALT, AST and GGT from baseline to end of IGB therapy using the inverse variance method and random-effect model, with 95% confidence interval (CI) and p-value. A p-value <0.05 was considered statistically significant. Publication bias was evaluated by Egger’s test and funnel plot.22,23 Heterogeneity was evaluated with inconsistency index (I2), classified as a low (I2≥25%), substantial (I2≥50%), or considerable (I2≥75%).24 Stratified analyses were conducted to investigate sources of heterogeneity based on the following characteristics: method of IGB implantation; mean basal level of serum markers (HOMA-IR, ALT, AST, or GGT); age and BMI of the participants; study region; and NOS score. When meta-regression analysis was performed, univariate and multivariate linear regression models were utilized to evaluate the slope coefficient between the reduced value of serum marker (HOMA-IR, ALT, AST, or GGT) after IGB therapy and the following covariates: mean basal level of serum marker; percentage of male individuals; and age and BMI of the participants. To summarize the results, the scatter plots were mapped to materialize the linear relationship between the changed value after IGB therapy and covariates which had statistical significance with both univariate and multivariate meta-regression analysis (p<0.05). Each study was represented by a circle of size proportional to the inverse of the variance of MD.

Results

Literature search results

Figure 1 summarizes the flow diagram of the selection process performed to identify eligible studies in this systematic review. Out of 152 references, a total of 14 studies25–38 comprising 624 participants met the predefined inclusion criteria. All studies were published prior to September 13, 2020.

Flow diagram of the study selection process.
Fig. 1  Flow diagram of the study selection process.

Improvement of insulin resistance after IGB on therapy

Summary of study characteristics

Eight studies25–27,29,33,35,36,38 with a total of 352 individuals were included in this meta-analysis of HOMA-IR level, and their characteristics are summarized in Table 1. All included studies were published after 2007. Of these, one38 was a two-arm RCT, and the rest25–27,29,33,35,36 were observational longitudinal studies, meaning that a total of nine intervention arms were included in this analysis. The participants came from three countries (Brazil, Italy, Japan). Seven intervention arms25–27,29,33,38 applied the Orbera IGB system, one arm36 used the Orbera/Spatz IGB system, and the single remaining arm35 reported results with the Elipse IGB system. Furthermore, the range of average baseline HOMA-IR was from 2.36 to 12.30. The results of the quality assessment using the modified NOS and Cochrane Collaboration’s tool can be found in Table S3 and Figure S1.

Table 1

Characteristics of the included studies

AuthorIGB group, nType of study; Country; Prevalence of diabetesNutrition and exercise prescriptionIGB type; IGB duration; Filling; Implantation of IGBLiver disease excluded
Bazerbachi et al.3721Observational study; USA; 52%Low-calorie diet + lifestyle therapyOrbera; 6 months; Liquid-filled; EndoscopicallyOther primary causes of liver disease
Maekawa et al.3818RCT; Japan; Not describedLow-carbohydrate dietOrbera; 6 months; Liquid-filled; EndoscopicallyNot described
Maekawa et al.3813RCT; Japan; Not describedLow-calorie dietOrbera; 6 months; Liquid-filled; EndoscopicallyNot described
Guedes et al.3642Observational study; Brazil; Not describedLow-calorie dietOrbera/Spatz; 6 months; Liquid-filled; EndoscopicallyNot described
Genco et al.3538Observational study; Italy; Not describedLow-calorie diet + exercise counselingElipse; 4 months; Liquid-filled; SwallowableNot described
Raftopoulos et al.3412Observational study; Greece; Not describedDiet and exercise counselingElipse; 4 months; Liquid-filled; SwallowableNo history of alcohol
Folini et al.3213Observational study; Italy; Not describedLow-calorie diet + exercise counselingOrbera; 6 months; Liquid-filled; EndoscopicallyAlcohol consumption, presence of any predisposing disorders for liver diseases, pregnancy, and lactation
Takihata et al.338Observational study; Japan; Not describedLow-calorie dietOrbera; 6 months; Liquid-filled; EndoscopicallyNot described
Tai et al.3128Observational study; China (Taiwan); Not describedLow-calorie dietOrbera; Median 200 days; Liquid-filled; EndoscopicallyAlcoholism or drug addiction
Nikolic et al.2833Observational study; Croatia; Not describedLow-calorie dietOrbera; 6 months; Liquid-filled; EndoscopicallyNot described
Sekino et al.298Observational study; Japan; Not describedNot describedOrbera; 6 months; Liquid-filled; EndoscopicallyNot described
Stimac et al.30165Observational study; Croatia; Not describedNot describedOrbera; 6 months; Liquid-filled; EndoscopicallyPresent alcohol or drug abuse
Forlano et al.27120Observational study; Italy; 13.3%Low-calorie dietOrbera; 6 months; Liquid-filled; EndoscopicallyUse of drugs reported to cause liver damage, alcohol intake of 30 g/day or more, and viral hepatitis
Donadio et al.2640Observational study; Italy; Not describedNot describedOrbera; 6 months; Liquid-filled; EndoscopicallyAlcoholism
Ricci et al.2565Observational study; Italy; Not describedLow-calorie dietOrbera; 6 months; Liquid-filled; EndoscopicallyPositivity for hepatitis B virus or hepatitis C virus, previous or current alcohol consumption >30 g/day, use of medications with reported hepatosteatogenic effect (amiodarone, tamoxifen, estrogens), and type 1 diabetes

