A Randomized Phase III Clinical Trial of Plecanatide, a Uroguanylin Analog, in Patients With Chronic Idiopathic Constipation

Philip B Miner Jr, William D Koltun, Gregory J Wiener, Marianela De La Portilla, Blas Prieto, Kunwar Shailubhai, Mary Beth Layton, Laura Barrow, Leslie Magnus, Patrick H Griffin, Philip B Miner Jr, William D Koltun, Gregory J Wiener, Marianela De La Portilla, Blas Prieto, Kunwar Shailubhai, Mary Beth Layton, Laura Barrow, Leslie Magnus, Patrick H Griffin

Abstract

Objectives: This study assessed the efficacy and safety of plecanatide, a guanylate cyclase-C (GC-C) agonist and the first uroguanylin analog approved for the treatment of chronic idiopathic constipation (CIC).

Methods: This phase III, multicenter, double-blind, placebo-controlled study randomized 1,394 patients with CIC. Patients received either plecanatide (3 or 6 mg) or placebo, orally, once daily, for 12 weeks. The primary efficacy endpoint was the percentage of patients who were durable overall complete spontaneous bowel movement (CSBM) responders over the 12-week treatment period. Patients were instructed to record their daily bowel movements, stool consistency scores, and abdominal symptoms in an electronic diary. Treatment-emergent adverse events (AEs) were collected.

Results: Each dose of plecanatide resulted in a significantly greater percentage of durable overall CSBM responders (21.0%, 3 mg; 19.5%, 6 mg) as compared with placebo (10.2%; P<0.001 for both). Plecanatide (3 and 6 mg) also significantly increased mean weekly CSBM frequency from baseline (increase of 2.5 and 2.2/week, respectively) vs. placebo (1.2/week; P<0.001 for both) and mean weekly spontaneous bowel movement frequency (increase of 3.2 and 3.1/week, respectively) vs. placebo (1.3/week; P<0.001, for both) over the 12-week treatment period. Both plecanatide doses significantly improved all secondary and additional efficacy endpoints. The most common AE, diarrhea, occurred in 1.3% (placebo), 5.9% (3 mg) and 5.7% (6 mg) of patients.

Conclusions: Plecanatide significantly improved constipation and its related symptoms with a low rate of adverse events. These results suggest that plecanatide will be a useful treatment option in the management of CIC. ClinicalTrials.gov: NCT01982240.

Conflict of interest statement

Guarantor of the article: Leslie Magnus, MD.

Specific author contributions: Study concept: Drs Miner and Shailubhai; study design: Drs Miner, Barrow, and Griffin; data acquisition: Drs Miner, Koltun, Wiener, De La Portilla, and Prieto; data analysis: Drs Miner, Barrow, and Griffin and Ms Layton; data interpretation, drafting of the manuscript, critical revision of the manuscript for important intellectual content, and final approval of the manuscript: all authors.

Financial support: Funding for this trial and manuscript support were provided by Synergy Pharmaceuticals Inc. Synergy Pharmaceuticals Inc. also provided the plecanatide and placebo used for the study.

Potential competing interests: Drs Shailubhai, Barrow, Magnus, and Griffin and Ms Layton are employees and stockholders of Synergy Pharmaceuticals Inc. Drs Miner, Koltun, Wiener, De La Portilla, and Prieto were study investigators paid for their participation in this clinical trial. Dr Koltun has received funding for either advisory boards or speaking from the following institutions: Synergy Pharmaceuticals, Symiomix Theraputics, Ferring Pharmaceuticals, Enteris Pharmaceuticals, and Shionogi Inc. Dr Miner has received funding for advisory board meetings. Transcript Profiling: does not apply.

Figures

Figure 1
Figure 1
Disposition of the study population.
Figure 2
Figure 2
(a) Percentage of patients in each treatment group assessed as a durable overall complete spontaneous bowel movement (CSBM) responder in the intention-to-treat (ITT) population, the primary efficacy endpoint. Durable overall CSBM responders were defined as patients who fulfilled both ≥3 CSBMs per week and an increase of ≥1 CSBM from baseline, in the same week, for ≥9 of the 12 treatment weeks, including ≥3 of the last 4 weeks of treatment. Error bars represent 95% confidence intervals. (b) Weekly evolution of the percentage of CSBM responders in the ITT population. Values are LS means; bars represent 95% confidence intervals. *P=0.001, **P=0.003, †P=0.005, ‡P=0.011 vs. placebo.
Figure 3
Figure 3
(a) Changes from baseline in weekly complete spontaneous bowel movement (CSBM) frequency. Values are least squares (LS) mean; bars represent s.e. *P<0.001 vs. placebo. (b) Changes from baseline in mean weekly spontaneous bowel movement (SBM) frequency. Values are LS mean; bars represent s.e. *P<0.001 vs. placebo. (c) Percentage of patients experiencing a CSBM or SBM within 24 h after the first dose of study medication. *P<0.001 vs. placebo.
Figure 4
Figure 4
(a) Changes from baseline in weekly stool consistency. Stool consistency was measured by the Bristol Stool Form Scale (BSFS). Values are least squares (LS) mean; bars represent s.e. *P<0.001 vs. placebo. (b) Weekly BSFS scores during the study. Values are LS mean.

References

    1. Suares NC, Ford AC. Prevalence of, and risk factors for, chronic idiopathic constipation in the community: systematic review and meta-analysis. Am J Gastroenterol 2011;106:1582–1591.
    1. Higgins PD, Johanson JF. Epidemiology of constipation in North America: a systematic review. Am J Gastroenterol 2004;99:750–759.
    1. American College of Gastroenterology Chronic Constipation Task Force. An evidence-based approach to the management of chronic constipation in North America. Am J Gastroenterol 2005;100 (Suppl 1): S1–S4.
    1. Johanson JF, Kralstein J. Chronic constipation: a survey of the patient perspective. Aliment Pharmacol Ther 2007;25:599–608.
    1. Bharucha AE, Dorn SD, Lembo A et al. American Gastroenterological Association medical position statement on constipation. Gastroenterology 2013;144:211–217.
    1. Bharucha AE, Pemberton JH, Locke GR 3rd. American Gastroenterological Association technical review on constipation. Gastroenterology 2013;144:218–238.
    1. FDA Approves Linzess To Treat Certain Cases Of Irritable Bowel Syndrome And Constipation [press release]. Silver Spring, MD: US Food and Drug Administration (FDA). 2012.
    1. FDA Approves New Prescription Drug For Adults For Treatment of Chronic "Idiopathic" Constipation [press release]. Silver Spring, MD: US Food and Drug Administration (FDA). 2006.
    1. Wald A. Constipation: advances in diagnosis and treatment. JAMA 2016;315:185–191.
    1. Kita T, Smith CE, Fok KF et al. Characterization of human uroguanylin: a member of the guanylin peptide family. Am J Physiol 1994;266:F342–F348.
    1. Hamra FK, Eber SL, Chin DT et al. Regulation of intestinal uroguanylin/guanylin receptor-mediated responses by mucosal acidity. Proc Natl Acad Sci USA 1997;94:2705–2710.
    1. Shailubhai K, Comiskey S, Foss JA et al. Plecanatide, an oral guanylate cyclase C agonist acting locally in the gastrointestinal tract, is safe and well-tolerated in single doses. Dig Dis Sci 2013;58:2580–2586.
    1. Shailubhai K, Palejwala V, Arjunan KP et al. Plecanatide and dolcanatide, novel guanylate cyclase-C agonists, ameliorate gastrointestinal inflammation in experimental models of murine colitis. World J Gastrointest Pharmacol Ther 2015;6:213–222.
    1. Liu D, Overbey D, Watkinson LD et al. In vivo imaging of human colorectal cancer using radiolabeled analogs of the uroguanylin peptide hormone. Anticancer Res 2009;29:3777–3783.
    1. Vaandrager AB, Smolenski A, Tilly BC et al. Membrane targeting of cGMP-dependent protein kinase is required for cystic fibrosis transmembrane conductance regulator Cl- channel activation. Proc Natl Acad Sci USA 1998;95:1466–1471.
    1. Toriano R, Ozu M, Politi MT et al. Uroguanylin regulates net fluid secretion via the NHE2 isoform of the Na+/H+ exchanger in an intestinal cellular model. Cell Physiol Biochem 2011;28:733–742.
    1. Busby RW, Bryant AP, Bartolini WP et al. Linaclotide, through activation of guanylate cyclase C, acts locally in the gastrointestinal tract to elicit enhanced intestinal secretion and transit. Eur J Pharmacol 2010;649:328–335.
    1. Brierley SM. Guanylate cyclase-C receptor activation: unexpected biology. Curr Opin Pharmacol 2012;12:632–640.
    1. Field M, Graf LH Jr, Laird WJ et al. Heat-stable enterotoxin of Escherichia coli: in vitro effects on guanylate cyclase activity, cyclic GMP concentration, and ion transport in small intestine. Proc Natl Acad Sci USA 1978;75:2800–2804.
    1. Shailubhai K. Therapeutic applications of guanylate cyclase-C receptor agonists. Curr Opin Drug Discov Devel 2002;5:261–268.
    1. Sindic A. Current understanding of guanylin peptides actions. ISRN Nephrol 2013;2013:813648.
    1. Thayalasekeran S, Ali H, Tsai HH. Novel therapies for constipation. World J Gastroenterol 2013;19:8247–8251.
    1. Castro J, Harrington AM, Hughes PA et al. Linaclotide inhibits colonic nociceptors and relieves abdominal pain via guanylate cyclase-C and extracellular cyclic guanosine 3′,5′-monophosphate. Gastroenterology 2013;145:1334-46.e1–11.
    1. Miner PB, Surowitz R, Fogel R et al. 925g Plecanatide, a novel guanylate cyclase-C (GC-C) receptor agonist, is efficacious and safe in patients with chronic idiopathic constipation (CIC): results from a 951 patient, 12 week, multi-center trial [abstract]. Gastroenterology 2013;144:S-163.
    1. Drossman DA. The functional gastrointestinal disorders and the Rome III process. Gastroenterology 2006;130:1377–1390.
    1. Study SP304-20210: A Randomized, 12-Week, Double-Blind, Placebo-Controlled, Repeat-Dose, Oral, Dose-Ranging Study to Assess the Safety and Efficacy of Plecanatide in Patients With Chronic Idiopathic Constipation. In: [Internet]. Bethesda (MD): National Library of Medicine (US). 2012, Accessed on 25 August 2015. Available at .

Source: PubMed

3
Sottoscrivi