Effect of Continued Weekly Subcutaneous Semaglutide vs Placebo on Weight Loss Maintenance in Adults With Overweight or Obesity: The STEP 4 Randomized Clinical Trial

Domenica Rubino, Niclas Abrahamsson, Melanie Davies, Dan Hesse, Frank L Greenway, Camilla Jensen, Ildiko Lingvay, Ofri Mosenzon, Julio Rosenstock, Miguel A Rubio, Gottfried Rudofsky, Sayeh Tadayon, Thomas A Wadden, Dror Dicker, STEP 4 Investigators, Mette Friberg, Anders Sjödin, Dror Dicker, Gabriella Segal, Ofri Mosenzon, Muhammad Sabbah, Yael Sofer, Victor Vishlitzky, Eelco W Meesters, Mirelle Serlie, Arianne van Bon, Helena Cardoso, Paula Freitas, Pedro Carneiro de Melo, Margarida Monteiro, Mariana Monteiro, Dírcea Rodrigues, Aysha Badat, Pankaj Joshi, Gulam Latiff, Essack A Mitha, Hans H Snyman, Elane van Nieuwenhuizen, Olga González Albarrán, Assumpta Caixas, Carmen de Al Cuesta, Pedro Pablo Garcia Luna, Cristobal Morales Portillo, Pedro Mezquita Raya, Miguel Angel Rubio, Niclas Abrahamsson, Johan Hoffstedt, Fredrik von Wowern, Erik Uddman, Birgit Bach-Kliegel, Felix Beuschlein, Stefan Bilz, Alain Golay, Gottfried Rudofsky, Christopher Strey, Galyna Fadieienko, Nataliia Kosei, Tetiana Tatarchuk, Valentyna Velychko, Olesya Zinych, Stephen L Aronoff, Harold E Bays, Andrew P Brockmyre, Robert S Call, Charles Crump, Cyrus V Desouza, Valerie Espinosa, Almena L Free, Winston H Gandy, Steven A Geller, Gregory M Gottschlich, Frank L Greenway, Laurie Han-Conrad, Wayne Harper, Lee Herman, Mitzie Hewitt, Priscilla Hollander, Steven R Kaster, Anastasios Manessis, Frederick A Martin, Robert E McNeill, Alexander V Murray, Paul C Norwood, John C H Reed, Julio Rosenstock, Domenica M Rubino, Martin J Schear, Mark L Warren, Domenica Rubino, Niclas Abrahamsson, Melanie Davies, Dan Hesse, Frank L Greenway, Camilla Jensen, Ildiko Lingvay, Ofri Mosenzon, Julio Rosenstock, Miguel A Rubio, Gottfried Rudofsky, Sayeh Tadayon, Thomas A Wadden, Dror Dicker, STEP 4 Investigators, Mette Friberg, Anders Sjödin, Dror Dicker, Gabriella Segal, Ofri Mosenzon, Muhammad Sabbah, Yael Sofer, Victor Vishlitzky, Eelco W Meesters, Mirelle Serlie, Arianne van Bon, Helena Cardoso, Paula Freitas, Pedro Carneiro de Melo, Margarida Monteiro, Mariana Monteiro, Dírcea Rodrigues, Aysha Badat, Pankaj Joshi, Gulam Latiff, Essack A Mitha, Hans H Snyman, Elane van Nieuwenhuizen, Olga González Albarrán, Assumpta Caixas, Carmen de Al Cuesta, Pedro Pablo Garcia Luna, Cristobal Morales Portillo, Pedro Mezquita Raya, Miguel Angel Rubio, Niclas Abrahamsson, Johan Hoffstedt, Fredrik von Wowern, Erik Uddman, Birgit Bach-Kliegel, Felix Beuschlein, Stefan Bilz, Alain Golay, Gottfried Rudofsky, Christopher Strey, Galyna Fadieienko, Nataliia Kosei, Tetiana Tatarchuk, Valentyna Velychko, Olesya Zinych, Stephen L Aronoff, Harold E Bays, Andrew P Brockmyre, Robert S Call, Charles Crump, Cyrus V Desouza, Valerie Espinosa, Almena L Free, Winston H Gandy, Steven A Geller, Gregory M Gottschlich, Frank L Greenway, Laurie Han-Conrad, Wayne Harper, Lee Herman, Mitzie Hewitt, Priscilla Hollander, Steven R Kaster, Anastasios Manessis, Frederick A Martin, Robert E McNeill, Alexander V Murray, Paul C Norwood, John C H Reed, Julio Rosenstock, Domenica M Rubino, Martin J Schear, Mark L Warren

Abstract

Importance: The effect of continuing vs withdrawing treatment with semaglutide, a glucagon-like peptide 1 receptor agonist, on weight loss maintenance in people with overweight or obesity is unknown.

