Promelaxin Microenemas Are Non-inferior to Oral Polyethylene Glycol for the Treatment of Functional Constipation in Young Children: A Randomized Clinical Trial

Caterina Strisciuglio, Vincenzo Coppola, Marina Russo, Carlo Tolone, Gian Luigi Marseglia, Alberto Verrotti, Silvia Caimmi, Claudia Caloisi, Valeria D'Argenio, Lucia Sacchetti, Annamaria Staiano, Caterina Strisciuglio, Vincenzo Coppola, Marina Russo, Carlo Tolone, Gian Luigi Marseglia, Alberto Verrotti, Silvia Caimmi, Claudia Caloisi, Valeria D'Argenio, Lucia Sacchetti, Annamaria Staiano

Abstract

Background: Polyethylene glycol (PEG) is recommended as first-line treatment of pediatric functional constipation. However, the oral route of administration is often poorly feasible in children mostly due to poor palatability. Promelaxin microenemas exert a topical evacuative action and may offer a valuable option in pediatric FC. Aim: To assess whether Promelaxin microenemas would be non-inferior to PEG 4000 in young children with FC. Methods: This is a randomized, open-label, multi-centric, non-inferiority trial enrolling infants and young children aged 6-48 months, with FC according to Rome III criteria. After 1 week of run in, children were randomized to 2 weeks of Promelaxin or PEG, followed by a 6-week on-demand treatment period. Primary endpoint was response rate to randomized treatment, with "response" defined as at least 3 evacuations per week and an average increase of at least one evacuation per week as compared to baseline. Safety, stool consistency and the analysis of fecal microbiota were secondary endpoints. Results: Out of the 158 patients who entered the trial, 153 patients were treated (77 and 76, PEG and Promelaxin arm, respectively). In the primary analysis, the 95% confidence interval (CI) for the treatment's effect lay entirely above the non-inferiority margin in both Full Set (FAS) and Per Protocol (PP) analyses, providing evidence of the non-inferiority of Promelaxin vs. PEG 4000 [response rate difference: 16.5% (CI 1.55-31.49%) and 11.03% (CI -5.58 to 27.64%), FAS and PP analyses, respectively]. Mean compliance to the randomized treatment was >80% in both arms. Secondary endpoints did not significantly differ between the two arms, except for the average number of total days of on-demand treatment that was significantly lower in the Promelaxin arm [14.6 (12.7) vs. 9.8 (9.1), mean (SD); primary endpoint responders in PEG and Promelaxin arm, respectively; p = 0.027]. Microbiota evenness significantly increased in the PEG 4000 arm at V4 as compared to the Promelaxin arm (p < 0.05). In addition, at V5, patients treated with PEG showed a significantly decreased microbiota density as compared to patients treated with Promelaxin (p = 0.036). Conclusions: Promelaxin microenemas are non-inferior to oral PEG in children with FC. Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT02751411.

Keywords: Promelaxin microenemas; functional constipation; medical devices based on substances; polyethylene glycol; young children.

Conflict of interest statement

AS is clinical investigator for Janssen Biologics B.V. and consultant for Angelini; she was clinical investigator for Aboca and PAREXEL International Srl; she was consultant for Aboca, for D.M.G. Italy and Nestlé, she was data safety monitoring board member for Sucampo AG and speaker for Aboca, Angelini, D.M.G. Italy and Valeas. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2021 Strisciuglio, Coppola, Russo, Tolone, Marseglia, Verrotti, Caimmi, Caloisi, D'Argenio, Sacchetti and Staiano.

