Efficacy of pregabalin in post-traumatic peripheral neuropathic pain: a randomized, double-blind, placebo-controlled phase 3 trial

John Markman, Malca Resnick, Scott Greenberg, Nathaniel Katz, Ruoyong Yang, Joseph Scavone, Ed Whalen, Gabriela Gregorian, Bruce Parsons, Lloyd Knapp, John Markman, Malca Resnick, Scott Greenberg, Nathaniel Katz, Ruoyong Yang, Joseph Scavone, Ed Whalen, Gabriela Gregorian, Bruce Parsons, Lloyd Knapp

Abstract

The growing need for symptomatic treatment of post-traumatic neuropathic pain (PTNP) continues to be unmet. Studies evaluating the efficacy of pregabalin for reducing neuropathic pain following trauma and surgery yielded positive results over ≤ 8-week treatment. To assess the efficacy and tolerability of pregabalin over 3 months in patients with PTNP, a randomized, double-blind, placebo-controlled, parallel-group trial evaluated patients with PTNP at 101 centers in 11 countries-the longest, largest such trial. Adults diagnosed with PTNP were randomly assigned (1:1) to 15 weeks of pregabalin (flexibly dosed 150-600 mg/day) or matching placebo. Primary efficacy analysis was by mixed-model repeated measures comparing change from baseline to week 15 in weekly mean pain scores between active and placebo groups. Evaluable patients included 274 in the pregabalin group and 265 in the placebo group. Trauma was surgical in 49.6% of patients, non-surgical in the remainder. The primary efficacy analysis showed no statistically significant difference between pregabalin and placebo groups in the change from baseline to week 15 [mean difference, - 0.22 points (95% confidence interval, 0.54-0.10); p = 0.1823]. However, comparisons for key secondary outcome measures yielded p values < 0.05 favoring pregabalin. Consistent with the known safety profile of pregabalin, the most common adverse events were dizziness and somnolence (14.6 and 9.9% of patients, respectively) with pregabalin (vs 4.2 and 3.4% with placebo). These findings demonstrate the feasibility of conducting a large, phase 3 registration trial in the heterogeneous PTNP study population.ClinicalTrials.gov NCT01701362.

Keywords: Neuropathic pain; Post-surgical neuropathic pain; Post-traumatic neuropathic pain; Pregabalin.

Conflict of interest statement

Ethical standards

The protocol complied with the Declaration of Helsinki (1964), and was reviewed and approved by the institutional review board at each participating center.

Informed consent

All participants provided written, informed consent.

Conflicts of interest

JM has participated in advisory boards (Pfizer, Editas Medicine, Flexion Therapeutics, Teva, Quark, Pacira, Inspirion Delivery Sciences, Quartet, Pacira Egalet, Biogen, Nektar, Endo, Immune Pharma, Chromocell, Collegium, Purdue, Novartis, Sanofi, Convergence, Aptinyx, Daiichi Sankyo, Allergan, Plasmasurgical, and Grunenthal), received research funding (Depomed, Pfizer), and served on Data Safety Monitoring Boards (Allergan, Novartis). M Resnick, R Yang, J Scavone, E Whalen, G Gregorian, B Parsons, and L Knapp are employees of Pfizer Inc. S Greenberg was an employee of Pfizer at the time of the study and development of the manuscript. N Katz is employed by Analgesic Solutions, which provided a central eligibility verification service to identify patients with PTNP.

Data sharing statement

Upon request, and subject to certain criteria, conditions and exceptions (see https://www.pfizer.com/science/clinical-trials/trial-data-and-results for more information), Pfizer will provide access to individual de-identified participant data from Pfizer-sponsored global interventional clinical studies conducted for medicines, vaccines and medical devices (1) for indications that have been approved in the US and/or EU or (2) in programs that have been terminated (i.e., development for all indications has been discontinued). Pfizer will also consider requests for the protocol, data dictionary, and statistical analysis plan. Data may be requested from Pfizer trials 24 months after study completion. The de-identified participant data will be made available to researchers whose proposals meet the research criteria and other conditions, and for which an exception does not apply, via a secure portal. To gain access, data requestors must enter into a data access agreement with Pfizer.

Figures

Fig. 1
Fig. 1
Trial profile. Subjects may have met more than one criterion for exclusion. Of 584 not meeting eligibility criteria, 190 did not meet requirements for neuropathic pain assessment; 71 did not meet the required duration of PTNP; 69 did not meet pain diary criteria prior to randomization; 65 were unwilling/unable to comply with study procedures; 44 had exclusionary pain conditions; 43 did not have the implicated peripheral nerve identified; 34 had other exclusionary NeP conditions; 34 had creatinine clearance ≤ 60 mL/min; and 22 were taking prohibited medications
Fig. 2
Fig. 2
Change from baseline in weekly mean pain score (daily pain NRS; ITT population). *Unadjusted p < 0.05 from MMRM analysis. Changes in weekly mean pain score ± standard error were estimated from mixed-model repeated measures (MMRM) model. Weekly mean pain numerical rating scale (NRS) scores are derived from the daily pain NRS and calculated as the mean of the available scores in the 7 days. Generally, week “n” mean pain score is defined as the mean of the 7 daily diary pain ratings from day 2 + 7*(n–1) to day 1 + 7*n. At least four entries within the last 7 days are required to calculate a mean score. NRS ranged from 0 (“no pain”) to 10 (“worst possible pain”), with higher scores indicating increased pain. ITT intention to treat

References

    1. Crombie IK, Davies HT, Macrae WA. Cut and thrust: antecedent surgery and trauma among patients attending a chronic pain clinic. Pain. 1998;76(1–2):167–171. doi: 10.1016/S0304-3959(98)00038-4.
    1. Kehlet H, Jensen TS, Woolf CJ. Persistent postsurgical pain: risk factors and prevention. Lancet. 2006;367(9522):1618–1625. doi: 10.1016/S0140-6736(06)68700-X.
    1. Treede RD, Jensen TS, Campbell JN, Cruccu G, Dostrovsky JO, Griffin JW, Hansson P, Hughes R, Nurmikko T, Serra J. Neuropathic pain: redefinition and a grading system for clinical and research purposes. Neurology. 2008;70(18):1630–1635. doi: 10.1212/01.wnl.0000282763.29778.59.
    1. Haanpaa M, Attal N, Backonja M, Baron R, Bennett M, Bouhassira D, Cruccu G, Hansson P, Haythornthwaite JA, Iannetti GD, Jensen TS, Kauppila T, Nurmikko TJ, Rice AS, Rowbotham M, Serra J, Sommer C, Smith BH, Treede RD. NeuPSIG guidelines on neuropathic pain assessment. Pain. 2011;152(1):14–27. doi: 10.1016/j.pain.2010.07.031.
    1. von Hehn CA, Baron R, Woolf CJ. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron. 2012;73(4):638–652. doi: 10.1016/j.neuron.2012.02.008.
    1. Gilron I, Kehlet H. Prevention of chronic pain after surgery: new insights for future research and patient care. Can J Anaesth. 2014;61(2):101–111. doi: 10.1007/s12630-013-0067-8.
    1. Macrae WA. Chronic post-surgical pain: 10 years on. Br J Anaesth. 2008;101(1):77–86. doi: 10.1093/bja/aen099.
    1. Wong K, Phelan R, Kalso E, Galvin I, Goldstein D, Raja S, Gilron I. Antidepressant drugs for prevention of acute and chronic postsurgical pain: early evidence and recommended future directions. Anesthesiology. 2014;121(3):591–608. doi: 10.1097/ALN.0000000000000307.
    1. Lyrica (pregabalin) [Prescribing Information]. Pfizer Inc. NY, NY (2016) . Accessed 25 Jan 2018
    1. van Seventer R, Bach FW, Toth CC, Serpell M, Temple J, Murphy TK, Nimour M. Pregabalin in the treatment of post-traumatic peripheral neuropathic pain: a randomized double-blind trial. Eur J Neurol. 2010;17(8):1082–1089. doi: 10.1111/j.1468-1331.2010.02979.x.
    1. US Department of Health and Human Services FaDA, Center for Drug Evaluation and Research (CDER) (2014) Guidance for industry analgesic indications: developing drug and biological products. . Accessed 10 Nov 2017
    1. Dworkin RH, Turk DC, Peirce-Sandner S, Burke LB, Farrar JT, Gilron I, Jensen MP, Katz NP, Raja SN, Rappaport BA, Rowbotham MC, Backonja MM, Baron R, Bellamy N, Bhagwagar Z, Costello A, Cowan P, Fang WC, Hertz S, Jay GW, Junor R, Kerns RD, Kerwin R, Kopecky EA, Lissin D, Malamut R, Markman JD, McDermott MP, Munera C, Porter L, Rauschkolb C, Rice AS, Sampaio C, Skljarevski V, Sommerville K, Stacey BR, Steigerwald I, Tobias J, Trentacosti AM, Wasan AD, Wells GA, Williams J, Witter J, Ziegler D. Considerations for improving assay sensitivity in chronic pain clinical trials: IMMPACT recommendations. Pain. 2012;153(6):1148–1158. doi: 10.1016/j.pain.2012.03.003.
    1. Freynhagen R, Baron R, Gockel U, Tolle TR. painDETECT: a new screening questionnaire to identify neuropathic components in patients with back pain. Curr Med Res Opin. 2006;22(10):1911–1920. doi: 10.1185/030079906X132488.
    1. Burstein R, Collins B, Jakubowski M. Defeating migraine pain with triptans: a race against the development of cutaneous allodynia. Ann Neurol. 2004;55(1):19–26. doi: 10.1002/ana.10786.
    1. Kroenke K, Spitzer RL, Williams JB. The PHQ-15: validity of a new measure for evaluating the severity of somatic symptoms. Psychosom Med. 2002;64(2):258–266. doi: 10.1097/00006842-200203000-00008.
    1. Posner K, Brent D, Lucas C, Gould M, Stanley B, Brown G, Fisher P, Zelazny J, Burke A, Oquendo M, Mann J. Columbia-suicide severity rating scale. New York: C-SSRS) The Research Foundation for Mental Hygiene; 2009.
    1. Hays RD, Stewart A. Measuring functioning and well-being: the medical outcomes study approach. Durham: Duke University Press; 1992.
    1. Cleeland CS. Measurement and prevalence of pain in cancer. Semin Oncol Nurs. 1985;1:87–92. doi: 10.1016/S0749-2081(85)80041-3.
    1. EuroQol Group EuroQo—a new facility for the measurement of health-related quality of life. Health Policy. 1990;16:199–208. doi: 10.1016/0168-8510(90)90421-9.
    1. Reilly MC, Zbrozek AS, Dukes EM. The validity and reproducibility of a work productivity and activity impairment instrument. Pharmacoeconomics. 1993;4(5):353–365. doi: 10.2165/00019053-199304050-00006.
    1. Quessy SN, Rowbotham MC. Placebo response in neuropathic pain trials. Pain. 2008;138(3):479–483. doi: 10.1016/j.pain.2008.06.024.
    1. Rosenstock J, Tuchman M, LaMoreaux L, Sharma U. Pregabalin for the treatment of painful diabetic peripheral neuropathy: a double-blind, placebo-controlled trial. Pain. 2004;110(3):628–638. doi: 10.1016/j.pain.2004.05.001.
    1. Sabatowski R, Galvez R, Cherry DA, Jacquot F, Vincent E, Maisonobe P, Versavel M, Study G. Pregabalin reduces pain and improves sleep and mood disturbances in patients with post-herpetic neuralgia: results of a randomised, placebo-controlled clinical trial. Pain. 2004;109(1–2):26–35. doi: 10.1016/j.pain.2004.01.001.
    1. Demant DT, Lund K, Vollert J, Maier C, Segerdahl M, Finnerup NB, Jensen TS, Sindrup SH. The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. Pain. 2014;155(11):2263–2273. doi: 10.1016/j.pain.2014.08.014.

Source: PubMed

3
S'abonner