Next Article in Journal
Congruence between Physical Activity Patterns and Dietary Patterns Inferred from Analysis of Sex Differences in Lifestyle Behaviors of Late Adolescents from Poland: Cophylogenetic Approach
Previous Article in Journal
Inflammatory Response: A Crucial Way for Gut Microbes to Regulate Cardiovascular Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Medicines for Obesity: Appraisal of Clinical Studies with Grading of Recommendations, Assessment, Development, and Evaluation Tool

by
Eleni A. Karavia
1,
Panagiota C. Giannopoulou
1,
Vassiliki Konstantinopoulou
1,
Katerina Athanasopoulou
1,
Theodosios D. Filippatos
2,
Demosthenes Panagiotakos
3 and
Kyriakos E. Kypreos
1,4,*
1
Pharmacology Laboratory, Department of Medicine, University of Patras School of Health Sciences, Patras 26504, Greece
2
Department of Internal Medicine, School of Medicine, University of Crete, Heraklion 71500, Greece
3
School of Health Sciences and Education, Harokopio University, Athens 17676, Greece
4
Department of Life Sciences, School of Sciences, European University Cyprus, Nicosia 1516, Cyprus
*
Author to whom correspondence should be addressed.
Nutrients 2023, 15(3), 606; https://doi.org/10.3390/nu15030606
Submission received: 18 December 2022 / Revised: 13 January 2023 / Accepted: 19 January 2023 / Published: 24 January 2023
(This article belongs to the Section Nutrition and Obesity)

Abstract

:
We evaluated the quality of evidence from phase III/IV clinical trials of drugs against obesity using the principles of Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) tool. Our systematic review evaluates the quality of clinical evidence from existing clinical trials and not the pharmacological efficacy of anti-obesity therapies. A literature search using select keywords in separate was performed in PubMed and ClinicalTrials.gov databases for phase III/IV clinical trials during the last ten years. Our findings indicate that the quality of existing clinical evidence from anti-obesity trials generally ranges from low to moderate. Most trials suffered from publication bias. Less frequently, trials suffered from the risk of bias mainly due to lack of blindness in the treatment. Our work indicates that additional higher-quality clinical trials are needed to gain more confidence in the estimate of the effect of currently used anti-obesity medicines, to allow more informed clinical decisions, thus reducing the risk of implementing potentially ineffective or even harmful therapeutic strategies.

1. Introduction

“Lifestyle diseases” is a contemporary, modern definition used to describe a range of disorders related to unhealthy habits such as excess food consumption and caloric intake, low physical activity, and disturbed circadian clock. They include coronary heart and fatty liver disease, obesity, and type 2 diabetes mellitus (T2DM) [1]. These disorders affect a very large part of the global population, and even though current treatment strategies are constantly updated, including the use of novel drugs, there are still numerous unmet medical needs to be addressed and many pharmacological challenges to tackle.
Obesity is a chronic, metabolic disorder characterized by increased fat depots, either in number or in size [2,3]. It is considered as the central pathology of metabolic syndrome, which may also include two of the following conditions: hypertension, disturbed plasma glucose homeostasis, and a distinct type of dyslipidemia characterized by increased plasma triglyceride levels and reduced HDL cholesterol levels. Phenotypically, obesity combines increased body mass, mainly due to adipose tissue expansion, and lean mass augmentation [4]. Undoubtedly, a prolonged positive energy balance contributes to the development of the disease [5]; yet, many other biological, environmental, and social parameters may also be implicated in the etiology of the disease [5,6]. In 2016, the World Health Organization reported that 39% of adults were overweight and 13% were obese, while the prevalence of overweight and obesity among children and adolescents was 18%. Impressively, it was reported that in 2019, 38 million children younger than the age of five were overweight and obese [7] (accessed on 24 June 2022). Obesity, abdominal fat, and dysfunctional adipose tissue are associated with the development of cardiovascular diseases, T2DM, several cancers, and hepatic or renal diseases [8], which are considered some of the primary causes of disability and death [9]. Therapeutic lifestyle changes (TLCs), such as changes in dietary habits and exercise, are often associated with significant weight loss and a normalization of body weight. Nevertheless, often times TLCs fail to produce sustained results and the use of medicines is mandated. In cases of morbid obesity with very high body mass indices, surgical weight loss appears the most effective solution. In recent years, bariatric surgical interventions have been used very effectively for sustained weight loss and normalization of plasma lipid levels, glucose tolerance, and insulin sensitivity in morbidly obese patients [10]. In particular, malabsorptive bariatric procedures have been proven more effective compared to restrictive bariatric procedures aiming at reducing food intake [11].
Therapeutic strategies for obesity aim not only at weight loss but also at prevention of weight regain [6,12], refinement of body composition, treatment of comorbidities, decrease in health risk, and improvement of life quality. Both lifestyle interventions and pharmacotherapy contribute significantly to this direction [6,12,13]; however, the use of medicines is usually recommended only for individuals with a body mass index (BMI) ≥30 kg/m2 without comorbidities or a BMI ≥27 kg/m2 if at least one obesity-related comorbidity, such as dyslipidemia or diabetes, is present [14]. Currently, both the Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have given their approval to semaglutide orlistat, liraglutide, and the combination of naltrexone/bupropion for the indication of obesity. The combination of phentermine/topiramate is approved only by the FDA. These active pharmaceutical compounds (APC) act through various mechanisms; for example, orlistat diminishes the intestinal digestion of lipids via highly specific inhibition of pancreatic and gastric lipases in the gastrointestinal tract [15], while liraglutide and semaglutide are long-acting agonists for the receptor of glucagon-like peptide-1 (GLP-1) [16]. Notably, the approved fixed-dose combinations aim at improving effectiveness and safety [12]. Particularly, a pooled-data clinical trial showed that phentermine/topiramate combination therapy is more effective in reducing body weight compared to either topiramate or phentermine monotherapies [17]. The combination of an opioid receptor antagonist (naltrexone) with a dopamine and norepinephrine reuptake inhibitor (bupropion) in an extended-release tablet is more efficacious in terms of weight loss compared with bupropion monotherapy [18].
The use of medicines for obesity poses a significant financial burden on health care systems all over the world and it should be mandated by strong clinical evidence supporting their efficacy. In the absence of quality clinical data, the risk and cost may far exceed the benefit, advocating against the use of these drugs.
In the present systematic review, we applied the Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) tool [19] to assess if medicines currently used for obesity are effective based on the quality of existing clinical evidence. Addressing this question could also be very important for health technology assessment organizations and guideline committees, in assisting their decisions.

