The efficacy and safety of setmelanotide in individuals with Bardet-Biedl syndrome or Alström syndrome: Phase 3 trial design

Robert M Haws, Gregory Gordon, Joan C Han, Jack A Yanovski, Guojun Yuan, Murray W Stewart, Robert M Haws, Gregory Gordon, Joan C Han, Jack A Yanovski, Guojun Yuan, Murray W Stewart

Abstract

Background: A phase 2 trial has suggested that treatment with the melanocortin-4 receptor (MC4R) agonist setmelanotide is associated with a decrease in hunger and weight-related outcomes in participants with Bardet-Biedl syndrome (BBS) and Alström syndrome. Here, we present the study design of an ongoing, randomized, double-blind, placebo-controlled, phase 3 trial to assess the long-term efficacy and safety of setmelanotide for the treatment of obesity and hyperphagia in individuals with BBS or Alström syndrome (ClinicalTrials.gov identifier: NCT03746522).

Methods: It was initially planned that ~30 participants aged ≥6 years with a clinical diagnosis of BBS or Alström syndrome would be enrolled. Participants with obesity as defined by a body mass index ≥30 kg/m2 (in those aged ≥16 years) or a weight >97th percentile (in those aged 6-15 years) are included. Participants are initially randomized in a 1:1 ratio to receive setmelanotide or placebo for 14 weeks (period 1). Following period 1, all participants receive 38 weeks of open-label treatment with setmelanotide (period 2). In each treatment period, setmelanotide is administered at 3 mg once a day following completion of dose escalation. The primary endpoint is the proportion of participants aged ≥12 years achieving a clinically meaningful reduction from baseline (≥10%) in body weight after ~52 weeks (eg, following period 2). Safety and tolerability are assessed by frequency of adverse events.

Conclusions: This pivotal trial is designed to evaluate the efficacy and safety of setmelanotide for the treatment of obesity and hyperphagia in individuals with BBS and Alström syndrome.

Submission category: Study Design, Statistical Design, Study Protocols.

Keywords: Antiobesity drug; Appetite control; Obesity therapy; Phase III study.

Conflict of interest statement

RMH is a consultant for Rhythm Pharmaceuticals, Inc and Trinity Life Sciences and receives grant funding from the Bardet-Biedl Syndrome Foundation. GG, GY, and MWS are employed by and may own stock in Rhythm Pharmaceuticals, Inc. JCH has received grant support for clinical investigations from the Memphis Research Consortium, Le Bonheur Children's Foundation Research Institute, and Rhythm Pharmaceuticals, Inc. JAY receives grant support for clinical investigations from the NICHD, NIH, Soleno Therapeutics Inc, and Rhythm Pharmaceuticals, Inc.

© 2021 The Authors. Published by Elsevier Inc.

Figures

Fig. 1
Fig. 1
Schematic for the design of the overall study. Escal., escalation; QD, once a day; SET, setmelanotide. aDuring dose escalation, participants who are ≥16 years of age receive setmelanotide 2 mg QD for 2 weeks, which increases to 3 mg at the beginning of week 3; participants who are <16 years of age receive setmelanotide 1 mg QD for the first week, 2 mg for the second week, and 3 mg at the beginning of week 3.

References

    1. Yazdi F.T., Clee S.M., Meyre D. Obesity genetics in mouse and human: back and forth, and back again. PeerJ. 2015;3:e856. doi: 10.7717/peerj.856.
    1. Huvenne H., Dubern B., Clement K., Poitou C. Rare genetic forms of obesity: clinical approach and current treatments in 2016. Obesity Facts. 2016;9(3):158–173. doi: 10.1159/000445061.
    1. Sohn J.W. Network of hypothalamic neurons that control appetite. BMB Rep. 2015;48(4):229–233.
    1. Seo S., Guo D.F., Bugge K., Morgan D.A., Rahmouni K., Sheffield V.C. Requirement of Bardet-Biedl syndrome proteins for leptin receptor signaling. Hum. Mol. Genet. 2009;18(7):1323–1331. doi: 10.1093/hmg/ddp031.
    1. Davenport J.R., Watts A.J., Roper V.C., Croyle M.J., van Groen T., Wyss J.M., Nagy T.R., Kesterson R.A., Yoder B.K. Disruption of intraflagellar transport in adult mice leads to obesity and slow-onset cystic kidney disease. Curr. Biol. 2007;17(18):1586–1594. doi: 10.1016/j.cub.2007.08.034.
    1. Hearn T., Spalluto C., Phillips V.J., Renforth G.L., Copin N., Hanley N.A., Wilson D.I. Subcellular localization of ALMS1 supports involvement of centrosome and basal body dysfunction in the pathogenesis of obesity, insulin resistance, and type 2 diabetes. Diabetes. 2005;54(5):1581–1587. doi: 10.2337/diabetes.54.5.1581.
    1. Li G., Vega R., Nelms K., Gekakis N., Goodnow C., McNamara P., Wu H., Hong N.A., Glynne R. A role for Alstrom syndrome protein, alms1, in kidney ciliogenesis and cellular quiescence. PLoS Genet. 2007;3(1):e8. doi: 10.1371/journal.pgen.0030008.
    1. Beales P.L., Elcioglu N., Woolf A.S., Parker D., Flinter F.A. New criteria for improved diagnosis of Bardet-Biedl syndrome: results of a population survey. J. Med. Genet. 1999;36(6):437–446.
    1. Marshall J.D., Beck S., Maffei P., Naggert J.K. Alstrom syndrome. Eur. J. Hum. Genet. 2007;15(12):1193–1202. doi: 10.1038/sj.ejhg.5201933.
    1. Feuillan P.P., Ng D., Han J.C., Sapp J.C., Wetsch K., Spaulding E., Zheng Y.C., Caruso R.C., Brooks B.P., Johnston J.J., Yanovski J.A., Biesecker L.G. Patients with Bardet-Biedl syndrome have hyperleptinemia suggestive of leptin resistance. J. Clin. Endocrinol. Metab. 2011;96(3):E528–E535. doi: 10.1210/jc.2010-2290.
    1. Sherafat-Kazemzadeh R., Ivey L., Kahn S.R., Sapp J.C., Hicks M.D., Kim R.C., Krause A.J., Shomaker L.B., Biesecker L.G., Han J.C., Yanovski J.A. Hyperphagia among patients with Bardet-Biedl syndrome. Pediatr. Obes. 2013;8(5):e64–e67. doi: 10.1111/j.2047-6310.2013.00182.x.
    1. Han J.C., Reyes-Capo D.P., Liu C.Y., Reynolds J.C., Turkbey E., Turkbey I.B., Bryant J., Marshall J.D., Naggert J.K., Gahl W.A., Yanovski J.A., Gunay-Aygun M. Comprehensive endocrine-metabolic evaluation of patients with Alström syndrome compared with BMI-matched controls. J. Clin. Endocrinol. Metab. 2018;103(7):2707–2719. doi: 10.1210/jc.2018-00496.
    1. Hicks M.D., Marshall J.D., Maffei P., Hanish A.E., Hunter L.A., Brady S.M., Sedaka N.M., Sherafat Kazemzadeh R., Tsao J.W., Milan G., Naggert J., Yanovski J.A., Han J.C. 8 November 2012. Hyperphagia, Leptin, and Brain-Derived Neurotrophic Factor in Subjects with Alström Syndrome and BMI-Z Matched Controls (Poster 2702T), Poster Presented at the Annual Scientific Meeting of the American Society for Human Genetics. San Francisco, CA.
    1. Clement K., Biebermann H., Farooqi I.S., Van der Ploeg L., Wolters B., Poitou C., Puder L., Fiedorek F., Gottesdiener K., Kleinau G., Heyder N., Scheerer P., Blume-Peytavi U., Jahnke I., Sharma S., Mokrosinski J., Wiegand S., Muller A., Weiss K., Mai K., Spranger J., Gruters A., Blankenstein O., Krude H., Kuhnen P. MC4R agonism promotes durable weight loss in patients with leptin receptor deficiency. Nat. Med. 2018;24(5):551–555. doi: 10.1038/s41591-018-0015-9.
    1. Kühnen P., Clement K., Wiegand S., Blankenstein O., Gottesdiener K., Martini L.L., Mai K., Blume-Peytavi U., Gruters A., Krude H. Proopiomelanocortin deficiency treated with a melanocortin-4 receptor agonist. N. Engl. J. Med. 2016;375(3):240–246. doi: 10.1056/NEJMoa1512693.
    1. Haws R., Stewart M., Han J.C. vol. 10. Banff; Alberta, Canada: February 2019. (Clinical Study Experience of the MC4R Agonist Setmelanotide in the Treatment of Rare Genetic Disorders of Obesity: Results from Bardet-Biedl Syndrome and Alström Syndrome Cohorts in a Phase 2 Open-Label Study, Poster Presented at the Keystone Symposia on Molecular and Cellular Biology 2019).
    1. Haws R., Brady S., Davis E., Fletty K., Yuan G., Gordon G., Stewart M., Yanovski J. Effect of setmelanotide, a melanocortin-4 receptor agonist, on obesity in Bardet-Biedl syndrome. Diabetes Obes. Metabol. 2020 doi: 10.1111/dom.14133.
    1. Castro-Sánchez S., Álvarez-Satta M., Valverde D. Bardet-Biedl syndrome: a rare genetic disease. J. Pediatr. Genet. 2013;2(2):77–83.
    1. Rath A., Salamon V., Peixoto S., Hivert V., Laville M., Segrestin B., Neugebauer E.A.M., Eikermann M., Bertele V., Garattini S., Wetterslev J., Banzi R., Jakobsen J.C., Djurisic S., Kubiak C., Demotes-Mainard J., Gluud C. A systematic literature review of evidence-based clinical practice for rare diseases: what are the perceived and real barriers for improving the evidence and how can they be overcome? Trials. 2017;18(1):556. doi: 10.1186/s13063-017-2287-7.
    1. Augustine E.F., Adams H.R., Mink J.W. Clinical trials in rare disease: challenges and opportunities. J. Child Neurol. 2013;28(9):1142–1150. doi: 10.1177/0883073813495959.
    1. Day S., Jonker A.H., Lau L.P.L., Hilgers R.D., Irony I., Larsson K., Roes K.C., Stallard N. Recommendations for the design of small population clinical trials. Orphanet J. Rare Dis. 2018;13(1):195. doi: 10.1186/s13023-018-0931-2.
    1. Sriram P. Beyond placebo: alternative options to the randomized control trial design in rare disease studies. Clin. Trial Pract. Open J. 2020;1(1):42–45.
    1. Dykens E.M., Miller J., Angulo M., Roof E., Reidy M., Hatoum H.T., Willey R., Bolton G., Korner P. Intranasal carbetocin reduces hyperphagia in individuals with Prader-Willi syndrome. JCI Insight. 2018;3(12) doi: 10.1172/jci.insight.98333.
    1. Heymsfield S.B., Avena N.M., Baier L., Brantley P., Bray G.A., Burnett L.C., Butler M.G., Driscoll D.J., Egli D., Elmquist J., Forster J.L., Goldstone A.P., Gourash L.M., Greenway F.L., Han J.C., Kane J.G., Leibel R.L., Loos R.J., Scheimann A.O., Roth C.L., Seeley R.J., Sheffield V., Tauber M., Vaisse C., Wang L., Waterland R.A., Wevrick R., Yanovski J.A., Zinn A.R. Hyperphagia: current concepts and future directions proceedings of the 2nd international conference on hyperphagia. Obesity. 2014;22(Suppl 1):S1–s17. doi: 10.1002/oby.20646.
    1. Forsyth Rl R.L., Gunay-Aygun M. Bardet-Biedl syndrome overview. In: Adam M.P., Ardinger H.H., Pagon R.A., Wallace S.E., editors. GeneReviews®. University of Washington, Seattle; 2020. NBK1363.

Source: PubMed

3
購読する