Figures

Figure 1
Figure 1
Study Design. The figure depicts the design of the study, including study Visits.
Figure 2
Figure 2
CONSORT diagram. The figure shows the Consort Diagram of the randomized trial.
Figure 3
Figure 3
Primary endpoint. The differences in response rates between Promelaxin and PEG and relative 95% confidence interval (CI) are shown according to the full analysis set (FAS) and the PP analyses.
Figure 4
Figure 4
Response rate for combined stool frequency and consistency and average days of treatment. (A,C) show the response rate (%) for the combined stool frequency and consistency secondary endpoints at V4 and V5, respectively. (B,D) show the average days of treatment (mean ± SE) at V4 and V5, respectively. In each panel, results of separate analyses either according to the PP population or to the subgroup of PP patients evaluated as responders to the primary endpoint (Primary Endpoint Responders) are depicted. Error bars represent Standard Error. (A) Overall PP: PEG, n = 38; Promelaxin, n = 36; Primary Endpoint responders: PEG, n = 25; Promelaxin, n = 31. (B) Overall PP: PEG, n = 15; Promelaxin, n = 19; Primary Endpoint responders: PEG, n = 13; Promelaxin, n = 18. (C) Overall PP: PEG, n = 35; Promelaxin, n = 33; Primary Endpoint responders: PEG, n = 22; Promelaxin, n = 28. (D) Overall PP: PEG, n = 10; Promelaxin, n = 15; Primary Endpoint responders: PEG, n = 9; Promelaxin, n = 15. *p < 0.05.
Figure 5
Figure 5
Stool consistency, QoL improvement and gastrointestinal symptoms in the randomized arms. (A) Percentage of patients with improved stool consistency at V3, V4, and V5. (B) Percentage of parents with improved QoL at V3, V4, and V5. (C–E) Number of days (mean ± SD) with gastrointestinal symptoms at V3, V4, and V5. Data are reported as mean ± SD. *p < 0.05, **p < 0.01, ***p < 0.001, and ****p < 0.0001. (A) V3: PEG, n = 45; Promelaxin, n = 56; V4: PEG, n = 42; Promelaxin, n = 45; V5: PEG, n = 37; Promelaxin, n = 38; (B) V3 (Father): PEG, n = 44; Promelaxin, n = 49. V4 (Father): PEG, n = 38; Promelaxin, n = 39. V5 (Father). PEG, n = 38; Promelaxin, n = 39; (B) V3 (Mother): PEG, n = 44; Promelaxin, n = 52. V4 (Mother): PEG, n = 44; Promelaxin, n = 52. V5 (Mother): PEG, n = 44; Promelaxin, n = 52. (C–E) V3: PEG, n = 45; Promelaxin, n = 56. V4: PEG, n = 40; Promelaxin, n = 47. V5: PEG, n = 40; Promelaxin, n = 47.
Figure 6
Figure 6
Microbiota analysis. The figure shows individual data and box-plots as median and IQR. (A) Microbiota evenness. (B) Microbiota density. Kruskal-Wallis-test and Student T-test were used for testing differences in microbiota evenness (Pielou's index) and microbiota density, respectively. *p < 0.05.

References

    1. van den Berg MM, Benninga MA, Di Lorenzo C. Epidemiology of childhood constipation: a systematic review. Am J Gastroenterol. (2006) 101:2401–9. 10.1111/j.1572-0241.2006.00771.x
    1. Vriesman MH, Koppen IJN, Camilleri M, Di Lorenzo C, Benninga MA. Management of functional constipation in children and adults. Nat Rev Gastroenterol Hepatol. (2020) 17:21–39. 10.1038/s41575-019-0222-y
    1. Koppen IJN, Vriesman MH, Saps M, Rajindrajith S, Shi X, van Etten-Jamaludin FS, et al. . Prevalence of functional defecation disorders in children: a systematic review and meta-analysis. J Pediatr. (2018) 198:121–30.e6. 10.1016/j.jpeds.2018.02.029
    1. Leung AKC, Hon KL. Paediatrics: how to manage functional constipation. Drugs Context. (2021) 10:2020-11-2. 10.7573/dic.2020-11-2
    1. Tang J, Li H, Tang W. Efficacy of non-pharmacologic auxiliary treatments in improving defecation function in children with chronic idiopathic constipation: a systematic review and network meta-analysis. Front Pediatr. (2021) 9:667225. 10.3389/fped.2021.667225
    1. Koppen IJ, Lammers LA, Benninga MA, Tabbers MM. Management of functional constipation in children: therapy in practice. Paediatr Drugs. (2015) 17:349–60. 10.1007/s40272-015-0142-4
    1. Jannin V, Lemagnen G, Gueroult P, Larrouture D, Tuleu C. Rectal route in the 21st century to treat children. Adv Drug Deliv Rev. (2014) 73:34–49. 10.1016/j.addr.2014.05.012
    1. Waterham M, Kaufman J, Gibb S. Childhood constipation. Aust Fam Physician. (2017) 46:908–12.
    1. Tabbers MM, DiLorenzo C, Berger MY, Faure C, Langendam MW, Nurko S, et al. . Evaluation and treatment of functional constipation in infants and children: evidence-based recommendations from ESPGHAN and NASPGHAN. J Pediatr Gastroenterol Nutr. (2014) 58:258–74. 10.1097/MPG.0000000000000266
    1. Venables R, Batchelor H, Hodson J, Stirling H, Marriott J. Determination of formulation factors that affect oral medicines acceptability in a domiciliary paediatric population. Int J Pharm. (2015) 480:55–62. 10.1016/j.ijpharm.2015.01.023
    1. Koppen IJN, van Wassenaer EA, Barendsen RW, Brand PL, Benninga MA. Adherence to polyethylene glycol treatment in children with functional constipation is associated with parental illness perceptions, satisfaction with treatment, and perceived treatment convenience. J Pediatr. (2018) 199:132–9.e1. 10.1016/j.jpeds.2018.03.066
    1. Reflection Paper: Formulations of Choice for the Paediatric Population EMEA/CHMP/PEG/194810/2005.
    1. Bekkali NL, van den Berg MM, Dijkgraaf MG, van Wijk MP, Bongers ME, Liem O, et al. . Rectal fecal impaction treatment in childhood constipation: enemas versus high doses oral PEG. Pediatrics. (2009) 124:e1108–15. 10.1542/peds.2009-0022
    1. Contijoch EJ, Britton GJ, Yang C, Mogno I, Li Z, Ng R, et al. . Gut microbiota density influences host physiology and is shaped by host and microbial factors. Elife. (2019) 8:e40553. 10.7554/eLife.40553
    1. van der Wulp MY, Derrien M, Stellaard F, Wolters H, Kleerebezem M, Dekker J, et al. . Laxative treatment with polyethylene glycol decreases microbial primary bile salt dehydroxylation and lipid metabolism in the intestine of rats. Am J Physiol Gastrointest Liver Physiol. (2013) 305:G474–82. 10.1152/ajpgi.00375.2012
    1. Tropini C, Moss EL, Merrill BD, Ng KM, Higginbottom SK, Casavant EP, et al. . Transient osmotic perturbation causes long-term alteration to the gut microbiota. Cell. (2018) 173:1742–54.e17. 10.1016/j.cell.2018.05.008
    1. Gorkiewicz G, Thallinger GG, Trajanoski S, Lackner S, Stocker G, Hinterleitner T, et al. . Alterations in the colonic microbiota in response to osmotic diarrhea. PLoS ONE. (2013) 8:e55817. 10.1371/journal.pone.0055817
    1. Vich Vila A, Collij V, Sanna S, Sinha T, Imhann F, Bourgonje AR, et al. . Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat Commun. (2020) 11:362. 10.1038/s41467-019-14177-z
    1. Hyman PE, Milla PJ, Benninga MA, Davidson GP, Fleisher DF, Taminiau J. Childhood functional gastrointestinal disorders: neonate/toddler. Gastroenterology. (2006) 130:1519–26. 10.1053/j.gastro.2005.11.065
    1. Bekkali N, Hamers SL, Reitsma JB, Van Toledo L, Benninga MA. Infant stool form scale: development and results. J Pediatr. (2009) 154:521–6.e1. 10.1016/j.jpeds.2008.10.010
    1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand J Gastroenterol. (1997) 32:920–4. 10.3109/00365529709011203
    1. Varni JW, Seid M. Kurtin PS. PedsQL 40: reliability and validity of the Pediatric Quality of Life Inventory version 40 generic core scales in healthy and patient populations. Med Care. (2001) 39:800–12. 10.1097/00005650-200108000-00006
    1. Iaffaldano L, Granata I, Pagliuca C, Esposito MV, Casaburi G, Salerno G, et al. . Oropharyngeal microbiome evaluation highlights Neisseria abundance in active celiac patients. Sci Rep. (2018) 8:11047. 10.1038/s41598-018-29443-1
    1. Nardelli C, Granata I, D'Argenio V, Tramontano S, Compare D, Guarracino MR, et al. . Characterization of the duodenal mucosal microbiome in obese adult subjects by 16S rRNA sequencing. Microorganisms. (2020) 8:485. 10.3390/microorganisms8040485
    1. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, et al. . Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol. (2019) 37:852–7. 10.1038/s41587-019-0209-9
    1. Kern SE. Challenges in conducting clinical trials in children: approaches for improving performance. Expert Rev Clin Pharmacol. (2009) 2:609–17. 10.1586/ecp.09.40
    1. Joseph PD, Craig JC, Caldwell PH. Clinical trials in children. Br J Clin Pharmacol. (2015) 79:357–69. 10.1111/bcp.12305
    1. Dziechciarz P, Horvath A, Szajewska H. Polyethylene glycol 4000 for treatment of functional constipation in children. J Pediatr Gastroenterol Nutr. (2015) 60:65–8. 10.1097/MPG.0000000000000543
    1. Walsh J, Schaufelberger D, Iurian S, Klein S, Batchelor H, Turner R, et al. . Path towards efficient paediatric formulation development based on partnering with clinical pharmacologists and clinicians, a c4c expert group White paper. Br J Clin Pharmacol. (2021). 10.1111/bcp.14989. [Epub ahead of print].
    1. Benninga MA, Faure C, Hyman PE, St James Roberts I, Schechter NL, Nurko S. Childhood functional gastrointestinal disorders: neonate/toddler. Gastroenterology. (2016) 15:S0016-5085(16)00182-7. 10.1053/j.gastro.2016.02.016
    1. Petraroli M, Castellone E, Patianna V, Esposito S. Gut microbiota and obesity in adults and children: the state of the art. Front Pediatr. (2021) 9:657020. 10.3389/fped.2021.657020
    1. Derrien M, Alvarez AS, de Vos WM. The gut microbiota in the first decade of life. Trend Microbiol. (2019) 27:997–1010. 10.1016/j.tim.2019.08.001
    1. Young VB, Schmidt TM. Overview of the gastrointestinal microbiota. Adv Exp Med Biol. (2008) 635:29–40. 10.1007/978-0-387-09550-9_3

Source: PubMed

3
Abonneren