2. Methods

GRADE is typically an instrument to evaluate a body of evidence for a particular patient/intervention/comparison/outcome, also known as PICO. Briefly, GRADE provides a methodology for the classification of quality of evidence focusing on specific quality factors [20]. These factors include risk of bias [21], inconsistency [22], indirectness [23], imprecision [24], and publication bias [25], all affected by individual parameters, as presented in brief in Table 1. In our paper, emphasis is placed on the evaluation of clinical evidence rather than on the efficacy of each medicine, which is not a subject of GRADE; that is, to determine what the evidence from a set of studies for each drug tells us about a given PICO. To facilitate our work, prior to GRADE implementation in each set of trials, every single trial was pre-evaluated based on critical “YES or NO” questions regarding the GRADE factors shown in Table 1. The limiting factors, which, according to GRADE, were identified to affect the quality of evidence in each trial are listed in Table 2 and in more detail, in Table S1 of Supplementary Data.
Following this initial screening of individual trials, for each drug, we then proceeded to an overall evaluation of the body of relevant clinical evidence from all studies, according to the GRADE handbook: https://gdt.gradepro.org/app/handbook/handbook.html [49] (accessed on 24 June 2022), and the results are listed in Table 3. The evaluation of clinical evidence for each PICO was performed using the academic version of GRADEpro tool (GRADEpro GDT: GRADEpro Guideline Development Tool [Software], McMaster University, 2020 (developed by Evidence Prime, Inc., available from gradepro.org, accessed on 10 January 2023)). The results of the overall evaluations for the anti-obesity drugs are presented in Supplemental Data.
The clinical trials included in the present study were extracted from PubMed using the following keywords in separate: “obesity”, “anti-obesity drugs”, “anti-obesity effects”, “anti-obesity drug effectiveness”, “anti-obesity medicine effectiveness”, “orlistat obesity effectiveness”, “liraglutide obesity effectiveness”, “naltrexone/bupropion obesity effectiveness”, “semaglutide obesity effectiveness” “phentermine obesity effectiveness”, “topiramate obesity effectiveness”, and the following filters were applied: Clinical Trial, Phase III, Clinical Trial, Phase IV, 10 years, Humans, English. The search retrieved studies published over the last 10 years, based on human species and in English language. In addition, the ClinicalTrials.gov database was used to identify additional studies and collect more relevant information. Advanced research was performed with the following fields: “Condition or disease: obesity”, “Other terms: weight loss or weight loss effect or orlistat or liraglutide or naltrexon/bupropion or semaglutide”, “Study Types: All Studies”, “Study Results: Studies with results”, “Phase: 3”, “Phase: 4”, “Study start: From 1 January 2012to 8 November 2022”. The following characteristics were extracted from the original papers using a standardized data extraction form: design of the study, lead author, year of publication, country of origin, and sample size. The drugs evaluated in this systematic review are drugs tested in multiple clinical trials, namely, naltrexone/bupropion, liraglutide, lorcaserin, semaglutide, and exenatide. Other drugs, such as bupropion, phentermine/topiramate, empagliflozin, ertugliflozin, amfepramone, rimonabant, setmelanotide, glargine, and tirzepatide, found only in single clinical trials were not included since GRADE is not applied to single studies.
PCG, VK, and KA performed a literature search independently, and EAK, GP, and KEK participated in the independent appraisal of the collected manuscripts and GRADE evaluation of the clinical evidence. Disagreements were solved by consensus and by the expert opinion of TF, AK, and DP. All authors contributed to the drafting of the final submitted version of the manuscript. The search and sorting strategies for our qualitative synthesis are summarized in Figure 1. The PRISMA flow diagram was created according to the guidelines described in [50].

3. Results

3.1. Naltrexone/Bupropion

The combination of naltrexone, an opioid μ-receptor antagonist, with bupropion, a dual norepinephrine and dopamine reuptake inhibitor, was approved by the FDA for obesity treatment in 2014.
A clinical trial (CONTRAVE Obesity Research-II (COR-II), NCT00567255) [26] showed that 1496 individuals with obesity (BMI: 30–45 kg/m2) or overweight (27–45 kg/m2) and with dyslipidemia and/or hypertension who received naltrexone/bupropion experienced greater weight loss compared to the placebo group.
Another clinical trial (NCT01764386) [27] showed that in 242 subjects with obesity or overweight with dyslipidemia and/or controlled hypertension, treatment with naltrexone SR/bupropion SR in conjunction with a lifestyle intervention program for 26 weeks improved weight loss, which was sustained for 78 weeks, compared to usual care.
The clinical evidence supporting the effectiveness of naltrexone/bupropion combination compared to placebo or usual care for weight loss is of low quality (Table 2 and Table 3).

3.2. Liraglutide

Liraglutide is a GLP-1 agonist used for the improvement of glycemic control in patients with type 2 diabetes mellitus (T2DM). Different clinical trials indicated a beneficial effect of GLP-1 analogs on obesity; thus, liraglutide was also licensed as a weight loss agent, with encouraging results in phase III clinical trials [34,52].
A recent post hoc analysis of pooled data from four phase IIIa trials in patients with either a minimum BMI of 27 kg/m2 and (at least) one comorbidity or a minimum BMI of 30 kg/m2 compared the efficacy and safety of liraglutide versus placebo, combined with a low-calorie diet and physical activity (NCT01272219, NCT01272232, NCT01557166, NCT00781937) [28]. The study showed that both Hispanic (n = 534) and non-Hispanic (n = 4597) subgroups had clinically significant mean weight loss after treatment with liraglutide, while more patients in both subgroups lost ≥5%, >10%, and >15% of their initial weight compared to placebo.
Furthermore, another randomized placebo-controlled, 12-week clinical trial in 72 overweight individuals and individuals with obesity, all of which had type 1 diabetes as comorbidity, showed that the addition of an either high (1.2 and 1.8 mg) or low (0.6 mg) liraglutide dose to insulin therapy resulted in significant weight reduction (NCT01722266) [29].
A 56-week, randomized, double-blind, placebo-controlled, multinational, multicenter trial with 396 participants (SCALE™ Insulin, NCT02963922) [30] showed that overweight individuals or individuals with obesity, all of which were insulin-treated for T2DM, increased weight loss compared to placebo when liraglutide was added as an adjunct to intensive behavioral therapy (IBT).
Similarly, a 56-week, randomized, double-blind, placebo-controlled, multicenter trial with 282 participants (SCALE™ IBT, NCT02963935) [31] showed that at week 56, the mean weight loss with liraglutide plus IBT was greater compared to placebo combined with IBT.
Furthermore, a randomized, double-blind trial (NCT02918279) showed that the use of liraglutide (3.0 mg) for 56 weeks plus lifestyle therapy led to a significantly greater reduction in the BMI standard deviation score compared to placebo plus lifestyle therapy in adolescents with obesity [32].
Moreover, in a 56-week, double-blind trial involving 3731 patients with either a minimum BMI of 30 kg/m2 or a minimum BMI of 27 kg/m2 with treated or untreated dyslipidemia or hypertension, treatment with 3.0 mg of liraglutide, as an adjunct to diet and exercise, was associated with reduced body weight and improved metabolic control (NCT01272219) [33].
Additionally, the use of subcutaneous liraglutide (3.0 mg) daily as an adjunct to 500 kcal/d dietary deficit and increased physical activity, compared to placebo, resulted in weight loss in overweight individuals and participants with obesity and T2DM over 56 weeks (NCT01272232) [34].
Finally, a clinical trial (NCT02911818) with 150 adults with obesity suggested a potential benefit on weight loss from adding 3.0 mg liraglutide to IBT for obesity compared to IBT alone [53].
Overall, the current clinical evidence for liraglutide is of moderate quality (Table 2 and Table 3).

3.3. Lorcaserin

Lorcaserin is a selective 5-HT2C receptor agonist with indication for weight management, in combination with lifestyle modification, for overweight participants or participants affected by obesity with more than one weight-related comorbidity [54].
Two phase III clinical trials of similar design, the Behavioral Modification and Lorcaserin for Overweight and Obesity Management (BLOOM, NCT00395135, n = 3182) and the Behavioral Modification and Lorcaserin Second Study for Obesity Management (BLOSSOM, NCT00603902, n = 4008), assessed the safety and effectiveness of lorcaserin in adults without T2DM. A pooled analysis of these clinical trials showed that lorcaserin-treated patients, in combination with diet and physical activity, lost significantly more body weight compared to the placebo group [35].
Moreover, a post hoc analysis of the BLOOM-DM trial (NCT00603291) suggested that lorcaserin treatment of overweight patients and patients affected by obesity with T2DM for 52 weeks promoted weight reduction and facilitated glycemic control compared to placebo [36].
Finally, in a 12-week, randomized, double-blind pilot safety study (NCT01987427) with 238 nondiabetic patients with obesity or overweight with more than one comorbidity, the use of lorcaserin added to phentermine enhanced short-term weight loss [37].
Overall, current clinical evidence for lorcaserin is of moderate quality (Table 2 and Table 3). On 13 February 2020, FDA requested that the manufacturer of pharmaceutical products containing the active ingredient lorcaserin voluntarily withdraw the weight loss drug from the U.S. market because a safety clinical trial showed an increased occurrence of cancer [55] (accessed on 24 June 2022) .

3.4. Semaglutide

In 2014, the FDA approved a subcutaneous injection of semaglutide, a GLP-1 receptor agonist, (2.4 mg once weekly) for chronic weight management in adults with a BMI ≥30 kg/m2 or BMI ≥27 kg/m2 with at least one obesity-related disorder. The use of the drug is recommended along with a low-calorie diet and increased physical activity.
In a double-blind trial (NCT03548935) with 1961 adults with a BMI ≥30 kg/m2 or a BMI ≥27 kg/m2 with at least one weight-related coexisting condition, except T2DM, semaglutide was administered subcutaneous once weekly for 68 weeks at a dose of 2.4 mg in combination with lifestyle intervention. Compared to placebo, semaglutide was associated with a sustained, clinically relevant reduction in body weight [38].
A phase IIIa, open-label, parallel-group, randomized controlled trial (SUSTAIN 3, NCT01885208) [39] with 813 participants with T2DM indicated that semaglutide was superior to exenatide extended-release in improving glycemic control and reducing body weight at week 56 of treatment.
A randomized, double-blind study (STEP 3, NCT03611582) including 611 overweight adults or adults affected by obesity without diabetes showed that once-a-week subcutaneous semaglutide, used as an adjunct to IBT and an initial low-calorie diet, compared to placebo, resulted in significantly greater weight loss over 68 weeks [40].
Furthermore, treatment once a week of subcutaneous semaglutide at a does of 2.4 mg plus a lifestyle intervention for 68 weeks for weight management in overweight adults or adults with obesity and T2DM achieved a superior decrease in body weight compared to placebo (STEP 2, NCT03552757) [41].
Moreover, a randomized, open-label, 68-week trial showed that among 338 overweight patients or patients affected by obesity without diabetes, once-weekly subcutaneous semaglutide, compared to once-daily subcutaneous liraglutide in combination with counseling for diet and physical activity, resulted in significantly greater weight loss (STEP 8, NCT04074161) [42].
In addition, in a phase IIIb, open-label trial where 577 adults with T2DM were randomized to subcutaneous once-weekly semaglutide (1.0 mg) or subcutaneous once-daily liraglutide (1.2 mg), semaglutide was superior to liraglutide in reducing body weight (SUSTAIN 10, NCT03191396) [43].
Another randomized, double-blind, 68-week phase IIIa withdrawal study suggested that in patients who completed a 20-week run-in period with subcutaneous semaglutide, 2.4 mg once weekly, maintaining treatment with semaglutide compared to switching to placebo resulted in continued weight loss over the following 48 weeks (STEP 4, NCT03548987) [44].
Finally, another clinical study with adults with T2DM who were treated with a sodium glucose co-transporter-2 (SGLT-2) inhibitor and were randomly assigned to receive subcutaneous semaglutide (1.0 mg) or placebo once weekly for 30 weeks showed that adding semaglutide to SGLT-2 inhibitor therapy significantly reduce bodyweight (SUSTAIN 9, NCT03086330) [45].
Overall, current clinical evidence for semaglutide is of low quality (Table 2 and Table 3).

3.5. Exenatide

Exenatide, another GLP-1 receptor agonist [56], is indicated as an adjunct to diet and exercise to improve glycemic control in adults with T2DM; however, it, also appears to reduce body weight in clinical trials [57].
A recent randomized, multicenter, double-blind study (DURATION-7, NCT02229383) including 464 patients with persistent hyperglycemia showed that exenatide once weekly significantly decreased body weight, a secondary clinical outcome of the trial, compared to placebo [46].
Recently, exenatide was evaluated in combination with dapagliflozin, another antidiabetic drug that inhibits SGLT-2 in the kidney. Specifically, in a post hoc analysis of 695 patients with T2DM inadequately controlled with metformin monotherapy (subpopulations from the DURATION-8 trial, NCT02229396), it was found that 28-week treatment with exenatide once weekly plus dapagliflozin resulted in a body weight reduction that was no greater than that observed with exenatide once weekly or dapagliflozin alone [47]. For each group, weight loss was numerically greater as baseline BMI increased.
Finally, a randomized, placebo-controlled study including 182 overweight women and women with obesity (NCT01590433) showed that in women who demonstrated ≥5% weight loss at 12 weeks, longer-term weight loss with exenatide treatment was similar to that achieved with a hypocaloric diet [48].
The use of exenatide compared to placebo or dapagliflozin for weight loss is based on evidence of low quality (Table 2 and Table 3).

