Efficacy of Tenapanor in Treating Patients With Irritable Bowel Syndrome With Constipation: A 12-Week, Placebo-Controlled Phase 3 Trial (T3MPO-1)

William D Chey, Anthony J Lembo, David P Rosenbaum, William D Chey, Anthony J Lembo, David P Rosenbaum

Abstract

Objectives: Tenapanor is a first-in-class, minimally absorbed, small-molecule inhibitor of the gastrointestinal sodium/hydrogen exchanger isoform 3. This phase 3 trial assessed the efficacy and safety of tenapanor 50 mg b.i.d. for the treatment of patients with constipation-predominant irritable bowel syndrome (IBS-C).

Methods: In this phase 3, double-blind study (ClinicalTrials.gov identifier NCT02621892), patients with IBS-C were randomized to tenapanor 50 mg b.i.d. or placebo b.i.d. for 12 weeks followed by a 4-week randomized withdrawal period. The primary efficacy variable was the proportion of patients who reported a reduction in average weekly worst abdominal pain of ≥30.0% and an increase of ≥1 complete spontaneous bowel movement from baseline, both in the same week, for ≥6 weeks of the 12-week treatment period.

Results: Of the 629 randomized patients with IBS-C, 606 (96.3%) were included in the intention-to-treat analysis set (tenapanor: n = 307; placebo: n = 299) and 533 (84.7%) completed the 12-week treatment period. In the intention-to-treat analysis set (mean age 45 years, 81.4% women), a significantly greater proportion of patients treated with tenapanor met the primary endpoint than patients treated with placebo (27.0% vs 18.7%, P = 0.020). Abdominal symptoms and global symptoms of IBS also improved with tenapanor (P < 0.05 vs placebo). Diarrhea was the most commonly reported adverse event, resulting in study drug discontinuation in 6.5% and 0.7% of patients receiving tenapanor and placebo, respectively, during the 12-week treatment period.

Discussion: Tenapanor 50 mg b.i.d. improved IBS-C symptoms and was generally well tolerated, offering a potential new treatment option for patients with IBS-C.

Conflict of interest statement

Guarantor of the article: William D. Chey, MD, AGAF, FACG, FACP, RFF.

Specific author contributions: W.D.C. and A.J.L. contributed to the planning of the study, interpretation of the data, and critical revision of the manuscript for important intellectual content; D.P.R. contributed to the planning of the study, conduct of the study, interpretation of the data, and critical revision of the manuscript for important intellectual content; all authors approved the final version of the manuscript for submission.

Financial support: Medical writing support was provided by Svetha Sankar, BSc BVMS, and Steven Inglis, PhD, of Oxford PharmaGenesis and was funded by Ardelyx.

Potential competing interests: W.D.C. is a consultant for Allergan, Biomerica, IM Health, Ironwood, Outpost, Prometheus, QOL Medical, Ritter Pharmaceuticals, and Salix/Valeant and has received research funding from Ardelyx, Biomerica, Commonwealth Diagnostics International, Ironwood, Nestlé, QOL Medical, Salix/Valeant, Vibrant, and Zespri. A.J.L. is a consultant for Allergan, Ardelyx, Bioamerica, Ironwood, Prometheus, and Valeant and has received research funding from Prometheus, Bioamerica, Vibrant, and Ironwood. D.P.R. is an employee of, and has ownership interest in, Ardelyx, Inc.