Quantitative synthesis and stratified analyses

Nine intervention arms25–27,29,33,35,36,38 of 352 participants evaluated the effect of IGB on HOMA-IR. The pooled mean decrease in HOMA-IR levels with IGB therapy was 1.56 (95% CI=1.16–1.95, I2=61.1; Fig. 2A). According to the Egger’s test and funnel plot, no significant publication bias was present (p=0.2665; Fig. S2A). Table 2 presents the results of the stratified analyses. Both endoscopic IGB (MD=1.68, 95% CI=1.24–2.11) and swallowable IGB (MD=0.90, 95% CI=0.26–1.54) were effective in inducing HOMA-IR loss. There were trends showing the advanced age group had less change in HOMA-IR (MD=1.07, 95% CI=0.57–1.56) compared to those ≤40 years (MD=1.82, 95% CI=1.25–2.40), but the findings were not statistically significant (p=0.0502). Higher baseline HOMA-IR (>5) was associated with more significant reductions in HOMA-IR [MD=3.48 (95% CI=2.46–4.50) vs. MD=1.40 (95% CI=1.25–1.54), p<0.0001)]. Consequently, intra-subgroup heterogeneity was significantly diminished and almost absent with different basal HOMA-IR (basal HOMA-IR ≤5: I2=0.0; basal HOMA-IR >5: I2=0.0).

Forest plots.
Fig. 2  Forest plots.

HOMA-IR (A), ALT (B), AST (C), and GGT (D) decreased after IGB treatment and removal. ALT, alanine aminotransferase; AST, aspartate aminotransferase; GGT, gamma-glutamyl transpeptidase; HOMA-IR, homeostasis model assessment of insulin resistance; IGB, intragastric balloon.

Table 2

Pooled change in HOMA-IR, ALT, AST, and GGT after IGB treatment and removal: Stratified analyses

Intervention arm, nMD (95% CI)I2
Pooled change in HOMA-IR after IGB treatment and removal
Insertion of IGB (IGB type)
  Endoscopic (Orbera/Spatz)81.68 (1.24–2.11)60.3
  Swallowable (Elipse)10.90 (0.26–1.54)
Basal HOMA-IR
  ≤571.40 (1.25–1.54)0.0
  >523.48 (2.46–4.50)0.0
Mean age, years
  ≤4041.82 (1.25–2.40)82.0
  >4051.07 (0.57–1.56)0.0
Mean BMI, kg/m2
  ≤4041.35 (1.19–1.51)0.0
  >4052.01 (1.25–2.77)66.4
Region
  Asia41.35 (1.19–1.51)72.4
  Europe41.41 (1.01–1.81)29.1
  South America11.39 (1.22–1.56)
NOS scale
  High31.37 (0.88–1.87)52.0
  Fair42.16 (0.87–3.44)81.2
Pooled change in ALT after IGB treatment and removal
Insertion of IGB (IGB type)
  Endoscopic (Orbera)1010.85 (6.31–15.39)55.9
  Swallowable (Elipse)120.27 (6.49–34.05)
Basal ALT, U/L
  ≤4079.58 (6.18–12.98)38.7
  >40432.43 (18.49–46.37)0.0
Mean age, years
  ≤40610.40 (5.38–15.41)54.6
  >40515.57 (5.20–25.93)64.6
Mean BMI, kg/m2
  ≤40222.61 (11.49–33.74)0.0
  >4099.98 (5.59–14.38)53.7
Region
  Asia325.80 (9.69–41.91)0.0
  Europe79.58 (6.18–12.98)38.7
  North America19.88 (7.33–12.44)
NOS scale
  High412.71 (5.27–20.16)78.0
  Fair710.59 (4.84–16.35)29.4
Pooled change in AST after IGB treatment and removal
Insertion of IGB (IGB type)
  Endoscopic (Orbera)66.74 (0.53–12.96)60.4
  Swallowable (Elipse)18.60 (2.41–14.79)
Basal AST, U/L
  ≤4064.52 (1.05–7.99)29.8
  >40136.18 (13.62–58.74)0
Mean age, years
  ≤4043.30 (−0.66 to 7.26)29.9
  >40314.54 (−0.04 to 29.12)63.5
Mean BMI, kg/m2
  ≤4028.77 (2.95–14.58)0.0
  >4056.64 (−0.20 to 13.49)66.8
Region
  Asia311.15 (1.77–20.53)0
  Europe33.59 (−0.34 to 7.52)52.1
  North America136.18 (13.62–58.74)
NOS scale
  High217.67 (−14.52 to 49.86)87.7
  Fair56.17 (0.53–11.81)38.2
Pooled change in GGT after IGB treatment and removal
Insertion of IGB (IGB type)
  Endoscopic (Orbera)89.45 (4.46–14.45)53.0
  Swallowable (Elipse)0
Basal GGT, U/L
  ≤4068.74 (2.89–14.59)66.2
  >40212.96 (−0.23 to 26.15)0.0
Mean age, years
  ≤4058.75 (1.71–15.79)71.3
  >4038.80 (2.02–15.58)0.0
Mean BMI, kg/m2
  ≤4089.45 (4.46–14.45)53.0
  >400
Region
  Asia212.96 (−0.23 to 26.15)0.0
  Europe68.74 (2.89–14.59)66.2
NOS scale
  High310.10 (2.49–17.72)80.1
  Fair57.88 (1.86–13.89)0.0

Meta-regression

Table 3 presents the meta-regression findings of HOMA-IR. In univariate meta-regression, basal HOMA-IR of the participants (slope coefficient=0.3966, 95% CI=0.1119–0.6814, p=0.0063) and percentage of male individuals (slope coefficient=0.0433, 95% CI=0.0183 to 0.0684, p=0.0007) seemed to be factors significantly associated with reductions in HOMA-IR. Subsequently, using a multivariate meta-regression approach, our final model consisted of four covariates: basal HOMA-IR, percentage of male individuals, age and BMI of the participants. Greater HOMA-IR loss was predicted by younger age (slope coefficient=−0.0932, 95% CI=−0.1647 to −0.0216, p=0.0107).

Table 3

Univariate and multivariate meta-regression analyses on the mean deference of HOMA-IR, ALT, AST, and GGT after IGB treatment and removal

ModeratorsIntervention arm, nUnivariable analysis
Multivariable analysis
Slope coefficient (95% CI)pSlope coefficient (95% CI)p
Mean deference of HOMA-IR after IGB treatment and removal
Basal HOMA-IR90.3966 (0.1119–0.6814)0.00630.2361 (−0.2764 to 0.7487)0.3665
Mean age9−0.0753 (−0.1891 to 0.0386)0.1950−0.0932 (−0.1647 to −0.0216)0.0107
Mean BMI90.0975 (−0.0288 to 0.2238)0.1302−0.0473 (−0.1298 to 0.0351)0.2607
Male90.0433 (0.0183–0.0684)0.00070.0381 (−0.0094 to 0.0856)0.1159
Mean deference of ALT after IGB treatment and removal
Basal ALT110.7314 (0.3862–1.0767)<0.00010.7135 (0.3213–1.1057)0.0004
Mean age110.8603 (−0.3376 to 2.0582)0.15930.3408 (−0.5602 to 1.2417)0.4585
Mean BMI11−1.1489 (−2.6746 to 0.3767)0.1399−0.5035 (−1.6750 to 0.6681)0.3996
Male110.0851 (−0.3480 to 0.5181)0.7002−0.0652 (−0.3742 to 0.2437)0.6791
Mean deference of AST after IGB treatment and removal
Basal AST70.7650 (0.3319–1.1982)0.00050.5438 (−0.0501 to 1.1378)0.0727
Mean age71.4430 (0.5644–2.3216)0.00130.5348 (−0.8576 to 1.9272)0.4516
Mean BMI7−0.1374 (−1.5495 to 1.2747)0.8488−0.1086 (−0.8199 to 0.6028)0.7648
Male70.1362 (−0.2020 to 0.4744)0.42990.0699 (−0.2378 to 0.3777)0.6562
Mean deference of GGT after IGB treatment and removal
Basal GGT80.7968 (0.2032–1.3904)0.00851.3773 (0.4793–2.2754)0.0026
Mean age80.5022 (−2.0156 to 3.0201)0.69580.3219 (−1.9139 to 2.5577)0.7778
Mean BMI8−0.2371 (−4.2184 to 3.7441)0.9071−1.3277 (−4.6165 to 1.9612)0.4288
Male80.0996 (−0.3201 to 0.5193)0.6418−0.4310 (−0.9670 to 0.1051)0.1151

Decrease in ALT after IGB therapy

Summary of study characteristics

Eleven observational longitudinal studies25–34,37 with a total of 513 individuals were included in this meta-analysis of ALT level, and their characteristics are summarized in Table 1. All included studies were published after 2007. The participants included in the meta-analysis of ALT level came from six countries (China, Croatia, Greece, Italy, Japan, USA). Ten studies25–33,37 applied the Orbera IGB system, and one study34 reported results with the Elipse IGB system. Furthermore, the range of average baseline ALT was from 26.0 to 91.6 U/L. The results of the quality assessment using the modified NOS can be found in Table S3.