4. Conclusions

GRADE is composed of several criteria aimed at evaluating the quality of a body of evidence for a particular patient cohort, intervention, population, and outcome. Even though the assessment of some GRADE criteria may be subjected to the opinion of the evaluator (for example, the type of a study population or the magnitude of effect), most of them are largely objective in nature as they pertain to the design of the clinical trial. Indeed, GRADE has gained global acceptance and has been endorsed by a large number of societies and institutions in many different medical specialties [58,59,60].
Based on our assessment, the quality of evidence from most trials assessing anti-obesity medicines ranges from low to moderate (Table 3). As such, in cases where the rating was low, further research is very likely to have an important impact on our confidence in the estimate of effect and is likely to change the estimate. In all other cases where the rating of the quality of evidence was moderate, further research is likely to have an impact, and may change the estimate. Most trials suffer from publication bias since the evidence arose from small trials (number of participants < 1000) funded by the marketing authorization holder. Less frequently, trials suffered from the risk of bias mainly due to a lack of blindness in the allocation of treatment(s). In addition, imprecision due to inappropriate optimal information size was found in a few trials, while inconsistency was not found.
Though the quality of evidence is important, we should keep in mind that clinical recommendations may consider other parameters such as balance between desirable and undesirable effects, patient values, patient preferences, and treatment cost. Therefore, it should be noted that in addition to objective criteria for deciding on a rating, GRADE also includes some subjective parameters that may be hard to evaluate [61,62].
Our work is unique and quite novel in its nature, context, and impact in the sense that in addition to reviewing phase III/IV clinical evidence, it engages in a critical assessment of this evidence based on the strict methodology and impartial, highly objective approach of GRADE. Despite encouraging data, some crucial questions remain regarding the quality of clinical evidence that supports the effectiveness of many of these drugs. It should be noted that our systematic review evaluates the quality of clinical evidence from existing clinical trials and not the pharmacological efficacy of anti-obesity therapies. In addition, in our work, we did not perform a comparison of pharmacological efficacy among different medications. In the absence of head-to-head clinical trials, comparing different APCs for efficacy would lead to the activation of the indirectness criterion, which would further reduce the confidence in clinical evidence. A limitation that we identified in our study is the fact that our analysis did not include studies currently in progress whose data have not been published yet. Such studies, when completed, could impact the quality of clinical evidence. Overall, our findings raise the possibility that additional higher-quality, free-of-limitations clinical trials are needed to gain more confidence in the estimate of the effect of currently used anti-obesity medicines and to allow more informed clinical decisions, thus reducing the risk of implementing potentially ineffective or even harmful therapeutic strategies.

Supplementary Materials

The following supporting information can be downloaded at https://www.mdpi.com/article/10.3390/nu15030606/s1, Table S1: Results from the PICO analysis for each medicine evaluated in the study. The factors identified to affect the quality of clinical evidence are listed in detail in the Explanations under each PICO table.

Author Contributions

Conceptualization, K.E.K. and E.A.K.; methodology, K.E.K., E.A.K., D.P. and T.D.F.; literature search P.C.G., V.K. and K.A.; GRADE evaluation of clinical evidence: E.A.K., K.E.K., D.P. and T.D.F.; original draft preparation, K.E.K. and E.A.K.; writing—review and editing, K.E.K., E.A.K., D.P., T.D.F., V.K. and K.A.; project administration, K.E.K.; funding acquisition, K.E.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the project “INSPIRED” (MIS 5002550), under the Action “Reinforcement of the Research and Innovation Infrastructure”, funded by the Operational Program “Competitiveness, Entrepreneurship and Innovation” (NSRF 2014–2020).

Institutional Review Board Statement

Not Applicable.

Informed Consent Statement

Not Applicable.

Data Availability Statement

Not Applicable.

Conflicts of Interest

Theodosios D. Filippatos reports lecture honoraria from Boehringer Ingelheim, Mylan, Vi-atris, Astra Zeneca, Lilly, Recordati, Bausch Health, Servier and Innovis. All other authors report no conflicting interests.

