A Model-Based Meta-analysis to Compare Efficacy and Tolerability of Tramadol and Tapentadol for the Treatment of Chronic Non-Malignant Pain

François Mercier, Laurent Claret, Klaas Prins, René Bruno, François Mercier, Laurent Claret, Klaas Prins, René Bruno

Abstract

Introduction: Pain is a major symptom in many medical conditions which can be relieved thanks to analgesics. The goal of this work was to present an indirect comparison of efficacy and tolerability profiles of two analgesics, tramadol and tapentadol, in patients with chronic non-malignant pain.

Methods: In the absence of a head-to-head comparison between these two opioid drugs, model-based meta-analyses were used to characterize the pain intensity time dynamics and evaluate the proportions of most frequent adverse events (constipation, nausea, vomiting, dizziness, and somnolence) and drop-outs (due to adverse event, as well as due to lack of efficacy) in each treatment group. Using these models, the investigational treatments were compared on the basis of Monte Carlo simulation outcomes.

Results: Data were extracted from 45 Phase II and Phase III studies representing a total of 81 treatment arms, i.e., approximately 13,000 patients. The pain intensity model shows, that after having adjusted for differences in baseline pain intensity and placebo effects, tramadol 300 mg once daily (qd) was slightly more effective in reducing pain than tapentadol 100-250 mg twice daily (bid), with a 46% change from baseline for the former versus 36% for the latter. From a tolerability standpoint, both drugs showed, as expected, increased risks of adverse events compared to placebo. Yet, tapentadol was associated with slightly lower risks of constipation, and nausea than tramadol.

Conclusion: Overall, the analysis showed that the benefit-risk profiles of tramadol 300 mg qd and tapentadol 100-250 mg bid were approximately even. The amount of data to characterize dose-response relationships was sufficient only in the tramadol group; public access to tapentadol efficacy and tolerability readouts across a wide dose range in chronic non-malignant pain would allow a comparison of therapeutic indices, a straight quantitation of the benefit-risk ratio. Knowing that their side-effects have been identified as potential hindrance to prescription, a broad and open access to clinical trial data in this indication is encouraged in order to facilitate the evaluation of the opiate analgesics clinical utility.

Figures

Fig. 1
Fig. 1
Pain intensity (normalized to a 0–10 scale) over time, in patients treated for chronic non-malignant pain, with placebo, tapentadol, or tramadol. Each circle represents the arm-level average score, with a diameter proportional to the sample size in the arm. The outer curves give the 95% predictive interval and the bold curve the predicted median, using the final pain intensity model
Fig. 2
Fig. 2
Distribution of 1,000 differences in predicted group-level pain intensity (normalized to a 0–10 scale) after 12 weeks of treatment with tramadol (300 mg one daily) versus tapentadol (100–250 mg twice daily) (ΔPI). Predictions of pain intensity were simulated from the final model, assuming 1,000 patients per arm. The plain and dashed vertical lines materialize the median and 95% prediction interval, respectively
Fig. 3
Fig. 3
From left to right number of arms (Narms), frequency of adverse event, odds-ratio (with 95% confidence interval) for the active groups versus placebo and odds-ratio (with 95% confidence interval) for tramadol (300 mg once daily) versus tapentadol (100–250 mg twice daily), for each type of adverse event (Constip. constipation, Dizzin. dizziness, Somnol. somnolence)
Fig. 4
Fig. 4
From left to right number of arms (Narms), proportion of drop-out patients, odds-ratio (with 95% confidence interval) for the active groups versus placebo, and odds-ratio (with 95% confidence interval) for tramadol (300 mg once daily) versus tapentadol (100–25/0 mg twice daily), per reason of withdrawal (DO.AE, drop-out due to adverse event; or DO.LoE, drop-out due to lack of efficacy)

References

    1. Elliott AM, Smith BH, Penny KI, Smith WCM, Chambers WA. The epidemiology of chronic pain in the community. Lancet. 1999;354:1248–1252. doi: 10.1016/S0140-6736(99)03057-3.
    1. Gureje O, Simon GE, Von Korff M. A cross-national study of the course of persistent pain in primary care. Pain. 2001;95:195–200. doi: 10.1016/S0304-3959(00)00483-8.
    1. Sjoegren P, Ekholm O, Peuckmann V, Groenbaek M. Epidemiology of chronic pain in Denmark: an update. Eur J Pain. 2009;13:287–292. doi: 10.1016/j.ejpain.2008.04.007.
    1. Nuckols TK, Anderson L, Popescu I, et al. Opioid prescribing: a systematic review and critical appraisal of guidelines for chronic pain. Ann Intern Med. 2014;160:38–47.
    1. Nossaman VE, Ramadhyani U, Kadowitz PJ, Nossaman BD. Advances in perioperative pain management: use of medications with dual analgesic mechanisms, tramadol and tapentadol. Anesthesiol Clin. 2010;28:647–666. doi: 10.1016/j.anclin.2010.08.009.
    1. Mandema JW, Cox E, Alderman J. Therapeutic benefit of eletriptan compared to sumatriptan for the acute relief of migraine pain—results of a model-based meta-analysis that accounts for encapsulation. Cephalgia. 2005;25:715–725. doi: 10.1111/j.1468-2982.2004.00939.x.
    1. Ito K, Ahadieh S, Corrigan B, French J, Fullerton T, Tensfeldt T. Disease progression meta-analysis model in Alzheimer’s disease. Alzheimer’s Dement. 2010;6:39–53. doi: 10.1016/j.jalz.2009.05.665.
    1. Gibbs JP, Fredrickson J, Barbee T, et al. Quantitative model of the relationship between dipeptidyl peptidase-4 (DDP-4) inhibition and response: meta-analysis of alogliptin, saxagliptin, sitagliptin, and vildagliptin efficacy results. J Clin Pharmacol. 2012;52:1494–1505. doi: 10.1177/0091270011420153.
    1. Ahn JE, French JL. Longitudinal aggregate data model-based meta-analysis with NONMEM: approaches to handling within treatment arm correlation. J Pharmacokinet Pharmacodyn. 2010;37:179–201. doi: 10.1007/s10928-010-9152-6.
    1. Benyamin R, Trescot AM, Datta S, et al. Opioid complications and side effects. Pain Physician. 2008;11:S105–S120.
    1. Moore RA, McQuay HJ. Prevalence of opioid adverse events in chronic non-malignant pain: systematic review of randomised trials of oral opioids. Arthritis Res Ther. 2005;7:R1046–R1051. doi: 10.1186/ar1782.
    1. Chou R, Clark E, Helfand M. Comparative efficacy and safety of long-acting oral opioids for chronic non-cancer pain: a systematic review. J Pain Symptom Manag. 2003;26:1026–1048. doi: 10.1016/j.jpainsymman.2003.03.003.
    1. Harrick C, van Hove I, Stegmann J-U, Oh C, Upmalis D. Efficacy and tolerability of tapentadol immediate release and oxycodone HCl immediate release in patients awaiting primary joint replacement surgery for end-stage joint disease: a 10-day, phase III, randomized, double-blind, active- and placebo-controlled study. Clin Ther. 2009;31:260–271. doi: 10.1016/j.clinthera.2009.02.009.
    1. Hale M, Upmalis D, Okamoto A, Lange C, Rauschkolb C. Tolerability of tapentadol immediate release in patients with lower back pain or osteoarthritis of the hip or knee over 90 days: a randomized, double-blind study. Curr Med Res Opin. 2009;25:1095–1104. doi: 10.1185/03007990902816970.
    1. Kelly K. Dose stability of tapentadol extended release (ER) for the relief of moderate-to-severe chronic osteoarthritic knee pain. In: 26th Annual Meeting of the American Academy of Pain Medicine (AAPM), February 3–6, 2010 in San Antonio, TX, Abstract of Poster 125.
    1. Wild JE, Grond S, Kuperwasser B, et al. Long-term safety and tolerability of tapentadol extended release for the management of chronic low back pain or osteoarthritis pain. Pain Pract. 2010;10:416–427. doi: 10.1111/j.1533-2500.2010.00397.x.
    1. Afilalo M, Etropolski MS, Kuperwasser B, et al. Efficacy and safety of tapentadol extended release compared with oxycodone controlled release for the management of moderate to severe chronic pain related to osteoarthritis of the knee: a randomized, double-blind, placebo- and active-controlled phase III study. Clin Drug Investig. 2010;30:489–505. doi: 10.2165/11533440-000000000-00000.
    1. Buynak R, Shapiro DY, Okamoto A, et al. Efficacy and safety of tapentadol extended release for the management of chronic low back pain: results of a prospective, randomized, double-blind, placebo- and active-controlled phase III study. Expert Opin Pharmacother. 2010;11:1787–1804. doi: 10.1517/14656566.2010.497720.
    1. Adler L, McDonald C, O’Brien C, Wilson M. A comparison of once-daily tramadol with normal release tramadol in the treatment of pain in osteoarthritis. J Rheumatol. 2002;29:2196–2199.
    1. Mongin G, Yakusevich V, Köpe A, et al. Efficacy and safety assessment of a novel once-daily tablet formulation of tramadol: A randomized, controlled study versus twice-daily tramadol in patients with osteoarthritis of the knee. Clin Drug Investig. 2004;24:545–558. doi: 10.2165/00044011-200424090-00005.
    1. Beaulieu AD, Peloso P, Bensen W, et al. A randomized, double-blind, 8-week, crossover study of once-daily, controlled-release tramadol versus immediate-release tramadol taken as needed for chronic noncancer pain. Clin Ther. 2007;29:49–60. doi: 10.1016/j.clinthera.2007.01.001.
    1. Turner JA, Deyo RA, Loeser JD, Von Korff M, Fordyce WE. The importance of placebo effects in pain treatment and research. J Am Med Assoc. 1994;271:1609–1614. doi: 10.1001/jama.1994.03510440069036.
    1. Pinheiro J, Bates D, DebRoy S, Sarkar D, the R Development Core Team. nlme: linear and nonlinear mixed effects models. 2012 R package version 3.1-103.
    1. Bates D, Maechler M, Bolker B, the R Development Core Team. Lme4: linear mixed-effects models using S4 classes. 2011 R package version 0.999375.
    1. Butcher HG, Guyatt GH, Griffith LE, Walter SD. The results of direct and indirect treatment comparisons in meta-analysis of randomized controlled trials. J Clin Epidemiol. 1997;50:683–691. doi: 10.1016/S0895-4356(97)00049-8.
    1. Norrbrink C, Lundeberg T. Tramadol in neuropathic pain after spinal cord injury: a randomized, double-blind, placebo-controlled trial. Clin J Pain. 2009;25:177–184. doi: 10.1097/AJP.0b013e31818a744d.
    1. Katz J, Finnerup NB, Dworkin RH. Clinical trial outcome in neuropathic pain: relationship to study characteristics. Neurology. 2008;70:263–272. doi: 10.1212/01.wnl.0000275528.01263.6c.
    1. Cepeda MS, Africano JM, Polo R, Alcala R, Carr DB. What decline in pain intensity is meaningful to patients with acute pain? Pain. 2003;105:151–157. doi: 10.1016/S0304-3959(03)00176-3.
    1. Dunn AG, Bourgeois FT, Murthy S, Mandl KD, Day RO, Coiera E. The role and impact of research agendas on the comparative-effectiveness research among antihyperlipidemics. Clin Pharmacol Ther. 2012;91:685–691. doi: 10.1038/clpt.2011.279.
    1. Therneau TM, Grambsch PM. Modeling survival data: extending the Cox model. New York: Springer; 2000.
    1. Harnisch L, Shepard T, Pons G, Della Pasqua O. Modeling and simulation as a tool to bridge efficacy and safety data in special populations. CPT Pharmacomet Syst Pharmacol. 2013;2:e28. doi: 10.1038/psp.2013.6.
    1. Lumley T. Network meta-analysis for indirect treatment comparisons. Stat Med. 2002;21:2313–2324. doi: 10.1002/sim.1201.
    1. Lee EY, Lee EB, Park BJ, et al. Tramadol 37.5-mg/acetaminophen 325-mg combination tablets added to regular therapy for rheumatoid arthritis pain: a 1-week, randomized, double-blind, placebo-controlled trial. Clin Ther. 2006;28:2052–2060. doi: 10.1016/j.clinthera.2006.12.019.
    1. Bennett RM, Kamin M, Karim R, et al. Tramadol and acetaminophen combination tablets in the treatment of fibromyalgia pain: a double-blind, randomized, placebo-controlled study. Am J Med. 2003;114:537–545. doi: 10.1016/S0002-9343(03)00116-5.

Source: PubMed

3
Prenumerera