Quantitative synthesis and stratified analyses

Eleven studies25–34,37 of 513 participants evaluated the effect of IGB on ALT. The pooled mean decrease of ALT with IGB therapy was 11.53 U/L (95% CI=7.10–15.96, I2=55.4; Fig. 2B). According to the Egger’s test and funnel plot, no significant publication bias was present (p=0.2422; Fig. S2B). Table 2 presents the results of the stratified analyses. Both endoscopic IGB (MD=10.85 U/L, 95% CI=6.31–15.39) and swallowable IGB (MD=20.27 U/L, 95% CI=6.49–34.05) were effective in inducing ALT loss. The advanced age group had similar change in ALT (MD =15.57 U/L, 95% CI=5.20–25.93) compared to those ≤40 years (MD =10.40 U/L, 95% CI=5.38–15.41). Higher baseline ALT (>40 U/L) was associated with more significant reductions in ALT [MD=32.43 U/L (95% CI=18.49–46.37) vs. MD=9.58 U/L (95% CI=6.18–12.98), p=0.0018]. Overall, intra-subgroup heterogeneity in different basal ALT diminished significantly and was classified as a low (basal ALT ≤40 U/L: I2=38.7; basal ALT >40 U/L: I2=0.0).

Meta-regression

Table 3 presented the meta-regression findings of ALT. In univariate meta-regression, basal ALT of the participants (slope coefficient=0.7314, 95% CI=0.3862–1.0767, p<0.0001) seemed to be a factor significantly associated with reductions in ALT. Subsequently, using a multivariate meta-regression approach, our final model consisted of four covariates: basal ALT; percentage of male individuals; age; and BMI. Effectiveness on ALT was predicted by basal ALT (slope coefficient=0.7135, 95% CI=0.3213–1.1057, p=0.0004). The scatter plot showed a linear trend towards increasing effectiveness of IGB therapy with increasing basal ALT of the participants (Fig. 3A).

Scatter plots.
Fig. 3  Scatter plots.

(A) Change in ALT is positively correlated with basal ALT. (B) Change in GGT is positively correlated with basal GGT. ALT, alanine aminotransferase; GGT, gamma-glutamyl transpeptidase.

Decrease in AST after IGB therapy

Summary of study characteristics

Seven observational longitudinal studies26,28,29,31,33,34,37 with a total of 150 individuals were included in this meta-analysis of AST level, and their characteristics are summarized in Table 1. The participants included in the meta-analysis of AST level came from six countries (China, Croatia, Greece, Italy, Japan, USA). Six studies26,28,29,31,33,37 applied the Orbera IGB system, and one study34 reported results with the Elipse IGB system. Furthermore, the range of average baseline AST was from 21.7 to 67.5 U/L. The results of the quality assessment using the modified NOS can be found in Table S3.

Quantitative synthesis and stratified analyses

Seven studies of 150 participants evaluated the effect of IGB on AST. The pooled mean decrease of AST with IGB therapy was 6.79 U/L (95% CI=1.69–11.90, I2=59.9; Fig. 2C). According to the Egger’s test and funnel plot, no significant publication bias was present (p=0.3768; Fig. S2C). Table 2 presents the results of the stratified analyses. Both endoscopic IGB (MD=6.74 U/L, 95% CI=0.53–12.96) and swallowable IGB (MD=8.60 U/L, 95% CI=2.41–14.79) were effective in inducing AST loss. The advanced age group had a similar change in AST (MD =14.54 U/L, 95% CI=−0.04 to 29.12) compared to those ≤40 years (MD=3.30 U/L, 95% CI=−0.66 to 7.26). Higher baseline AST (>40 U/L) was associated with more significant reductions in AST [MD=36.18 U/L (95% CI=13.62–58.74) vs. MD=4.52 U/L (95% CI=1.05-7.99, p=0.0065)]. Overall, intra-subgroup heterogeneity in different basal AST diminished significantly and was classified as a low (basal AST ≤40 U/L: I2=29.8; basal AST >40 U/L: I2=0.0).

Meta-regression

Table 3 presents the meta-regression findings of AST. In univariate meta-regression, basal AST of the participants (slope coefficient=0.7650, 95% CI=0.3319–1.1982, p=0.0005) and age of the participants (slope coefficient=1.4430, 95% CI=0.5644–2.3216, p=0.0013) seemed to be factors significantly associated with reductions in AST. Subsequently, using a multivariate meta-regression approach, our final model consisted of four covariates: basal AST; percentage of male individuals; age; and BMI. Effectiveness on AST could not be predicted by all of the above covariates.

Decrease in GGT after IGB therapy

Summary of study characteristics

Eight observational longitudinal studies25–30,32,33 with a total of 452 individuals were included in this meta-analysis of GGT level, and their characteristics are summarized in Table 1. The participants included in the meta-analysis of GGT level came from three countries (Croatia, Italy, Japan). All eight studies25–30,32,33 applied the Orbera IGB system. Furthermore, the range of average baseline GGT was from 29.8 to 53.0 U/L. The results of the quality assessment using the modified NOS can be found in Table S3.

Quantitative synthesis and stratified analyses

Eight studies25–30,32,33 of 452 participants evaluated the effect of IGB on GGT. The pooled mean decrease of GGT with IGB therapy was 10.54 U/L (95% CI=6.32–14.75, I2=37.6; Fig. 2D). According to the Egger’s test and funnel plot, no significant publication bias was present (p=0.8620; Fig. S2D). Table 2 presented the results of the stratified analyses. The advanced age group had a similar change in GGT (MD =8.80 U/L, 95% CI=2.02–15.58) compared to those ≤40 years (MD=8.75 U/L, 95% CI=1.71–15.79). There were trends showing that the higher basal GGT group had more change in GGT (MD=12.96, 95% CI=−0.23 to 26.15) compared to those ≤40 U/L (MD=8.74, 95% CI=2.89–14.59) but the findings were not statistically significant (p=0.6919). Overall, intra-subgroup heterogeneity diminished significantly in the higher basal GGT group (I2=0.0).

Meta-regression

Table 3 presents the meta-regression findings of GGT. In univariate meta-regression, basal GGT of the participants (slope coefficient=0.7968, 95% CI=0.2032–1.3904, p=0.0085) seemed to be a factor significantly associated with reductions in GGT. Subsequently, using a multivariate meta-regression approach, our final model consisted of four covariates: basal GGT; percentage of male individuals; age; and BMI. Effectiveness on GGT was predicted by basal GGT (slope coefficient=1.3773, 95% CI=0.4793–2.2754, p=0.0026). The scatter plot showed a linear trend towards increasing effectiveness of IGB therapy with increasing basal GGT of the participants (Fig. 3B).

Discussion

Principal findings and relevant mechanisms

IGB is the most widely available EBMT with proven efficacy in inducing weight loss. According to the IGB type, an empty balloon is introduced into the stomach by an upper gastrointestinal endoscopy or by swallowing the balloon capsule directly. The liquid-filled IGB is inflated with saline and methylene blue to occupy the space in the stomach. After that, the IGB dwells in the stomach for 4 to 6 months until it ruptures or is removed.14,39 Due to its moderate efficacy of weight loss and excellent safety profiles, the potential utility of IGB was mentioned by the Asian-Pacific clinical practice guideline on MAFLD.40 IGB has also been employed for clinical research of fatty liver disease. However, there is still substantial heterogeneity in results across studies. One explanation is that patients with fatty liver disease can be subdivided into IGB responder and non-responder groups. In this systematic review with meta-analysis, we demonstrated that IGB could reverse the serum markers of MAFLD, including HOMA-IR, ALT, AST, and GGT levels. Furthermore, the change of ALT and GGT with IGB therapy had a positive linear relationship with the basal value. This means that even at higher levels of disease severity, abnormal liver enzymes can be controlled within the reported range of included studies (ALT: 26.0–91.6 U/L; GGT: 29.8–53.0 U/L).

Due to the dearth of eligible studies, the histological and radiological findings cannot be quantitatively pooled through meta-analyses and can only be described in the discussion. In terms of histological variables, a small RCT,17 with 18 patients who completed the study, reported that NAFLD activity score at post-therapy was significantly lower among the IGB-treated compared with the sham-treated arm. On the other hand, there seemed to be no difference between the IGB-treated arm and the sham-treated arm in improving fibrosis. Consistent with this finding, according to another observational study,37 significant improvement of NAFLD activity score was reached in most NAFLD patients treated with IGB (p<0.001). Apart from these, some of the studies assessed non-invasive radiological parameters of NAFLD. A prospective single-arm study27 showed that after 6 months of IGB therapy, the number of patients with severe hepatic steatosis confirmed by abdominal ultrasound decreased from 52% to 4%. Two other clinical studies,32,37 respectively, demonstrated that hepatic fat fraction and fibrosis by magnetic resonance imaging could be significantly alleviated by IGB therapy. Taken together, these histological and radiological findings were consistent with the results of serum markers (HOMA-IR, ALT, AST, and GGT) in our meta-analyses.

To date, no study has looked at the impact of age on insulin resistance amelioration in patients receiving IGB therapy. In our meta-analysis, multivariate linear meta-regression and stratified analyses indicated that participants with advanced age had less change in HOMA-IR after IGB therapy. Several weight-dependent and non-weight-dependent hypotheses may explain this phenomenon. A previously published study reported that advanced age was significantly correlated with less excess weight loss in females after IGB intervention.41 Given that clinically significant weight loss can alleviate insulin resistance,42 age-related differences in insulin resistance outcomes might be partly attributed to the different weight loss during treatment. Additionally, both obesity and aging are linked to and engender insulin resistance.43 Among elderly patients, the effect of aging is strongly amplified and cannot be eliminated by the obesity management tools. Taken together, age might be considered as a predictor of insulin resistance amelioration in patients undergoing IGB therapy.

Comparison with other studies or reviews

In terms of the impact of IGB on liver enzymes, a commendable meta-analysis published in 2016 showed that the use of IGB could decrease ALT (MD=10.02, 95% CI=6.8–13.2),19 which was in line with our findings. When their meta-analysis was published, swallowable IGB had not been widely used and investigated.14 To help clinicians and researchers keep up to date with current evidence, we performed this systematic review including more updated studies. Our stratified analysis revealed that the improvement of ALT, AST, and HOMA-IR with swallowable IGB therapy was no worse than that with endoscopic IGB. Future RCTs are needed to comprehensively compare the efficacy and safety between these two IGBs.

Limitations and strengths

Our systematic review does have some shortcomings. First, although our review included studies of both endoscopic and swallowable IGB, there were still a number of IGB types (such as ReShape Duo Balloon and Obalon Gastric Balloon) not mentioned in the current review due to the lack of relevant clinical research.14 Second, at the time of the preliminary search, we found that most of the clinical studies in this field were of longitudinal observational design. Thus, when formal screening of the search was performed, we defined the patient at baseline, but not the sham-treated group, as comparators. However, this approach ignored the potential for spontaneous remission of the disease.44 Despite these limitations, our systematic review provides the most comprehensive evaluation of the effect of IGB on the serum markers of MAFLD, with low intra-subgroup heterogeneity in stratified analysis, suggesting that the evidence is highly credible. More impressively, our observations demonstrate for the first time that age has an adverse effect on IGB treatment of insulin resistance.

Conclusions and perspectives

IGB therapy has led to improvements in the serum markers of MAFLD, including HOMA-IR, ALT, AST, and GGT. Significant reductions in HOMA-IR and liver biochemical parameters were seen across different methods of balloon implantation and different age/BMI classes. The improvement of insulin resistance and liver biochemistries with swallowable IGB therapy was no worse than that with endoscopic IGB. Furthermore, greater insulin resistance amelioration with IGB therapy was predicted by younger age and the relevant mechanism needs further investigation. Although IGB has the potential to become a multidisciplinary management tool of MAFLD, it cannot be ignored that IGB is a temporary measure. If the patient cannot maintain an active lifestyle after the first balloon is removed, relapse of MAFLD is an expected result. In this regard, IGB combined with other pharmacotherapy or sequential IGB therapy could be a potential solution, and further RCT is warranted.

Supporting information

Supporting Fig. 1

Quality assessment of included RCTs.

(TIF)

Supporting Fig. 2

Funnel plots.

HOMA-IR (A), ALT (C), AST (C), and GGT (D) decreased after IGB treatment and removal.

(TIF)

Supporting Table 1

Reporting checklist for a systematic review with meta-analysis.

(DOCX)

Supporting Table 2

Search strategy.

(DOCX)

Supporting Table 3

NOS quality assessment scale.

(DOCX)

Abbreviations

ALT: 

alanine aminotransferase

AST: 

aspartate aminotransferase

BMI: 

body mass index

CI: 

confidence interval

EBMT: 

endoscopic bariatric and metabolic therapies

GGT: 

gamma-glutamyl transpeptidase

HOMA-IR: 

homeostasis model assessment of insulin resistance

IGB: 

intragastric balloon

I2

inconsistency index

MD: 

mean difference

NAFLD: 

nonalcoholic fatty liver disease

NOS: 

Newcastle-Ottawa scale

MAFLD: 

metabolic dysfunction-associated fatty liver disease

RCT: 

randomized controlled trial

Declarations

Data sharing statement

No additional data are available.

Funding

This study was supported by the National Key R&D Program of China (2017YFC0908903), National Natural Science Foundation of China (81873565, 81900507), Shanghai Leading Talent Plan 2017, Innovative Research Team of High-Level Local Universities in Shanghai, and Hospital Funded Clinical Research, Clinical Research Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (17CSK04).

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

Conception and design (JGF), funding acquisition and supervision (JGF, TYR), collection and assembly of data (ZYZ, JZ), data analysis and interpretation (ZYZ), manuscript writing (ZYZ, JZ, TYR, YWS, RXY, JGF), final approval of manuscript (ZYZ, JZ, TYR, YWS, RXY, JGF).

References

  1. Sanyal AJ. Past, present and future perspectives in nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol 2019;16(6):377-386 View Article
  2. Paik JM, Golabi P, Younossi Y, Mishra A, Younossi ZM. Changes in the global burden of chronic liver diseases from 2012 to 2017: the growing impact of NAFLD. Hepatology 2020;72(5):1605-1616 View Article
  3. Eslam M, Newsome PN, Sarin SK, Anstee QM, Targher G, Romero-Gomez M, et al. A new definition for metabolic dysfunction-associated fatty liver disease: an international expert consensus statement. J Hepatol 2020;73(1):202-209 View Article
  4. Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nat Med 2018;24(7):908-922 View Article
  5. Baldwin D, Chennakesavalu M, Gangemi A. Systematic review and meta-analysis of Roux-en-Y gastric bypass against laparoscopic sleeve gastrectomy for amelioration of NAFLD using four criteria. Surg Obes Relat Dis 2019;15(12):2123-2130 View Article
  6. Fakhry TK, Mhaskar R, Schwitalla T, Muradova E, Gonzalvo JP, Murr MM. Bariatric surgery improves nonalcoholic fatty liver disease: a contemporary systematic review and meta-analysis. Surg Obes Relat Dis 2019;15(3):502-511 View Article
  7. Lee Y, Doumouras AG, Yu J, Brar K, Banfield L, Gmora S, et al. Complete resolution of nonalcoholic fatty liver disease after bariatric surgery: a systematic review and meta-analysis. Clin Gastroenterol Hepatol 2019;17(6):1040-1060.e11 View Article
  8. Martin M, Beekley A, Kjorstad R, Sebesta J. Socioeconomic disparities in eligibility and access to bariatric surgery: a national population-based analysis. Surg Obes Relat Dis 2010;6(1):8-15 View Article
  9. Penn L, Moffatt SM, White M. Participants’ perspective on maintaining behaviour change: a qualitative study within the European Diabetes Prevention Study. BMC Public Health 2008;8:235 View Article
  10. Chalasani N, Younossi Z, Lavine JE, Charlton M, Cusi K, Rinella M, et al. The diagnosis and management of nonalcoholic fatty liver disease: practice guidance from the American Association for the Study of Liver Diseases. Hepatology 2018;67(1):328-357 View Article
  11. Abu Dayyeh BK, Bazerbachi F, Graupera I, Cardenas A. Endoscopic bariatric and metabolic therapies for non-alcoholic fatty liver disease. J Hepatol 2019;71(6):1246-1248 View Article
  12. Salomone F, Sharaiha RZ, Boškoski I. Endoscopic bariatric and metabolic therapies for non-alcoholic fatty liver disease: evidence and perspectives. Liver Int 2020;40(6):1262-1268 View Article
  13. Abu Dayyeh BK, Edmundowicz S, Thompson CC. Clinical practice update: expert review on endoscopic bariatric therapies. Gastroenterology 2017;152(4):716-729 View Article
  14. Bazerbachi F, Vargas EJ, Abu Dayyeh BK. Endoscopic bariatric therapy: a guide to the intragastric balloon. Am J Gastroenterol 2019;114(9):1421-1431 View Article
  15. Frutos MD, Morales MD, Luján J, Hernández Q, Valero G, Parrilla P. Intragastric balloon reduces liver volume in super-obese patients, facilitating subsequent laparoscopic gastric bypass. Obes Surg 2007;17(2):150-154 View Article
  16. Lewis MC, Phillips ML, Slavotinek JP, Kow L, Thompson CH, Toouli J. Change in liver size and fat content after treatment with optifast very low calorie diet. Obes Surg 2006;16(6):697-701 View Article
  17. Lee YM, Low HC, Lim LG, Dan YY, Aung MO, Cheng CL, et al. Intragastric balloon significantly improves nonalcoholic fatty liver disease activity score in obese patients with nonalcoholic steatohepatitis: a pilot study. Gastrointest Endosc 2012;76(4):756-760 View Article
  18. Page MJ, Moher D. Evaluations of the uptake and impact of the Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement and extensions: a scoping review. Syst Rev 2017;6(1):263 View Article
  19. Popov VB, Thompson CC, Kumar N, Ciarleglio MM, Deng Y, Laine L. Effect of intragastric balloons on liver enzymes: a systematic review and meta-analysis. Dig Dis Sci 2016;61(9):2477-2487 View Article
  20. Stang A. Critical evaluation of the Newcastle-Ottawa scale for the assessment of the quality of nonrandomized studies in meta-analyses. Eur J Epidemiol 2010;25(9):603-605 View Article
  21. Higgins JP, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, et al. The cochrane collaboration’s tool for assessing risk of bias in randomised trials. BMJ 2011;343:d5928 View Article
  22. Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ 1997;315(7109):629-634 View Article
  23. Egger M, Smith GD, Phillips AN. Meta-analysis: principles and procedures. BMJ 1997;315(7121):1533-1537 View Article
  24. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ 2003;327(7414):557-560 View Article
  25. Ricci G, Bersani G, Rossi A, Pigò F, De Fabritiis G, Alvisi V. Bariatric therapy with intragastric balloon improves liver dysfunction and insulin resistance in obese patients. Obes Surg 2008;18(11):1438-1442 View Article
  26. Donadio F, Sburlati LF, Masserini B, Lunati EM, Lattuada E, Zappa MA, et al. Metabolic parameters after BioEnterics intragastric balloon placement in obese patients. J Endocrinol Invest 2009;32(2):165-168 View Article
  27. Forlano R, Ippolito AM, Iacobellis A, Merla A, Valvano MR, Niro G, et al. Effect of the BioEnterics intragastric balloon on weight, insulin resistance, and liver steatosis in obese patients. Gastrointest Endosc 2010;71(6):927-933 View Article
  28. Nikolic M, Mirosevic G, Ljubicic N, Boban M, Supanc V, Nikolic BP, et al. Obesity treatment using a Bioenterics intragastric balloon (BIB)—preliminary Croatian results. Obes Surg 2011;21(8):1305-1310 View Article
  29. Sekino Y, Imajo K, Sakai E, Uchiyama T, Iida H, Endo H, et al. Time-course of changes of visceral fat area, liver volume and liver fat area during intragastric balloon therapy in Japanese super-obese patients. Intern Med 2011;50(21):2449-2455 View Article
  30. Stimac D, Majanović SK, Turk T, Kezele B, Licul V, Orlić ZC. Intragastric balloon treatment for obesity: results of a large single center prospective study. Obes Surg 2011;21(5):551-555 View Article
  31. Tai CM, Lin HY, Yen YC, Huang CK, Hsu WL, Huang YW, et al. Effectiveness of intragastric balloon treatment for obese patients: one-year follow-up after balloon removal. Obes Surg 2013;23(12):2068-2074 View Article
  32. Folini L, Veronelli A, Benetti A, Pozzato C, Cappelletti M, Masci E, et al. Liver steatosis (LS) evaluated through chemical-shift magnetic resonance imaging liver enzymes in morbid obesity; effect of weight loss obtained with intragastric balloon gastric banding. Acta Diabetol 2014;51(3):361-368 View Article
  33. Takihata M, Nakamura A, Aoki K, Kimura M, Sekino Y, Inamori M, et al. Comparison of intragastric balloon therapy and intensive lifestyle modification therapy with respect to weight reduction and abdominal fat distribution in super-obese Japanese patients. Obes Res Clin Pract 2014;8(4):e331-338 View Article
  34. Raftopoulos I, Giannakou A. The Elipse Balloon, a swallowable gastric balloon for weight loss not requiring sedation, anesthesia or endoscopy: a pilot study with 12-month outcomes. Surg Obes Relat Dis 2017;13(7):1174-1182 View Article
  35. Genco A, Ernesti I, Ienca R, Casella G, Mariani S, Francomano D, et al. Safety and efficacy of a new swallowable intragastric balloon not needing endoscopy: early Italian experience. Obes Surg 2018;28(2):405-409 View Article
  36. Guedes MR, Fittipaldi-Fernandez RJ, Diestel CF, Klein MRST. Impact of intragastric balloon treatment on adipokines, cytokines, and metabolic profile in obese individuals. Obes Surg 2019;29(8):2600-2608 View Article
  37. Bazerbachi F, Vargas EJ, Rizk M, Maselli DB, Mounajjed T, Venkatesh SK, et al. Intragastric balloon placement induces significant metabolic and histologic improvement in patients with nonalcoholic steatohepatitis. Clin Gastroenterol Hepatol 2021;19(1):146-154.e4 View Article
  38. Maekawa S, Niizawa M, Harada M. A comparison of the weight loss effect between a low-carbohydrate diet and a calorie-restricted diet in combination with intragastric balloon therapy. Intern Med 2020;59(9):1133-1139 View Article
  39. Laing P, Pham T, Taylor LJ, Fang J. Filling the void: a review of intragastric balloons for obesity. Dig Dis Sci 2017;62(6):1399-1408 View Article
  40. Eslam M, Sarin SK, Wong VW, Fan JG, Kawaguchi T, Ahn SH, et al. The asian pacific association for the study of the liver clinical practice guidelines for the diagnosis and management of metabolic associated fatty liver disease. Hepatol Int 2020;14(6):889-919 View Article
  41. Diab AF, Abdurasul EM, Diab FH. The effect of age, gender, and baseline BMI on weight loss outcomes in obese patients undergoing intragastric balloon therapy. Obes Surg 2019;29(11):3542-3546 View Article
  42. Swift DL, Johannsen NM, Lavie CJ, Earnest CP, Blair SN, Church TS. Effects of clinically significant weight loss with exercise training on insulin resistance and cardiometabolic adaptations. Obesity (Silver Spring) 2016;24(4):812-819 View Article
  43. Gong Z, Tas E, Yakar S, Muzumdar R. Hepatic lipid metabolism and non-alcoholic fatty liver disease in aging. Mol Cell Endocrinol 2017;455:115-130 View Article
  44. Han MAT, Altayar O, Hamdeh S, Takyar V, Rotman Y, Etzion O, et al. Rates of and factors associated with placebo response in trials of pharmacotherapies for nonalcoholic steatohepatitis: systematic review and meta-analysis. Clin Gastroenterol Hepatol 2019;17(4):616-629.e26 View Article
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Efficacy of Intragastric Balloons in the Markers of Metabolic Dysfunction-associated Fatty Liver Disease: Results from Meta-analyses

Zi-Yuan Zou, Jing Zeng, Tian-Yi Ren, Yi-Wen Shi, Rui-Xu Yang, Jian-Gao Fan
  • Reset Zoom
  • Download TIFF