References

  1. Sharma, M.; Majumdar, P.K. Occupational Lifestyle Diseases: An Emerging Issue. Indian J. Occup. Environ. Med. 2009, 13, 109–112. [Google Scholar] [CrossRef] [Green Version]
  2. Hollstein, T.; Piaggi, P. Metabolic Factors Determining the Susceptibility to Weight Gain: Current Evidence. Curr. Obes. Rep. 2020, 9, 121–135. [Google Scholar] [CrossRef]
  3. Knittle, J.L.; Timmers, K.; Ginsberg-Fellner, F.; Brown, R.E.; Katz, D.P. The Growth of Adipose Tissue in Children and Adolescents. Cross-Sectional and Longitudinal Studies of Adipose Cell Number and Size. J. Clin. Investig. 1979, 63, 239–246. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Webster, J.D.; Hesp, R.; Garrow, J.S. The Composition of Excess Weight in Obese Women Estimated by Body Density, Total Body Water and Total Body Potassium. Hum. Nutr. Clin. Nutr. 1984, 38, 299–306. [Google Scholar]
  5. Heymsfield, S.B.; Wadden, T.A. Mechanisms, Pathophysiology, and Management of Obesity. N. Engl. J. Med. 2017, 376, 254–266. [Google Scholar] [CrossRef] [PubMed]
  6. Yumuk, V.; Tsigos, C.; Fried, M.; Schindler, K.; Busetto, L.; Micic, D.; Toplak, H. European Guidelines for Obesity Management in Adults. Obes. Facts 2015, 8, 402–424. [Google Scholar] [CrossRef] [PubMed]
  7. Key Facts by WHO for Obesity and Overweight. Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 24 June 2022).
  8. MacMahon, S.; Baigent, C.; Duffy, S.; Rodgers, A.; Tominaga, S.; Chambless, L.; De Backer, G.; De Bacquer, D.; Kornitzer, M.; Whincup, P.; et al. Body-Mass Index and Cause-Specific Mortality in 900 000 Adults: Collaborative Analyses of 57 Prospective Studies. Lancet 2009, 373, 1083–1096. [Google Scholar] [CrossRef] [Green Version]
  9. Frühbeck, G.; Toplak, H.; Woodward, E.; Yumuk, V.; Maislos, M.; Oppert, J.M. Obesity: The Gateway to Ill Health—An EASO Position Statement on a Rising Public Health, Clinical and Scientific Challenge in Europe. Obes. Facts 2013, 6, 117–120. [Google Scholar] [CrossRef] [PubMed]
  10. Sjöström, L.; Lindroos, A.-K.; Peltonen, M.; Torgerson, J.; Bouchard, C.; Carlsson, B.; Dahlgren, S.; Larsson, B.; Narbro, K.; David Sjöström, C.; et al. Lifestyle, Diabetes, and Cardiovascular Risk Factors 10 Years after Bariatric Surgery From the Departments of Body Composi-Tion and Metabolism L. N. Engl. J. Med. 2004, 23, 351. [Google Scholar]
  11. Bult, M.J.F.; van Dalen, T.; Muller, A.F. Surgical Treatment of Obesity. Eur. J. Endocrinol. 2008, 158, 135–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Toplak, H.; Woodward, E.; Yumuk, V.; Oppert, J.M.; Halford, J.C.G.; Frühbeck, G. 2014 EASO Position Statement on the Use of Anti-Obesity Drugs. Obes. Facts 2015, 8, 166–174. [Google Scholar] [CrossRef]
  13. Bray, G.A. Medical Treatment of Obesity: The Past, the Present and the Future. Best Pract. Res. Clin. Gastroenterol. 2014, 28, 665–684. [Google Scholar] [CrossRef] [PubMed]
  14. Jensen, M.D.; Ryan, D.H.; Apovian, C.M.; Ard, J.D.; Comuzzie, A.G.; Donato, K.A.; Hu, F.B.; Hubbard, V.S.; Jakicic, J.M.; Kushner, R.F.; et al. 2013 AHA/ACC/TOS Guideline for the Management of Overweight and Obesity in Adults: A Report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Obesity Society. J. Am. Coll. Cardiol. 2014, 63, 2985–3023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ballinger, A.; Peikin, S.R. Orlistat: Its Current Status as an Anti-Obesity Drug. Eur. J. Pharmacol. 2002, 440, 109–117. [Google Scholar] [CrossRef]
  16. Crane, J.; McGowan, B. The GLP-1 Agonist, Liraglutide, as a Pharmacotherapy for Obesity. Ther. Adv. Chronic Dis. 2016, 7, 92–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Thompson, J.; Heaton, P.; Kelton, C. Efficacy of Phentermine Monotherapy, Topiramate Monotherapy, and Phentermine/Topiramate Combination Therapy on Weight Loss: A Network Meta-Analysis of Randomized Controlled Trial Data. Value Health 2014, 17, A224. [Google Scholar] [CrossRef] [Green Version]
  18. Plodkowski, R.A.; Nguyen, Q.; Sundaram, U.; Nguyen, L.; Chau, D.L.; St Jeor, S. Bupropion and Naltrexone: A Review of Their Use Individually and in Combination for the Treatment of Obesity. Expert Opin. Pharmacother. 2009, 10, 1069–1081. [Google Scholar] [CrossRef]
  19. Guyatt, G.H.; Oxman, A.D.; Vist, G.E.; Kunz, R.; Falck-Ytter, Y.; Schünemann, H.J. GRADE: What Is “Quality of Evidence” and Why Is It Important to Clinicians? BMJ 2008, 336, 995–998. [Google Scholar] [CrossRef] [Green Version]
  20. Guyatt, G.; Oxman, A.D.; Akl, E.A.; Kunz, R.; Vist, G.; Brozek, J.; Norris, S.; Falck-Ytter, Y.; Glasziou, P.; Debeer, H.; et al. GRADE Guidelines: 1. Introduction-GRADE Evidence Profiles and Summary of Findings Tables. J. Clin. Epidemiol. 2011, 64, 383–394. [Google Scholar] [CrossRef]
  21. Guyatt, G.H.; Oxman, A.D.; Vist, G.; Kunz, R.; Brozek, J.; Alonso-Coello, P.; Montori, V.; Akl, E.A.; Djulbegovic, B.; Falck-Ytter, Y.; et al. GRADE Guidelines: 4. Rating the Quality of Evidence—Study Limitations (Risk of Bias). J. Clin. Epidemiol. 2011, 64, 407–415. [Google Scholar] [CrossRef]
  22. Guyatt, G.H.; Oxman, A.D.; Kunz, R.; Woodcock, J.; Brozek, J.; Helfand, M.; Alonso-Coello, P.; Glasziou, P.; Jaeschke, R.; Akl, E.A.; et al. GRADE Guidelines: 7. Rating the Quality of Evidence—Inconsistency. J. Clin. Epidemiol. 2011, 64, 1294–1302. [Google Scholar] [CrossRef]
  23. Guyatt, G.H.; Oxman, A.D.; Kunz, R.; Woodcock, J.; Brozek, J.; Helfand, M.; Alonso-Coello, P.; Falck-Ytter, Y.; Jaeschke, R.; Vist, G.; et al. GRADE Guidelines: 8. Rating the Quality of Evidence—Indirectness. J. Clin. Epidemiol. 2011, 64, 1303–1310. [Google Scholar] [CrossRef] [PubMed]
  24. Guyatt, G.H.; Oxman, A.D.; Kunz, R.; Brozek, J.; Alonso-Coello, P.; Rind, D.; Devereaux, P.J.; Montori, V.M.; Freyschuss, B.; Vist, G.; et al. GRADE Guidelines 6. Rating the Quality of Evidence--Imprecision. J. Clin. Epidemiol. 2011, 64, 1283–1293. [Google Scholar] [CrossRef] [PubMed]
  25. Guyatt, G.H.; Oxman, A.D.; Montori, V.; Vist, G.; Kunz, R.; Brozek, J.; Alonso-Coello, P.; Djulbegovic, B.; Atkins, D.; Falck-Ytter, Y.; et al. GRADE Guidelines: 5. Rating the Quality of Evidence—Publication Bias. J. Clin. Epidemiol. 2011, 64, 1277–1282. [Google Scholar] [CrossRef]
  26. Apovian, C.M.; Aronne, L.; Rubino, D.; Still, C.; Wyatt, H.; Burns, C.; Kim, D.; Dunayevich, E. A Randomized, Phase 3 Trial of Naltrexone SR/Bupropion SR on Weight and Obesity-Related Risk Factors (COR-II). Obesity 2013, 21, 935–943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Halseth, A.; Shan, K.; Walsh, B.; Gilder, K.; Fujioka, K. Method-of-use Study of Naltrexone Sustained Release (SR)/Bupropion SR on Body Weight in Individuals with Obesity. Obes. (Silver Spring) 2017, 25, 338. [Google Scholar] [CrossRef] [PubMed]
  28. O’Neil, P.M.; Garvey, W.T.; Gonzalez-Campoy, J.M.; Mora, P.; Ortiz, R.V.; Guerrero, G.; Claudius, B.; Pi-Sunyer, X. Effects of Liraglutide 3.0 Mg on Weight and Risk Factors in Hispanic versus Non-Hipanic Populations: Subgroup Analysis from Scale Randomized Trials. Endocr. Pract. 2016, 22, 1277–1287. [Google Scholar] [CrossRef]
  29. Kuhadiya, N.D.; Dhindsa, S.; Ghanim, H.; Mehta, A.; Makdissi, A.; Batra, M.; Sandhu, S.; Hejna, J.; Green, K.; Bellini, N.; et al. Addition of Liraglutide to Insulin in Patients with Type 1 Diabetes: A Randomized Placebo-Controlled Clinical Trial of 12 Weeks. Diabetes Care 2016, 39, 1027–1035. [Google Scholar] [CrossRef] [Green Version]
  30. Garvey, W.T.; Birkenfeld, A.L.; Dicker, D.; Mingrone, G.; Pedersen, S.D.; Satylganova, A.; Skovgaard, D.; Sugimoto, D.; Jensen, C.; Mosenzon, O. Efficacy and Safety of Liraglutide 3.0 Mg in Individuals with Overweight or Obesity and Type 2 Diabetes Treated with Basal Insulin: The SCALE Insulin Randomized Controlled Trial. Diabetes Care 2020, 43, 1085–1093. [Google Scholar] [CrossRef] [Green Version]
  31. Wadden, T.A.; Tronieri, J.S.; Sugimoto, D.; Lund, M.T.; Auerbach, P.; Jensen, C.; Rubino, D. Liraglutide 3.0 Mg and Intensive Behavioral Therapy (IBT) for Obesity in Primary Care: The SCALE IBT Randomized Controlled Trial. Obesity 2020, 28, 529–536. [Google Scholar] [CrossRef]
  32. Kelly, A.S.; Auerbach, P.; Barrientos-Perez, M.; Gies, I.; Hale, P.M.; Marcus, C.; Mastrandrea, L.D.; Prabhu, N.; Arslanian, S. A Randomized, Controlled Trial of Liraglutide for Adolescents with Obesity. N. Engl. J. Med. 2020, 382, 2117–2128. [Google Scholar] [CrossRef] [PubMed]
  33. Pi-Sunyer, X.; Astrup, A.; Fujioka, K.; Greenway, F.; Halpern, A.; Krempf, M.; Lau, D.C.W.; le Roux, C.W.; Violante Ortiz, R.; Jensen, C.B.; et al. A Randomized, Controlled Trial of 3.0 Mg of Liraglutide in Weight Management. N. Engl. J. Med. 2015, 373, 11–22. [Google Scholar] [CrossRef] [PubMed]
  34. Davies, M.J.; Bergenstal, R.; Bode, B.; Kushner, R.F.; Lewin, A.; Skjøth, T.V.; Andreasen, A.H.; Jensen, C.B.; DeFronzo, R.A.; Valensi, P.; et al. Efficacy of Liraglutide for Weight Loss among Patients with Type 2 Diabetes: The SCALE Diabetes Randomized Clinical Trial. JAMA-J. Am. Med. Assoc. 2015, 314, 687–699. [Google Scholar] [CrossRef] [Green Version]
  35. Aronne, L.; Shanahan, W.; Fain, R.; Glicklich, A.; Soliman, W.; Li, Y.; Smith, S. Safety and Efficacy of Lorcaserin: A Combined Analysis of the BLOOM and BLOSSOM Trials. Postgrad. Med. 2014, 126, 7–18. [Google Scholar] [CrossRef] [PubMed]
  36. Pi-Sunyer, X.; Shanahan, W.; Fain, R.; Ma, T.; Garvey, W.T. Impact of Lorcaserin on Glycemic Control in Overweight and Obese Patients with Type 2 Diabetes: Analysis of Week 52 Responders and Nonresponders. Postgrad. Med. 2016, 128, 591–597. [Google Scholar] [CrossRef] [PubMed]
  37. Smith, S.R.; Garvey, W.T.; Greenway, F.L.; Zhou, S.; Fain, R.; Pilson, R.; Fujioka, K.; Aronne, L.J. Coadministration of Lorcaserin and Phentermine for Weight Management: A 12-Week, Randomized, Pilot Safety Study. Obes. (Silver Spring) 2017, 25, 857–865. [Google Scholar] [CrossRef]
  38. Wilding, J.P.H.; Batterham, R.L.; Calanna, S.; Davies, M.; Van Gaal, L.F.; Lingvay, I.; McGowan, B.M.; Rosenstock, J.; Tran, M.T.D.; Wadden, T.A.; et al. Once-Weekly Semaglutide in Adults with Overweight or Obesity. N. Engl. J. Med. 2021, 384, 989–1002. [Google Scholar] [CrossRef]
  39. Ahmann, A.J.; Capehorn, M.; Charpentier, G.; Dotta, F.; Henkel, E.; Lingvay, I.; Holst, A.G.; Annett, M.P.; Aroda, V.R. Efficacy and Safety of Once-Weekly Semaglutide versus Exenatide ER in Subjects with Type 2 Diabetes (SUSTAIN 3): A 56-Week, Open-Label, Randomized Clinical Trial. In Proceedings of the Diabetes Care; American Diabetes Association Inc.: Arlington County, VA, USA, 2018; Volume 41, pp. 258–266. [Google Scholar]
  40. Wadden, T.A.; Bailey, T.S.; Billings, L.K.; Davies, M.; Frias, J.P.; Koroleva, A.; Lingvay, I.; O’Neil, P.M.; Rubino, D.M.; Skovgaard, D.; et al. Effect of Subcutaneous Semaglutide vs Placebo as an Adjunct to Intensive Behavioral Therapy on Body Weight in Adults With Overweight or Obesity: The STEP 3 Randomized Clinical Trial. JAMA 2021, 325, 1403–1413. [Google Scholar] [CrossRef] [PubMed]
  41. Davies, M.; Færch, L.; Jeppesen, O.K.; Pakseresht, A.; Pedersen, S.D.; Perreault, L.; Rosenstock, J.; Shimomura, I.; Viljoen, A.; Wadden, T.A.; et al. Semaglutide 2·4 Mg Once a Week in Adults with Overweight or Obesity, and Type 2 Diabetes (STEP 2): A Randomised, Double-Blind, Double-Dummy, Placebo-Controlled, Phase 3 Trial. Lancet 2021, 397, 971–984. [Google Scholar] [CrossRef]
  42. Rubino, D.M.; Greenway, F.L.; Khalid, U.; O’Neil, P.M.; Rosenstock, J.; Sørrig, R.; Wadden, T.A.; Wizert, A.; Garvey, W.T. Effect of Weekly Subcutaneous Semaglutide vs Daily Liraglutide on Body Weight in Adults With Overweight or Obesity Without Diabetes: The STEP 8 Randomized Clinical Trial. JAMA 2022, 327, 138–150. [Google Scholar] [CrossRef]
  43. Capehorn, M.S.; Catarig, A.M.; Furberg, J.K.; Janez, A.; Price, H.C.; Tadayon, S.; Vergès, B.; Marre, M. Efficacy and Safety of Once-Weekly Semaglutide 1.0 mg vs. Once-Daily Liraglutide 1.2 mg as Add-on to 1-3 Oral Antidiabetic Drugs in Subjects with Type 2 Diabetes (SUSTAIN 10). Diabetes Metab. 2020, 46, 100–109. [Google Scholar] [CrossRef] [PubMed]
  44. Rubino, D.; Abrahamsson, N.; Davies, M.; Hesse, D.; Greenway, F.L.; Jensen, C.; Lingvay, I.; Mosenzon, O.; Rosenstock, J.; Rubio, M.A.; et al. Effect of Continued Weekly Subcutaneous Semaglutide vs Placebo on Weight Loss Maintenance in Adults With Overweight or Obesity: The STEP 4 Randomized Clinical Trial. JAMA 2021, 325, 1414–1425. [Google Scholar] [CrossRef] [PubMed]
  45. Zinman, B.; Bhosekar, V.; Busch, R.; Holst, I.; Ludvik, B.; Thielke, D.; Thrasher, J.; Woo, V.; Philis-Tsimikas, A. Semaglutide Once Weekly as Add-on to SGLT-2 Inhibitor Therapy in Type 2 Diabetes (SUSTAIN 9): A Randomised, Placebo-Controlled Trial. Lancet Diabetes Endocrinol. 2019, 7, 356–367. [Google Scholar] [CrossRef] [PubMed]
  46. Guja, C.; Frías, J.P.; Somogyi, A.; Jabbour, S.; Wang, H.; Hardy, E.; Rosenstock, J. Effect of Exenatide QW or Placebo, Both Added to Titrated Insulin Glargine, in Uncontrolled Type 2 Diabetes: The DURATION-7 Randomized Study. Diabetes Obes. Metab. 2018, 20, 1602–1614. [Google Scholar] [CrossRef] [Green Version]
  47. Jabbour, S.A.; Frías, J.P.; Guja, C.; Hardy, E.; Ahmed, A.; Öhman, P. Effects of Exenatide Once Weekly plus Dapagliflozin, Exenatide Once Weekly, or Dapagliflozin, Added to Metformin Monotherapy, on Body Weight, Systolic Blood Pressure, and Triglycerides in Patients with Type 2 Diabetes in the DURATION-8 Study. Diabetes Obes. Metab. 2018, 20, 1515–1519. [Google Scholar] [CrossRef] [Green Version]
  48. Rodgers, M.; Migdal, A.L.; Rodríguez, T.G.; Chen, Z.Z.; Nath, A.K.; Gerszten, R.E.; Kasid, N.; Toschi, E.; Tripaldi, J.; Heineman, B.; et al. Weight Loss Outcomes Among Early High Responders to Exenatide Treatment: A Randomized, Placebo Controlled Study in Overweight and Obese Women. Front. Endocrinol. (Lausanne) 2021, 12. [Google Scholar] [CrossRef] [PubMed]
  49. Available online: https://gdt.gradepro.org/app/handbook/handbook.html (accessed on 24 June 2022).
  50. Guidelines for Creating a PRISMA Flow Diagram. Available online: https://guides.lib.unc.edu/prisma/step-by-step#:~:text=Step%206%3A%20Reports%20Sought%20for,your%20number%20sought%20for%20retrieval (accessed on 24 June 2022).
  51. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  52. Astrup, A.; Rössner, S.; Van Gaal, L.; Rissanen, A.; Niskanen, L.; Al Hakim, M.; Madsen, J.; Rasmussen, M.F.; Lean, M.E. Effects of Liraglutide in the Treatment of Obesity: A Randomised, Double-Blind, Placebo-Controlled Study. Lancet 2009, 374, 1606–1616. [Google Scholar] [CrossRef]
  53. Wadden, T.A.; Walsh, O.A.; Berkowitz, R.I.; Chao, A.M.; Alamuddin, N.; Gruber, K.; Leonard, S.; Mugler, K.; Bakizada, Z.; Tronieri, J.S. Intensive Behavioral Therapy for Obesity Combined with Liraglutide 3.0 Mg: A Randomized Controlled Trial. Obes. (Silver Spring) 2019, 27, 75–86. [Google Scholar] [CrossRef]
  54. Thomsen, W.J.; Grottick, A.J.; Menzaghi, F.; Reyes-Saldana, H.; Espitia, S.; Yuskin, D.; Whelan, K.; Martin, M.; Morgan, M.; Chen, W.; et al. Lorcaserin, a Novel Selective Human 5-Hydroxytryptamine2C Agonist: In Vitro and in Vivo Pharmacological Characterization. J. Pharmacol. Exp. Ther. 2008, 325, 577–587. [Google Scholar] [CrossRef] [Green Version]
  55. FDA Drug Safety Podcast. Available online: https://www.fda.gov/drugs/fda-drug-safety-podcasts/fda-requests-withdrawal-weight-loss-drug-belviq-belviq-xr-lorcaserin-market (accessed on 24 June 2022).
  56. Parkes, D.G.; MacE, K.F.; Trautmann, M.E. Discovery and Development of Exenatide: The First Antidiabetic Agent to Leverage the Multiple Benefits of the Incretin Hormone, GLP-1. Expert Opin. Drug Discov. 2013, 8, 219–244. [Google Scholar] [CrossRef] [PubMed]
  57. Gupta, V. Glucagon-like Peptide-1 Analogues: An Overview. Indian J. Endocrinol. Metab. 2013, 17, 413. [Google Scholar] [CrossRef] [PubMed]
  58. Broek, J.L.; Akl, E.A.; Alonso-Coello, P.; Lang, D.; Jaeschke, R.; Williams, J.W.; Phillips, B.; Lelgemann, M.; Lethaby, A.; Bousquet, J.; et al. Grading Quality of Evidence and Strength of Recommendations in Clinical Practice Guidelines. Part 1 of 3. An Overview of the GRADE Approach and Grading Quality of Evidence about Interventions. Allergy 2009, 64, 669–677. [Google Scholar] [CrossRef]
  59. Swiglo, B.A.; Murad, M.H.; Schünemann, H.J.; Kunz, R.; Vigersky, R.A.; Guyatt, G.H.; Montori, V.M. A Case for Clarity, Consistency, and Helpfulness: State-of-the-Art Clinical Practice Guidelines in Endocrinology Using the Grading of Recommendations, Assessment, Development, and Evaluation System. J. Clin. Endocrinol. Metab. 2008, 93, 666–673. [Google Scholar] [CrossRef] [Green Version]
  60. De Palma, R.; Liberati, A.; Ciccone, G.; Bandieri, E.; Belfiglio, M.; Ceccarelli, M.; Leoni, M.; Longo, G.; Magrini, N.; Marangolo, M.; et al. Developing Clinical Recommendations for Breast, Colorectal, and Lung Cancer Adjuvant Treatments Using the GRADE System: A Study from the Programma Ricerca e Innovazione Emilia Romagna Oncology Research Group. J. Clin. Oncol. 2008, 26, 1033–1039. [Google Scholar] [CrossRef] [Green Version]
  61. Riva, J.J.; Bhatt, M.; Brunarski, D.J.; Busse, J.W.; Martins, C.C.; Xie, F.; Schünemann, H.J.; Brozek, J.L. Guidelines That Use the GRADE Approach Often Fail to Provide Complete Economic Information for Recommendations: A Systematic Survey. J. Clin. Epidemiol. 2021, 136, 203–215. [Google Scholar] [CrossRef]
  62. Hilton Boon, M.; Thomson, H.; Shaw, B.; Akl, E.A.; Lhachimi, S.K.; López-Alcalde, J.; Klugar, M.; Choi, L.; Saz-Parkinson, Z.; Mustafa, R.A.; et al. Challenges in Applying the GRADE Approach in Public Health Guidelines and Systematic Reviews: A Concept Article from the GRADE Public Health Group. J. Clin. Epidemiol. 2021, 135, 42–53. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Strategy for collecting and evaluating clinical evidence for anti-obesity medicines in the context of this systematic review. PRISMA 2020 flow diagram for new systematic reviews, which included searches of databases and registers only. * GRADE cannot be applied in a single study; thus, in this review, drugs found in only one study were excluded. ** Studies that include clinical trials from 2009 were excluded since our research focuses on the last ten years. *** Studies that include clinical trials evaluated in other included studies. ¥ This study includes two different studies, which we evaluated separately; thus, the number of “Studies included in review” is 24 and not 22 [51].
Figure 1. Strategy for collecting and evaluating clinical evidence for anti-obesity medicines in the context of this systematic review. PRISMA 2020 flow diagram for new systematic reviews, which included searches of databases and registers only. * GRADE cannot be applied in a single study; thus, in this review, drugs found in only one study were excluded. ** Studies that include clinical trials from 2009 were excluded since our research focuses on the last ten years. *** Studies that include clinical trials evaluated in other included studies. ¥ This study includes two different studies, which we evaluated separately; thus, the number of “Studies included in review” is 24 and not 22 [51].
Nutrients 15 00606 g001
Table 1. Critical questions to evaluate the clinical evidence of each study, based on GRADE factors.
Table 1. Critical questions to evaluate the clinical evidence of each study, based on GRADE factors.
QUESTIONSACTION
1. Risk of Bias
1a. Randomized?Yes = maintain, No = downgrade
1b. Double blind?Yes = maintain, No = downgrade
1c. Dropouts/Withdrawals accounted for?Yes = maintain, No = downgrade
1d. Large loses to follow-up?Yes = downgrade, No = maintain
1e. All data taken into consideration?Yes = maintain, No = downgrade
1f. Adherence to intention to treat analysis?Yes = maintain, No = downgrade
1g. Stop early for benefit?Yes = downgrade, No = maintain
1h. Failure to report outcomes?Yes = downgrade, No = maintain
2. Inconsistency
2a. Population Heterogeneity?Yes = downgrade, No = maintain
2b. Intervention Heterogeneity?Yes = downgrade, No = maintain
2c. Outcome heterogeneity?Yes = downgrade, No = maintain
3. Indirectness
3a. Indirect comparison?Yes = downgrade, No = maintain
3b. Study population differs from target population?Yes = downgrade, No = maintain
3c. Comparator differs among studies?Yes = downgrade, No = maintain
3d. Study outcomes differ from outcomes of interest?Yes = downgrade, No = maintain
4. Imprecision
4a. Optimal information size (OIS)?Yes = maintain, No = downgrade
4b. Mean difference 95% CI includes 0?Yes = downgrade, No = maintain
4c. Mean 95% CI ranges overlap?Yes = downgrade, No = maintain
4d. Risk ratio 95% CI includes 1?Yes = downgrade, No = maintain
5. Publication bias
5a. Failure to report studies, especially those showing no effect?Yes = downgrade, No = maintain
5b. Evidence arises from small trials funded by the drug company?Yes = downgrade, No = maintain
Table 2. Drugs, ATC, NCT, and the bias (limitations) found in each study. The factor contributing to bias according to Table 1 is indicated in parenthesis.
Table 2. Drugs, ATC, NCT, and the bias (limitations) found in each study. The factor contributing to bias according to Table 1 is indicated in parenthesis.
NameAnatomical Therapeutic Chemical (ATC)Authors/National Clinical Trial (NCT)Bias (Type Based on Table 1)
Naltrexone/BupropionA08AA62Apovian et al. [26]/NCT00567255None
Halseth et al. [27]/NCT01764386Risk of bias (1a), Publication bias (5b)
LiraglutideA10BJ02O’Neil et al. [28]/NCT01272219, NCT01272232, NCT01557166, NCT00781937None
Kuhadiya et al. [29]/NCT01722266Imprecision (4a)
Garvey et al. [30]/NCT02963922Publication bias (5b)
Wadden et al. [31]/NCT02963935Publication bias (5b)
Kelly et al. [32]/NCT02918279Publication bias (5b)
Pi-Sunyer et al. [33]/NCT01272219None
Davies et al. [34]/NCT01272232Publication bias (5b)
Wadden et al./NCT02911818Risk of bias (1b), Imprecision (4a), Publication bias (5b)
LorcaserinA08AA11Aronne et al. [35]/NCT00395135, NCT00603902Risk of bias (1d)
Pi-Sunyer et al. [36]/NCT00603291Publication bias (5b)
Smith et al. [37]/NCT01987427None
SemaglutideA10BJ06Wilding et al. [38]/NCT03548935None
Ahmann et al. [39]/NCT01885208Risk of bias (1a), Publication bias (5b)
Wadden et al. [40]/NCT03611582Publication bias (5b)
Davies et al. [41]/NCT03552757None
Rubino et al. [42]/NCT04074161Risk of bias (1a), Inconsistency (2b), Publication bias (5b)
Capehorn et al. [43]/NCT03191396Risk of bias (1a), Publication bias
Rubino et al. [44]/NCT03548987Publication bias (5b)
Zinman et al. [45]/ NCT03086330Publication bias (5b)
ExenatideA10BJ01Guja et al. [46]/NCT02229383Publication bias (5b)
Jabbour et al. [47]/NCT02229396Imprecision (4a), Publication bias (5b)
Rodgers et al. [48]/NCT01590433Imprecision (4a), Publication bias (5b)
Table 3. Assessment of quality using the academic version of GRADEpro tool.
Table 3. Assessment of quality using the academic version of GRADEpro tool.
No. of StudiesRisk of BiasInconsistencyIndirectnessImprecisionRisk of Publication BiasTotal Assessment
Naltrexone/
Bupropion
2SeriousNot seriousNot seriousNot seriousStrongly
suspected
Low
Liraglutide8Not seriousNot seriousNot seriousNot seriousStrongly
suspected
Moderate
Lorcaserin3Not seriousNot seriousNot seriousNot seriousStrongly
suspected
Moderate
Semaglutide8SeriousNot seriousNot seriousNot seriousStrongly
suspected
Low
Exenatide3Not seriousNot seriousNot seriousVery seriousStrongly
suspected
Low
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Karavia, E.A.; Giannopoulou, P.C.; Konstantinopoulou, V.; Athanasopoulou, K.; Filippatos, T.D.; Panagiotakos, D.; Kypreos, K.E. Medicines for Obesity: Appraisal of Clinical Studies with Grading of Recommendations, Assessment, Development, and Evaluation Tool. Nutrients 2023, 15, 606. https://doi.org/10.3390/nu15030606

AMA Style

Karavia EA, Giannopoulou PC, Konstantinopoulou V, Athanasopoulou K, Filippatos TD, Panagiotakos D, Kypreos KE. Medicines for Obesity: Appraisal of Clinical Studies with Grading of Recommendations, Assessment, Development, and Evaluation Tool. Nutrients. 2023; 15(3):606. https://doi.org/10.3390/nu15030606

Chicago/Turabian Style

Karavia, Eleni A., Panagiota C. Giannopoulou, Vassiliki Konstantinopoulou, Katerina Athanasopoulou, Theodosios D. Filippatos, Demosthenes Panagiotakos, and Kyriakos E. Kypreos. 2023. "Medicines for Obesity: Appraisal of Clinical Studies with Grading of Recommendations, Assessment, Development, and Evaluation Tool" Nutrients 15, no. 3: 606. https://doi.org/10.3390/nu15030606

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop