Efficacy and Safety of Analgesic Treatment for Depression in People with Advanced Dementia: Randomised, Multicentre, Double-Blind, Placebo-Controlled Trial (DEP.PAIN.DEM)

Ane Erdal, Elisabeth Flo, Dag Aarsland, Clive Ballard, Dagrun D Slettebo, Bettina S Husebo, Ane Erdal, Elisabeth Flo, Dag Aarsland, Clive Ballard, Dagrun D Slettebo, Bettina S Husebo

Abstract

Background: Chronic pain and depression often co-occur, and pain may exacerbate depression in people with dementia.

Objective: The objective of this study was to assess the efficacy and safety of analgesic treatment for depression in nursing home patients with advanced dementia and clinically significant depressive symptoms.

Methods: We conducted a multicentre, parallel-group, double-blind, placebo-controlled trial in 47 nursing homes, including 162 nursing home patients aged ≥ 60 years with dementia (Mini-Mental State Examination ≤ 20) and depression (Cornell Scale for Depression in Dementia ≥ 8). Patients were randomised to receive active analgesic treatment (paracetamol or buprenorphine transdermal system) or identical placebo for 13 weeks. The main outcome measure was the change in depression (Cornell Scale for Depression in Dementia) from baseline to 13 weeks, assessed using linear mixed models with fixed effects for time, intervention and their interaction in the models. Secondary outcomes were to assess whether any change in depression was secondary to change in pain (Mobilisation-Observation-Behaviour-Intensity-Dementia-2 Pain Scale) and adverse events.

Results: The mean depression change was - 0.66 (95% confidence interval - 2.27 to 0.94) in the active group (n = 80) and - 3.30 (- 4.68 to -1.92) in the placebo group (n = 82). The estimated treatment effect was 2.64 (0.55-4.72, p = 0.013), indicating that analgesic treatment had no effect on depressive symptoms from baseline to 13 weeks while placebo appeared to ameliorate depressive symptoms. There was no significant reduction in pain in the active treatment group (paracetamol and buprenorphine combined) vs. placebo; however, a subgroup analysis demonstrated a significant reduction in pain for paracetamol vs. placebo [by - 1.11 (- 2.16 to - 0.06, p = 0.037)] from week 6 to 13 without a change in depression. Buprenorphine did not have significant effects on depression [3.04 (- 0.11 to 6.19), p = 0.059] or pain [0.47 (- 0.77 to 1.71), p = 0.456] from 0 to 13 weeks. Thirty-five patients were withdrawn from the study because of adverse reactions, deterioration or death: 25 (31.3%) during active treatment [23 (52.3%) who received buprenorphine], and ten (12.2%) in the placebo group. The most frequently occurring adverse events were psychiatric (17 adverse reactions) and neurological (14 adverse reactions).

Conclusion: Analgesic treatment did not reduce depression while placebo appeared to improve depressive symptoms significantly by comparison, possibly owing to the adverse effects of active buprenorphine. The risk of adverse events warrants caution when prescribing buprenorphine for people with advanced dementia.

Trial registration: ClinicalTrials.gov NCT02267057 (registered 7 July, 2014) and Norwegian Medicines Agency EudraCT 2013-002226-23.

Conflict of interest statement

Conflict of interest

Clive Ballard has received consultancy honoraria from Acadia, Lundbeck, Heptares, Roche, Lilly, Otsuka, GSK, Pfizer and Synexus; speaker fees from Lundbeck, Lilly and Otsuka; and grant support from Acadia Pharmaceuticals 2014–2017. Ane Erdal, Elisabeth Flo, Dag Aarsland, Dagrun D. Slettebo and Bettina S. Husebo have no conflicts of interest directly relevant to the content of this article. The sponsors had no influence on the study design, data collection and analysis, decision to publish or preparation of the manuscript.

Ethics approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards. Prior to enrolment, the trial was registered in ClinicalTrials.gov (NCT02267057), and was approved by the Norwegian Medicines Agency (EudraCT 2013-002226-23) and the Regional Committee for Medical and Health Research Ethics (REC-West 2013/1474).

Consent to participate

Informed consent was obtained from all individual participants included in the study. Verbal and written informed consent was obtained in direct conversation with all patients who were deemed to have medical decision-making capacity. If participants did not have the capacity to give consent, the participant’s next of kin or legal guardian provided informed consent in accordance with ethics committee requirements and Norwegian legislation at the time of the study. We expected that patients with Mini-Mental State Examination scores ≥ 16 would be able to give informed consent [41], but nevertheless we included the closest relatives of all patients in a discussion about consent and provided written information about the trial to ensure full transparency. To empower those patients with a reduced ability to consent, we attempted to adjust the information procedure to enable them to understand the purpose and implications of study participation. We included a verbal and written statement assuring that their decision to give consent would not affect the quality of the medical care provided to the patient. Even though informed consent had been given, all participants were free to decline drug administration and other procedures at any time during the trial, irrespective of cognitive state.

Figures

Fig. 1
Fig. 1
Trial profile. Each patient was categorised by the first exclusion criterion that was fulfilled, after which no more assessments were made. CSDD Cornell Scale for Depression in Dementia, MMSE Mini-Mental State Examination, NHs nursing homes
Fig. 2
Fig. 2
Change in depressive symptoms (Cornell Scale for Depression in Dementia) throughout the study period. CI confidence interval, MMSE Mini-Mental State Examination, MOBID-2 Mobilisation-Observation-Behaviour-Intensity-Dementia-2 Pain Scale
Fig. 3
Fig. 3
Change in pain intensity [Mobilisation-Observation-Behaviour-Intensity-Dementia-2 Pain Scale (MOBID-2)] throughout the study period in patients with moderate-to-severe pain at baseline (MOBID-2 ≥ 3). CI confidence interval

References

    1. Janus SI, van Manen JG. IJzerman MJ, Zuidema SU. Psychotropic drug prescriptions in Western European nursing homes. Int Psychogeriatr. 2016;28(11):1775–1790. doi: 10.1017/S1041610216001150.
    1. Helvik AS, Engedal K, Benth JS, Selbaek G. Prevalence and severity of dementia in nursing home residents. Dement Geriatr Cogn Disord. 2015;40(3–4):166–177. doi: 10.1159/000433525.
    1. Bergh S, Selbaek G, Engedal K. Discontinuation of antidepressants in people with dementia and neuropsychiatric symptoms (DESEP study): double blind, randomised, parallel group, placebo controlled trial. BMJ. 2012;344:e1566. doi: 10.1136/bmj.e1566.
    1. Lyketsos CG, DelCampo L, Steinberg M, Miles Q, Steele CD, Munro C, et al. Treating depression in Alzheimer disease: efficacy and safety of sertraline therapy, and the benefits of depression reduction: the DIADS. Arch Gen Psychiatry. 2003;60(7):737–746. doi: 10.1001/archpsyc.60.7.737.
    1. Banerjee S, Hellier J, Dewey M, Romeo R, Ballard C, Baldwin R, et al. Sertraline or mirtazapine for depression in dementia (HTA-SADD): a randomised, multicentre, double-blind, placebo-controlled trial. Lancet. 2011;378(9789):403–411. doi: 10.1016/S0140-6736(11)60830-1.
    1. Rosenberg PB, Drye LT, Martin BK, Frangakis C, Mintzer JE, Weintraub D, et al. Sertraline for the treatment of depression in Alzheimer disease. Am J Geriatr Psychiatry. 2010;18(2):136–145. doi: 10.1097/JGP.0b013e3181c796eb.
    1. Rabins PV, Blacker D, Rovner BW, Rummans T, Schneider LS, Tariot PN, et al. American Psychiatric Association practice guideline for the treatment of patients with Alzheimer’s disease and other dementias. Second edition. Am J Psychiatry. 2007;164(12 Suppl.):5–56.
    1. Farina N, Morrell L, Banerjee S. What is the therapeutic value of antidepressants in dementia? A narrative review. Int J Geriatr Psychiatry. 2017;32(1):32–49. doi: 10.1002/gps.4566.
    1. Nelson JC, Devanand DP. A systematic review and meta-analysis of placebo-controlled antidepressant studies in people with depression and dementia. J Am Geriatr Soc. 2011;59(4):577–585. doi: 10.1111/j.1532-5415.2011.03355.x.
    1. Sandvik RK, Selbaek G, Seifert R, Aarsland D, Ballard C, Corbett A, et al. Impact of a stepwise protocol for treating pain on pain intensity in nursing home patients with dementia: a cluster randomized trial. Eur J Pain. 2014;18(10):1490–1500. doi: 10.1002/ejp.523.
    1. Achterberg WP, Gambassi G, Finne-Soveri H, Liperoti R, Noro A, Frijters DH, et al. Pain in European long-term care facilities: cross-national study in Finland. Italy and The Netherlands. Pain. 2010;148(1):70–74. doi: 10.1016/j.pain.2009.10.008.
    1. Hadjistavropoulos T, Herr K, Prkachin KM, Craig KD, Gibson SJ, Lukas A, et al. Pain assessment in elderly adults with dementia. Lancet Neurol. 2014;13(12):1216–1227. doi: 10.1016/S1474-4422(14)70103-6.
    1. Erdal A, Flo E, Selbaek G, Aarsland D, Bergh S, Slettebo DD, et al. Associations between pain and depression in nursing home patients at different stages of dementia. J Affect Disord. 2017;218:8–14. doi: 10.1016/j.jad.2017.04.038.
    1. Leong IY, Nuo TH. Prevalence of pain in nursing home residents with different cognitive and communicative abilities. Clin J Pain. 2007;23(2):119–127. doi: 10.1097/01.ajp.0000210951.01503.3b.
    1. Chopra K, Arora V. An intricate relationship between pain and depression: clinical correlates, coactivation factors and therapeutic targets. Expert Opin Ther Targets. 2014;18(2):159–176. doi: 10.1517/14728222.2014.855720.
    1. Husebo BS, Ballard C, Fritze F, Sandvik RK, Aarsland D. Efficacy of pain treatment on mood syndrome in patients with dementia: a randomized clinical trial. Int J Geriatr Psychiatry. 2014;29(8):828–836. doi: 10.1002/gps.4063.
    1. van Ojik AL, Jansen PA, Brouwers JR, van Roon EN. Treatment of chronic pain in older people: evidence-based choice of strong-acting opioids. Drugs Aging. 2012;29(8):615–625. doi: 10.1007/BF03262278.
    1. Karp JF, Butters MA, Begley AE, Miller MD, Lenze EJ, Blumberger DM, et al. Safety, tolerability, and clinical effect of low-dose buprenorphine for treatment-resistant depression in midlife and older adults. J Clin Psychiatry. 2014;75(8):e785–e793. doi: 10.4088/JCP.13m08725.
    1. Durso GR, Luttrell A, Way BM. Over-the-counter relief from pains and pleasures alike: acetaminophen blunts evaluation sensitivity to both negative and positive stimuli. Psychol Sci. 2015;26(6):750–758. doi: 10.1177/0956797615570366.
    1. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”: a practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189–198. doi: 10.1016/0022-3956(75)90026-6.
    1. Alexopoulos GS, Abrams RC, Young RC, Shamoian CA. Cornell Scale for Depression in Dementia. Biol Psychiatry. 1988;23(3):271–284. doi: 10.1016/0006-3223(88)90038-8.
    1. Husebo BS, Ostelo R, Strand LI. The MOBID-2 pain scale: reliability and responsiveness to pain in patients with dementia. Eur J Pain. 2014;18(10):1419–1430. doi: 10.1002/ejp.507.
    1. Cummings JL, Mega M, Gray K, Rosenberg-Thompson S, Carusi DA, Gornbein J. The Neuropsychiatric Inventory: comprehensive assessment of psychopathology in dementia. Neurology. 1994;44(12):2308–2314. doi: 10.1212/WNL.44.12.2308.
    1. Goodarzi ZS, Mele BS, Roberts DJ, Holroyd-Leduc J. Depression case finding in individuals with dementia: a systematic review and meta-analysis. J Am Geriatr Soc. 2017;65(5):937–948. doi: 10.1111/jgs.14713.
    1. Perneczky R, Wagenpfeil S, Komossa K, Grimmer T, Diehl J, Kurz A. Mapping scores onto stages: mini-mental state examination and clinical dementia rating. Am J Geriatr Psychiatry. 2006;14(2):139–144. doi: 10.1097/01.JGP.0000192478.82189.a8.
    1. Twisk JWR. Applied longitudinal data analysis for epidemiology: a practical guide. 2. Cambridge: Cambridge University Press; 2013.
    1. Benedetti F, Carlino E, Piedimonte A. Increasing uncertainty in CNS clinical trials: the role of placebo, nocebo, and Hawthorne effects. Lancet Neurol. 2016;15(7):736–747. doi: 10.1016/S1474-4422(16)00066-1.
    1. Benedetti F, Arduino C, Costa S, Vighetti S, Tarenzi L, Rainero I, et al. Loss of expectation-related mechanisms in Alzheimer’s disease makes analgesic therapies less effective. Pain. 2006;121(1–2):133–144. doi: 10.1016/j.pain.2005.12.016.
    1. Gilmartin JF, Vaatainen S, Tormalehto S, Bell JS, Lonnroos E, Salo L, et al. Depressive symptoms are associated with analgesic use in people with Alzheimer’s disease: Kuopio ALSOVA study. PloS One. 2015;10(2):e0117926. doi: 10.1371/journal.pone.0117926.
    1. Snowden M, Sato K, Roy-Byrne P. Assessment and treatment of nursing home residents with depression or behavioral symptoms associated with dementia: a review of the literature. J Am Geriatr Soc. 2003;51(9):1305–1317. doi: 10.1046/j.1532-5415.2003.51417.x.
    1. Iden KR, Hjorleifsson S, Ruths S. Treatment decisions on antidepressants in nursing homes: a qualitative study. Scand J Prim Health Care. 2011;29(4):252–256. doi: 10.3109/02813432.2011.628240.
    1. Gulla C, Selbaek G, Flo E, Kjome R, Kirkevold O, Husebo BS. Multi-psychotropic drug prescription and the association to neuropsychiatric symptoms in three Norwegian nursing home cohorts between 2004 and 2011. BMC Geriatr. 2016;16(1):115. doi: 10.1186/s12877-016-0287-1.
    1. Chen YH, Lin LC. Ability of the pain recognition and treatment (PRT) protocol to reduce expressions of pain among institutionalized residents with dementia: a cluster randomized controlled trial. Pain Manag Nurs. 2016;17(1):14–24. doi: 10.1016/j.pmn.2015.08.003.
    1. Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. BMJ. 2015;350:h369. doi: 10.1136/bmj.h369.
    1. