Effect of comorbid migraine on propranolol efficacy for painful TMD in a randomized controlled trial

Inna E Tchivileva, Richard Ohrbach, Roger B Fillingim, Pei Feng Lim, Massimiliano Di Giosia, Margarete Ribeiro-Dasilva, John H Campbell, Holly Hadgraft, Janet Willis, Samuel J Arbes Jr, Gary D Slade, Inna E Tchivileva, Richard Ohrbach, Roger B Fillingim, Pei Feng Lim, Massimiliano Di Giosia, Margarete Ribeiro-Dasilva, John H Campbell, Holly Hadgraft, Janet Willis, Samuel J Arbes Jr, Gary D Slade

Abstract

Introduction: The migraine-preventive drug propranolol is efficacious in reducing pain from temporomandibular disorder, suggesting potential modifying or mediating effects of comorbid migraine.

Methods: In this randomized controlled trial, myofascial temporomandibular disorder patients were treated with propranolol or placebo for 9 weeks. The primary endpoint was change in a facial pain index derived from daily symptom diaries. Linear and logistic regression models tested for a migraine × treatment-group interaction in reducing facial pain index. Counterfactual models explored changes in headache impact and heart rate as mediators of propranolol's efficacy.

Results: Propranolol's efficacy in reducing facial pain index was greater among the 104 migraineurs than the 95 non-migraineurs: For example, for the binary ≥ 30% reduction in facial pain index, odds ratios were 3.3 (95% confidence limits: 1.4, 8.1) versus 1.3 (0.5, 3.2), respectively, although the interaction was statistically non-significant (p = 0.139). Cumulative response curves confirmed greater efficacy for migraineurs than non-migraineurs (differences in area under the curve 26% and 6%, respectively; p = 0.081). While 9% of the treatment effect was mediated by reduced headache impact, 46% was mediated by reduced heart rate.

Conclusions: Propranolol was more efficacious in reducing temporomandibular disorder pain among migraineurs than non-migraineurs, with more of the effect mediated by reduced heart rate than by reduced headache impact.

Study identification and registration: SOPPRANO; NCT02437383; https://ichgcp.net/clinical-trials-registry/NCT02437383.

Keywords: Adrenergic beta-antagonists; autonomic nervous system; chronic pain; facial pain; headache; sympathetic nervous system.

Conflict of interest statement

Declaration of conflicting interests: The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Figures

Figure 1.
Figure 1.
Change in three endpoints at three follow-up visits for participants in two treatment groups stratified according to the baseline migraine status. Endpoints were estimated for propranolol () and placebo (O) groups in regression models (ITT population). Endpoints are: percentage of participants with a ≥ 30% reduction in FPI plotted for (a) non-migraineurs and (b) migraineurs; percentage of participants with a ≥ 50% reduction in FPI plotted for (c) non-migraineurs and (d) migraineurs; and percentage of participants reporting a reduction of ≥ 6 points in the HIT-6 scale plotted for (e) non-migraineurs and (f) migraineurs. FPI endpoints were recorded at up to three follow-up visits that occurred 1, 5 and 9 weeks after initiating treatment. Percentage reductions in FPI were calculated relative to baseline and dichotomized to signify the percentage of participants with ≥ 30% and ≥ 50% reductions. The HIT-6 score was reported at weeks 5 and 9 and dichotomized to signify the percentage of participants with a ≥ 6-point post-baseline reduction considered a clinically meaningful improvement. Adjusted percentages were estimated from binomial logistic generalized estimating equation models with predictor variables of the baseline migraine status, treatment group, visit, and all two-way and three-way interactions. Covariates were study site, sex, and race/ethnicity (and for the FPI models, the baseline value of FPI). Estimate statements for each model were used to calculate adjusted odds ratios and 95% confidence limits at week 9.
Figure 2.
Figure 2.
Cumulative proportion of FPI responders at week 9 for two treatment groups analyzed by migraine status (ITT population): (a) migraineurs and (b) non-migraineurs. FPI endpoints were recorded at up to three follow-up visits occurring 1, 5 and 9 weeks after initiating treatment with either propranolol () or placebo (O). Adjusted percentages of participants responding, plotted on the vertical axis, were calculated by dichotomizing relative reductions at week 9 compared to week 0. Thresholds for dichotomization are shown on the horizontal axis. Adjusted percentages were estimated with binary logistic regression models using the generalized estimating equation method allowing for repeated visits by study participants. Numbers above plotted values represent a number needed to treat. Area under the curve (AUC) was calculated using the trapezoid rule and expressed as the percentage of maximum response within the range of 20% to 70% reduction (shaded rectangle).
Figure 3.
Figure 3.
Causal mediation of propranolol effect on proportion of FPI responders at week 9. The mediation model is adjusted for study site, sex, race/ethnicity, and baseline FPI. (a) Mediation model with change in the HIT-6 score (continuous variable) as a potential mediator of the total effect of treatment group on a ≥ 50% reduction in FPI. (b) Mediation model with change in heart rate (continuous variable) as a potential mediator of the total effect of treatment group on a ≥ 50% reduction in FPI. Values are covariate-adjusted odds ratios (ORs) and 95% confidence limits.

References

    1. Costen JB. A syndrome of ear and sinus symptoms dependent upon disturbed function of the temporomandibular joint. 1934. Ann Otol Rhinol Laryngol 1997; 106: 805–819.
    1. May A, Svensson P. One nerve, three divisions, two professions and nearly no crosstalk?. Cephalalgia 2017; 37: 603–603.
    1. Plesh O, Adams SH, Gansky SA. Self-reported comorbid pains in severe headaches or migraines in a US national sample. Headache 2012; 52: 946–956.
    1. Ashraf J, Zaproudina N, Suominen AL, et al.. Association between temporomandibular disorders pain and migraine: Results of the Health 2000 Survey. J Oral Facial Pain Headache 2019; 33: 399–407.
    1. Mitrirattanakul S, Merrill RL. Headache impact in patients with orofacial pain. J Am Dent Assoc 2006; 137: 1267–1274.
    1. Franco AL, Goncalves DA, Castanharo SM, et al.. Migraine is the most prevalent primary headache in individuals with temporomandibular disorders. J Orofacial Pain 2010; 24: 287–292.
    1. Dahan H, Shir Y, Nicolau B, et al.. Self-reported migraine and chronic fatigue syndrome are more prevalent in people with myofascial vs nonmyofascial temporomandibular disorders. J Oral Facial Pain Headache 2016; 30: 7–13.
    1. Tchivileva IE, Ohrbach R, Fillingim RB, et al.. Temporal change in headache and its contribution to the risk of developing first-onset temporomandibular disorder in the Orofacial Pain: Prospective Evaluation and Risk Assessment (OPPERA) study. Pain 2017; 158: 120–129.
    1. Graff-Radford SB, Abbott JJ. Temporomandibular disorders and headache. Oral Maxillofacial Surg Clin N Am 2016; 28: 335–349.
    1. Miglis MG. Migraine and autonomic dysfunction: Which is the horse and which is the jockey?. Curr Pain Headache Rep 2018; 22: 19.
    1. Maixner W, Greenspan JD, Dubner R, et al.. Potential autonomic risk factors for chronic TMD: descriptive data and empirically identified domains from the OPPERA case-control study. J Pain 2011; 12: T75–T91.
    1. Conti PC, Costa YM, Goncalves DA, et al.. Headaches and myofascial temporomandibular disorders: Overlapping entities, separate managements?. J Oral Rehab 2016; 43: 702–715.
    1. Shields KG, Goadsby PJ. Propranolol modulates trigeminovascular responses in thalamic ventroposteromedial nucleus: A role in migraine?. Brain 2005; 128: 86–97.
    1. Boyer N, Signoret-Genest J, Artola A, et al.. Propranolol treatment prevents chronic central sensitization induced by repeated dural stimulation. Pain 2017; 158: 2025–2034.
    1. Khasar SG, McCarter G, Levine JD. Epinephrine produces a beta-adrenergic receptor-mediated mechanical hyperalgesia and in vitro sensitization of rat nociceptors. J Neurophysiol 1999; 81: 1104–1112.
    1. Chen YW, Chu CC, Chen YC, et al.. Propranolol elicits cutaneous analgesia against skin nociceptive stimuli in rats. Neurosci Lett 2012; 524: 129–132.
    1. Ciszek BP, O'Buckley SC, Nackley AG. Persistent catechol-O-methyltransferase-dependent pain is initiated by peripheral beta-adrenergic receptors. Anesthesiology 2016; 124: 1122–1135.
    1. Bruehl S, Chung OY. Interactions between the cardiovascular and pain regulatory systems: An updated review of mechanisms and possible alterations in chronic pain. Neurosci Biobehavioral Rev 2004; 28: 395–414.
    1. Petersen KK, Andersen HH, Tsukamoto M, et al.. The effects of propranolol on heart rate variability and quantitative, mechanistic, pain profiling: A randomized placebo-controlled crossover study. Scand J Pain 2018; 18: 479–489.
    1. Tchivileva IE, Hadgraft H, Lim PF, et al.. Efficacy and safety of propranolol for treatment of TMD pain: A randomized, placebo-controlled clinical trial. Pain 2020; 16: 1755–1767.
    1. Schiffman E, Ohrbach R, Truelove E, et al.. Diagnostic criteria for temporomandibular disorders (DC/TMD) for clinical and research applications: Recommendations of the International RDC/TMD Consortium Network and Orofacial Pain Special Interest Group. J Oral Facial Pain Headache 2014; 28: 6–27.
    1. Headache Classification Committee of the International Headache Society. The International Classification of Headache Disorders, 3rd edition (beta version). Cephalalgia 2013; 33: 629–808.
    1. Kosinski M, Bayliss MS, Bjorner JB, et al.. A six-item short-form survey for measuring headache impact: The HIT-6. Qual Life Res 2003; 12: 963–974.
    1. Von Korff M, Ormel J, Keefe FJ, et al.. Grading the severity of chronic pain. Pain 1992; 50: 133–149.
    1. Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand 1983; 67: 361–370.
    1. Dworkin RH, Turk DC, McDermott MP, et al.. Interpreting the clinical importance of group differences in chronic pain clinical trials: IMMPACT recommendations. Pain 2009; 146: 238–244.
    1. Smelt AF, Assendelft WJ, Terwee CB, et al.. What is a clinically relevant change on the HIT-6 questionnaire? An estimation in a primary-care population of migraine patients. Cephalalgia 2014; 34: 29–36.
    1. Davis S. Mixed models for repeated measures using categorical time effects (MMRM). In: O'Kelly M, Ratitch B. (eds) Clinical trials with missing data: A guide for practitioners. New Jersey: John Wiley & Sons, Ltd, 2014, pp. 131–184.
    1. Farrar JT. Advances in clinical research methodology for pain clinical trials. Nat Med 2010; 16: 1284–1293.
    1. Robins JM, Greenland S. Identifiability and exchangeability for direct and indirect effects. Epidemiology 1992; 3: 143–155.
    1. Sun X, Briel M, Walter SD, et al.. Is a subgroup effect believable? Updating criteria to evaluate the credibility of subgroup analyses. BMJ 2010; 340: c117.
    1. Gurung T, Ellard DR, Mistry D, et al.. Identifying potential moderators for response to treatment in low back pain: A systematic review. Physiotherapy 2015; 101: 243–251.
    1. Yunus MB. Central sensitivity syndromes: A new paradigm and group nosology for fibromyalgia and overlapping conditions, and the related issue of disease versus illness. Semin Arthritis Rheum 2008; 37: 339–352.
    1. Monfredi O, Lyashkov AE, Johnsen AB, et al.. Biophysical characterization of the underappreciated and important relationship between heart rate variability and heart rate. Hypertension 2014; 64: 1334–1343.
    1. Pincus T, Miles C, Froud R, et al.. Methodological criteria for the assessment of moderators in systematic reviews of randomised controlled trials: A consensus study. BMC Med Res Methodol 2011; 11: 14.

Source: PubMed

3
Subscribe