Objective: To compare continued once-weekly treatment with subcutaneous semaglutide, 2.4 mg, with switch to placebo for weight maintenance (both with lifestyle intervention) in adults with overweight or obesity after a 20-week run-in with subcutaneous semaglutide titrated to 2.4 mg weekly.

Design, setting, and participants: Randomized, double-blind, 68-week phase 3a withdrawal study conducted at 73 sites in 10 countries from June 2018 to March 2020 in adults with body mass index of at least 30 (or ≥27 with ≥1 weight-related comorbidity) and without diabetes.

Interventions: A total of 902 participants received once-weekly subcutaneous semaglutide during run-in. After 20 weeks (16 weeks of dose escalation; 4 weeks of maintenance dose), 803 participants (89.0%) who reached the 2.4-mg/wk semaglutide maintenance dose were randomized (2:1) to 48 weeks of continued subcutaneous semaglutide (n = 535) or switched to placebo (n = 268), plus lifestyle intervention in both groups.

Main outcomes and measures: The primary end point was percent change in body weight from week 20 to week 68; confirmatory secondary end points were changes in waist circumference, systolic blood pressure, and physical functioning (assessed using the Short Form 36 Version 2 Health Survey, Acute Version [SF-36]).

Results: Among 803 study participants who completed the 20-week run-in period (with a mean weight loss of 10.6%) and were randomized (mean age, 46 [SD, 12] years; 634 [79%] women; mean body weight, 107.2 kg [SD, 22.7 kg]), 787 participants (98.0%) completed the trial and 741 (92.3%) completed treatment. With continued semaglutide, mean body weight change from week 20 to week 68 was -7.9% vs +6.9% with the switch to placebo (difference, -14.8 [95% CI, -16.0 to -13.5] percentage points; P < .001). Waist circumference (-9.7 cm [95% CI, -10.9 to -8.5 cm]), systolic blood pressure (-3.9 mm Hg [95% CI, -5.8 to -2.0 mm Hg]), and SF-36 physical functioning score (2.5 [95% CI, 1.6-3.3]) also improved with continued subcutaneous semaglutide vs placebo (all P < .001). Gastrointestinal events were reported in 49.1% of participants who continued subcutaneous semaglutide vs 26.1% with placebo; similar proportions discontinued treatment because of adverse events with continued semaglutide (2.4%) and placebo (2.2%).

Conclusions and relevance: Among adults with overweight or obesity who completed a 20-week run-in period with subcutaneous semaglutide, 2.4 mg once weekly, maintaining treatment with semaglutide compared with switching to placebo resulted in continued weight loss over the following 48 weeks.

Trial registration: ClinicalTrials.gov Identifier: NCT03548987.

Conflict of interest statement

Conflict of Interest Disclosures: Dr Rubino reported being a clinical investigator for Boehringer Ingelheim and AstraZeneca and receiving speaker fees, consulting fees, and honoraria from Novo Nordisk and being a shareholder in Novo Nordisk. Dr Davies reported receipt of consultant, advisory board member, and speaker fees from Novo Nordisk, Sanofi, Eli Lilly, and Boehringer Ingelheim; advisory board member and speaker fees from AstraZeneca; advisory board member fees from Gilead Sciences, Janssen, and Lexicon; speaker fees from Napp Pharmaceuticals and Takeda Pharmaceuticals International, and grants from AstraZeneca, Novo Nordisk, Boehringer Ingelheim, Janssen, and Sanofi. Dr Hesse reported receipt of personal fees from Novo Nordisk. Dr Greenway reported receipt of grants from Pennington Biomedical Research Center and NuSirt to his institution; research funding from NovMeta Pharma and Melior Discoveries; receipt of consulting fees from Basic Research, Dr. Reddy’s Laboratories, Jazz Pharmaceuticals, General Nutrition Corporation, and Regeneron Pharmaceuticals; receipt of scientific advisory board fees from Jenny Craig and Pfizer; and stock ownership in Academic Technology Ventures, Ketogenic Health Systems, Plensat, UR Labs, and Rejuvenate Bio. In addition, Dr Greenway has a patent issued for orlistat and a patent pending for pramlintide/albuterol. Ms Jensen reported receipt of personal fees from Novo Nordisk. Dr Lingvay reported receipt of grants, personal fees, and nonfinancial support from Novo Nordisk and Sanofi; personal fees and nonfinancial support from Eli Lilly, AstraZeneca, and Boehringer Ingelheim; personal fees from Janssen, Intercept, Intarcia, TARGETPharma, Mannkind, Valeritas, Bayer, and Zealand Pharma; grants and nonfinancial support from Merck and Pfizer; and grants from Mylan. Dr Mosenzon reported receipt of grants from Novo Nordisk and AstraZeneca through Hadassah Medical Center; advisory board and speaker’s bureau fees from Novo Nordisk, AstraZeneca, Eli Lilly, Merck Sharp & Dohme, and Sanofi; speaker’s bureau fees from Boehringer Ingelheim and Janssen; and advisory board fees from BOL Pharma. Dr Rosenstock reported receipt of scientific advisory board fees, honoraria, consulting fees, and grants/research support from Novo Nordisk, Applied Therapeutics, Boehringer Ingelheim, Eli Lilly, Intarcia, Oramed, and Sanofi; honoraria or consulting fees from Zealand; and grants/research support from Genentech, Novartis, Pfizer, REMD Biotherapeutics, and vTv Therapeutics. Dr Rubio reported receipt of personal fees from Novo Nordisk. Dr Tadayon reported being a full-time employee of and shareholder in Novo Nordisk. Dr Wadden reported receipt of grants from Novo Nordisk received on behalf of the University of Pennsylvania and scientific advisory board fees from Novo Nordisk and WW (formerly Weight Watchers). Dr Dicker reported receipt of personal fees, nonfinancial support, and grants from Novo Nordisk and grants from Eli Lilly. No other disclosures were reported.

Figures

Figure 1.. Participant Flow in the Semaglutide…
Figure 1.. Participant Flow in the Semaglutide Treatment Effect in People With Obesity (STEP) 4 Trial
aThese participants received once-weekly semaglutide (safety analysis set). bRun-in failures were defined as participants not meeting all 3 randomization criteria: attend the randomization visit, reach the semaglutide maintenance dose of 2.4 mg by week 16 (±3 days), and be receiving semaglutide, 2.4 mg, at week 20. cOther reasons are listed in eTable 9 in Supplement 1. dOther reasons are listed in eTable 10 in Supplement 1.
Figure 2.. Effect of Semaglutide, 2.4 mg…
Figure 2.. Effect of Semaglutide, 2.4 mg Once Weekly, Compared With Placebo on Efficacy Outcomes During the Entire Trial (Full Analysis Set)
Data presented in panels A, B, and C are observed data for the full analysis set from the in-trial period (the time from week 0 to the date of last contact with trial site). Error bars represent 95% confidence intervals for the mean. Participant numbers shown denote those contributing to the mean. The dashed vertical line at week 20 represents the randomization time point. Data in the shaded area on the right in panel C are estimated mean changes from week 0 to week 68 for the treatment policy estimand, analyzed using the full analysis set. (The treatment policy estimand assessed the treatment effect regardless of treatment discontinuation or rescue intervention using analysis of covariance, with randomized treatment as a factor and baseline end point value as a covariate, and a multiple imputation approach for missing data.) Data in panel D are observed data among all randomized participants with a week 68 assessment from the in-trial period (the time from week 0 to the date of last contact with trial site).

References

    1. Frühbeck G, Busetto L, Dicker D, et al. . The ABCD of obesity: an EASO position statement on a diagnostic term with clinical and scientific implications. Obes Facts. 2019;12(2):131-136. doi:10.1159/000497124
    1. Leibel RL, Seeley RJ, Darsow T, Berg EG, Smith SR, Ratner R. Biologic responses to weight loss and weight regain: report from an American Diabetes Association research symposium. Diabetes. 2015;64(7):2299-2309. doi:10.2337/db15-0004
    1. Lemstra M, Bird Y, Nwankwo C, Rogers M, Moraros J. Weight loss intervention adherence and factors promoting adherence: a meta-analysis. Patient Prefer Adherence. 2016;10:1547-1559. doi:10.2147/PPA.S103649
    1. Kroeger CM, Hoddy KK, Varady KA. Impact of weight regain on metabolic disease risk: a review of human trials. J Obes. 2014;2014:614519. doi:10.1155/2014/614519
    1. Garvey WT, Mechanick JI, Brett EM, et al. ; Reviewers of the AACE/ACE Obesity Clinical Practice Guidelines . American Association of Clinical Endocrinologists and American College of Endocrinology comprehensive clinical practice guidelines for medical care of patients with obesity. Endocr Pract. 2016;22(suppl 3):1-203. doi:10.4158/
    1. Yumuk V, Tsigos C, Fried M, et al. ; Obesity Management Task Force of the European Association for the Study of Obesity . European guidelines for obesity management in adults. Obes Facts. 2015;8(6):402-424. doi:10.1159/000442721
    1. Bray GA, Frühbeck G, Ryan DH, Wilding JP. Management of obesity. Lancet. 2016;387(10031):1947-1956. doi:10.1016/S0140-6736(16)00271-3
    1. Bessesen DH, Van Gaal LF. Progress and challenges in anti-obesity pharmacotherapy. Lancet Diabetes Endocrinol. 2018;6(3):237-248. doi:10.1016/S2213-8587(17)30236-X
    1. Khera R, Murad MH, Chandar AK, et al. . Association of pharmacological treatments for obesity with weight loss and adverse events: a systematic review and meta-analysis. JAMA. 2016;315(22):2424-2434. doi:10.1001/jama.2016.7602
    1. Kushner RF, Calanna S, Davies M, et al. . Semaglutide 2.4 mg for the treatment of obesity: key elements of the STEP trials 1 to 5. Obesity (Silver Spring). 2020;28(6):1050-1061. doi:10.1002/oby.22794
    1. US Food and Drug Administration . Ozempic (semaglutide): prescribing information. Revised January 2020. Accessed July 22, 2020.
    1. Friedrichsen M, Breitschaft A, Tadayon S, Wizert A, Skovgaard D. The effect of semaglutide 2.4 mg once weekly on energy intake, appetite, control of eating, and gastric emptying in adults with obesity. Diabetes Obes Metab. 2021;23(3):754-762. doi:10.1111/dom.14280
    1. Gabery S, Salinas CG, Paulsen SJ, et al. . Semaglutide lowers body weight in rodents via distributed neural pathways. JCI Insight. 2020;5(6):e133429. doi:10.1172/jci.insight.133429
    1. O’Neil PM, Birkenfeld AL, McGowan B, et al. . Efficacy and safety of semaglutide compared with liraglutide and placebo for weight loss in patients with obesity: a randomised, double-blind, placebo and active controlled, dose-ranging, phase 2 trial. Lancet. 2018;392(10148):637-649. doi:10.1016/S0140-6736(18)31773-2
    1. Lingvay I, Desouza CV, Lalic KS, et al. . A 26-week randomized controlled trial of semaglutide once daily versus liraglutide and placebo in patients with type 2 diabetes suboptimally controlled on diet and exercise with or without metformin. Diabetes Care. 2018;41(9):1926-1937. doi:10.2337/dc17-2381
    1. World Medical Association . World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191-2194.
    1. US Food and Drug Administration . ICH E9 (R1): Statistical Principles for Clinical Trials: Addendum: Estimands and Sensitivity Analysis in Clinical Trials. Published October 2017. Accessed November 18, 2020.
    1. Aroda VR, Saugstrup T, Buse JB, Donsmark M, Zacho J, Davies MJ. Incorporating and interpreting regulatory guidance on estimands in diabetes clinical trials: the PIONEER 1 randomized clinical trial as an example. Diabetes Obes Metab. 2019;21(10):2203-2210. doi:10.1111/dom.13804
    1. International Council for Harmonisation . Addendum on Estimands and Sensitivity Analysis in Clinical Trials To The Guideline on Statistical Principles for Clinical Trials E9I. Accessed October 27, 2020.
    1. Wharton S, Astrup A, Endahl L, et al. . Estimating and reporting treatment effects in clinical trials for weight management: using estimands to interpret effects of intercurrent events and missing data. Int J Obes (Lond). 2021. doi:10.1038/s41366-020-00733-x
    1. McEvoy BW. Missing data in clinical trials for weight management. J Biopharm Stat. 2016;26(1):30-36. doi:10.1080/10543406.2015.1094814
    1. Little RJA, Rubin DB. Statistical Analysis With Missing Data. John Wiley & Sons; 1987.
    1. Sjöström L, Rissanen A, Andersen T, et al. ; European Multicentre Orlistat Study Group . Randomised placebo-controlled trial of orlistat for weight loss and prevention of weight regain in obese patients. Lancet. 1998;352(9123):167-172. doi:10.1016/S0140-6736(97)11509-4
    1. Smith SR, Weissman NJ, Anderson CM, et al. ; Behavioral Modification and Lorcaserin for Overweight and Obesity Management Study Group . Multicenter, placebo-controlled trial of lorcaserin for weight management. N Engl J Med. 2010;363(3):245-256. doi:10.1056/NEJMoa0909809
    1. Courcoulas AP, Christian NJ, Belle SH, et al. ; Longitudinal Assessment of Bariatric Surgery Consortium . Weight change and health outcomes at 3 years after bariatric surgery among individuals with severe obesity. JAMA. 2013;310(22):2416-2425. doi:10.1001/jama.2013.280928
    1. Singh M, Lee J, Gupta N, et al. . Weight loss can lead to resolution of gastroesophageal reflux disease symptoms: a prospective intervention trial. Obesity (Silver Spring). 2013;21(2):284-290. doi:10.1002/oby.20279
    1. Arterburn DE, Telem DA, Kushner RF, Courcoulas AP. Benefits and risks of bariatric surgery in adults: a review. JAMA. 2020;324(9):879-887. doi:10.1001/jama.2020.12567
    1. Lean MEJ, Leslie WS, Barnes AC, et al. . Primary care-led weight management for remission of type 2 diabetes (DiRECT): an open-label, cluster-randomised trial. Lancet. 2018;391(10120):541-551. doi:10.1016/S0140-6736(17)33102-1
    1. Apovian CM, Garvey WT, Ryan DH. Challenging obesity: patient, provider, and expert perspectives on the roles of available and emerging nonsurgical therapies. Obesity (Silver Spring). 2015;23(suppl 2):S1-S26. doi:10.1002/oby.21140
    1. Pi-Sunyer X, Astrup A, Fujioka K, et al. ; SCALE Obesity and Prediabetes NN8022-1839 Study Group . A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N Engl J Med. 2015;373(1):11-22. doi:10.1056/NEJMoa1411892
    1. Wadden TA, Hollander P, Klein S, et al. ; NN8022-1923 Investigators . Weight maintenance and additional weight loss with liraglutide after low-calorie-diet-induced weight loss: the SCALE Maintenance randomized study. Int J Obes (Lond). 2013;37(11):1443-1451. doi:10.1038/ijo.2013.120
    1. Hellström PM. GLP-1 analogue liraglutide as adjunct treatment in diabetes type 2 after failed bariatric/metabolic surgery. Ann Transl Med. 2019;7(suppl 6):S240. doi:10.21037/atm.2019.08.94
    1. Schauer PR, Kashyap SR, Wolski K, et al. . Bariatric surgery versus intensive medical therapy in obese patients with diabetes. N Engl J Med. 2012;366(17):1567-1576. doi:10.1056/NEJMoa1200225
    1. Aroda VR, Ahmann A, Cariou B, et al. . Comparative efficacy, safety, and cardiovascular outcomes with once-weekly subcutaneous semaglutide in the treatment of type 2 diabetes: insights from the SUSTAIN 1-7 trials. Diabetes Metab. 2019;45(5):409-418. doi:10.1016/j.diabet.2018.12.001
    1. Trujillo J. Safety and tolerability of once-weekly GLP-1 receptor agonists in type 2 diabetes. J Clin Pharm Ther. 2020;45(suppl 1):43-60. doi:10.1111/jcpt.13225
    1. US Food and Drug Administration . Enrichment Strategies for Clinical Trials to Support Determination of Effectiveness of Human Drugs and Biological Products: Guidance for Industry. Published March 2019. Accessed July 22, 2020.

Source: PubMed

3
Subscribe