Figures

Figure 1.
Figure 1.
Overview of patient flow through the study. The safety analysis set includes all patients who received ≥1 dose of treatment. The ITT analysis set includes all patients who met the study entry inclusion/exclusion criteria, were randomized, and received ≥1 dose of the study drug. b.i.d., twice daily; ITT, intention-to-treat; PRO, patient-reported outcome.
Figure 2.
Figure 2.
Six- of 12-week responder rates (ITT analysis set): proportions of patients with (a) combined response for ≥6 of the 12 treatment weeks (primary efficacy variable), (b) abdominal pain response for ≥6 of 12 treatment weeks (key secondary efficacy variable), and (c) CSBM response for ≥6 of 12 treatment weeks (key secondary efficacy variable). aThe adjusted RR was based on the ratio of responder rates for tenapanor 50 mg b.i.d. vs placebo b.i.d., stratified by pooled investigator sites using the Mantel–Haenszel method. bThe CMH P value was based on a 1 degree of freedom test for association between treatment (tenapanor and placebo), stratified by pooled investigator sites. An abdominal pain response is defined as a decrease in average weekly worst abdominal pain of ≥30.0% from baseline. A CSBM response is defined as an increase of ≥1 CSBM/week from baseline. A combined response is defined as an abdominal pain response and CSBM response both occurring in the same week. b.i.d., twice daily; CI, confidence interval; CMH, Cochran–Mantel–Haenszel; CSBM, complete spontaneous bowel movement; ITT, intention-to-treat; RR, relative risk.
Figure 3.
Figure 3.
Nine- of 12-week responder rates (ITT analysis set): proportions of patients with (a) combined response for ≥9 of 12 treatment weeks (key secondary efficacy variable), (b) abdominal pain response for ≥9 of 12 treatment weeks (key secondary efficacy variable), and (c) CSBM response for ≥9 of 12 treatment weeks (key secondary efficacy variable). aThe adjusted RR was based on the ratio of responder rates for tenapanor 50 mg b.i.d. vs placebo b.i.d., stratified by pooled investigator sites using the Mantel–Haenszel method. bThe CMH P value was based on a 1 degree of freedom test for association between treatment (tenapanor and placebo), stratified by pooled investigator sites. An abdominal pain response is defined as a decrease in average weekly worst abdominal pain of ≥30.0% from baseline. A CSBM response is defined as an increase of ≥1 CSBM/week from baseline. A combined response is defined as an abdominal pain response and CSBM response both occurring in the same week. b.i.d., twice daily; CI, confidence interval; CMH, Cochran–Mantel–Haenszel; CSBM, complete spontaneous bowel movement; ITT, intention-to-treat; RR, relative risk.
Figure 4.
Figure 4.
Durable responder rates (ITT analysis set): proportions of patients with (a) durable combined response (secondary efficacy variable), (b) durable abdominal pain response (secondary efficacy variable), and (c) durable CSBM response (secondary efficacy variable). aThe adjusted RR was based on the ratio of responder rates for tenapanor 50 mg b.i.d. vs placebo b.i.d., stratified by pooled investigator sites using the Mantel–Haenszel method. bThe CMH P value was based on a 1 degree of freedom test for association between treatment (tenapanor and placebo), stratified by pooled investigator sites. A durable abdominal pain response is defined as a decrease in average weekly worst abdominal pain of ≥30.0% from baseline for ≥9 of 12 treatment weeks, including ≥3 of the final 4 weeks of the treatment period. A durable CSBM response is defined as an increase of ≥1 CSBM/week from baseline for ≥9 of 12 treatment weeks, including ≥3 of the final 4 weeks of the treatment period. A durable combined response is defined as an abdominal pain response and CSBM response, both occurring in the same week, for ≥9 of 12 treatment weeks, including ≥3 of the final 4 weeks of the treatment period. b.i.d., twice daily; CI, confidence interval; CMH, Cochran–Mantel–Haenszel; CSBM, complete spontaneous bowel movement; ITT, intention-to-treat; RR, relative risk.
Figure 5.
Figure 5.
Mean change from baseline in average weekly number of CSBMs over time (ITT analysis set) during the (a) treatment period and (b) RW period. *P < 0.001, †P = 0.001 vs placebo. P values were based on an analysis of covariance model with treatment and pooled investigator site as factors and baseline value as a covariate. Baseline for the treatment period is defined as the average of the first and second weeks of the screening period. Baseline for the RW period is defined as the last valid week of the treatment period. A valid week required at least 4 days of SBM reporting. b.i.d., twice daily; CSBM, complete spontaneous bowel movement; ITT, intention-to-treat; RW, randomized withdrawal.
Figure 6.
Figure 6.
Mean percentage change from baseline in average weekly abdominal pain score over time (ITT analysis set) during the (a) treatment period and (b) RW period. *P < 0.05, †P < 0.001 vs placebo. P values were based on an analysis of covariance model with treatment and pooled investigator site as factors and baseline value as a covariate. Baseline for the treatment period is defined as the average of the first and second weeks of the screening period. Baseline for the RW period is defined as the last valid week of the treatment period. A valid week required at least 4 days of abdominal pain reporting. b.i.d., twice daily; ITT, intention-to-treat; RW, randomized withdrawal.

References

    1. Lacy BE, Chey WD, Lembo AJ. New and emerging treatment options for irritable bowel syndrome. Gastroenterol Hepatol (NY) 2015;11:1–19.
    1. Cash B, Sullivan S, Barghout V. Total costs of IBS: Employer and managed care perspective. Am J Manag Care 2005;11:S7–16.
    1. Saha L. Irritable bowel syndrome: Pathogenesis, diagnosis, treatment, and evidence-based medicine. World J Gastroenterol 2014;20:6759–73.
    1. Endo Y, Shoji T, Fukudo S. Epidemiology of irritable bowel syndrome. Ann Gastroenterol 2015;28:158–9.
    1. Drossman DA, Chey WD, Johanson JF, et al. Clinical trial: Lubiprostone in patients with constipation-associated irritable bowel syndrome—Results of two randomized, placebo-controlled studies. Aliment Pharmacol Ther 2009;29:329–41.
    1. Chey WD, Lembo AJ, Lavins BJ, et al. Linaclotide for irritable bowel syndrome with constipation: A 26-week, randomized, double-blind, placebo-controlled trial to evaluate efficacy and safety. Am J Gastroenterol 2012;107:1702–12.
    1. Brenner DM, Fogel R, Dorn SD, et al. Efficacy, safety, and tolerability of plecanatide in patients with irritable bowel syndrome with constipation: Results of two phase 3 randomized clinical trials. Am J Gastroenterol 2018;113:735–45.
    1. Schoenfeld PS. Advances in IBS 2016: A review of current and emerging data. Gastroenterol Hepatol (NY) 2016;12:1–11.
    1. Rosenbaum DP, Yan A, Jacobs JW. Pharmacodynamics, safety, and tolerability of the NHE3 inhibitor tenapanor: Two trials in healthy volunteers. Clin Drug Investig 2018;38:341–51.
    1. Spencer AG, Labonte ED, Rosenbaum DP, et al. Intestinal inhibition of the Na+/H+ exchanger 3 prevents cardiorenal damage in rats and inhibits Na+ uptake in humans. Sci Transl Med 2014;6:227–36.
    1. Chey WD, Lembo AJ, Rosenbaum DP. Tenapanor treatment of patients with constipation-predominant irritable bowel syndrome: A phase 2, randomized, placebo-controlled efficacy and safety trial. Am J Gastroenterol 2017;112:763–74.
    1. Rome Foundation. Guidelines–rome III diagnostic criteria for functional gastrointestinal disorders. J Gastrointestin Liver Dis 2006;15:307–12.
    1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol 1997;32:920–4.
    1. Mitsuhashi S, Ballou S, Jiang ZG, et al. Characterizing normal bowel frequency and consistency in a representative sample of adults in the United States (NHANES). Am J Gastroenterol 2018;113:115–23.
    1. Kanazawa M, Miwa H, Nakagawa A, et al. Abdominal bloating is the most bothersome symptom in irritable bowel syndrome with constipation (IBS-C): A large population-based internet survey in Japan. Biopsychosoc Med 2016;10:19.
    1. Ringel Y, Williams RE, Kalilani L, et al. Prevalence, characteristics, and impact of bloating symptoms in patients with irritable bowel syndrome. Clin Gastroenterol Hepatol 2009;7:68–72.
    1. Enck P, Aziz Q, Barbara G, et al. Irritable bowel syndrome. Nat Rev Dis Primers 2016;2:16014.
    1. Kiyatkin M, Feng B, Schwartz E, et al. Combined genetic and pharmacological inhibition of TRPV1 and P2X3 attenuates colorectal hypersensitivity and afferent sensitization. Am J Physiol Gastrointest Liver Physiol 2013;305:G638–48.
    1. Wouters M, Balemans D, Wanrooy SV, et al. Histamine receptor H1-mediated sensitization of TRPV1 mediates visceral hypersensitivity and symptoms in patients with irritable bowel syndrome. Gastroenterology 2016;150:875–87.
    1. Li Q, King A, Liu L, et al. Tenapanor reduces IBS pain through inhibition of TRPV1-dependent neuronal hyperexcitability in vivo. Presented at the World Congress of Gastroenterology, October 13–18, 2017, Orlando, USA: American College of Gastroenterology.
    1. Block GA, Rosenbaum DP, Leonsson-Zachrisson M, et al. Effect of tenapanor on serum phosphate in patients receiving hemodialysis. J Am Soc Nephrol 2017;28:1933–42.
    1. Aoki J, Ikari Y. Cardiovascular disease in patients with end-stage renal disease on hemodialysis. Ann Vasc Dis 2017;10:327–37.
    1. Center for Drug Evaluation and Research (CDER). Guidance for industry: Irritable bowel syndrome—Clinical evaluation of drugs for treatment (). Accessed January 2, 2019.
    1. Rey E, Talley NJ. Irritable bowel syndrome: Novel views on the epidemiology and potential risk factors. Dig Liver Dis 2009;41:772–80.
    1. Heitkemper M, Carter E, Ameen V, et al. Women with irritable bowel syndrome: Differences in patients' and physicians' perceptions. Gastroenterol Nurs 2002;25:192–200.

Source: PubMed

3
S'abonner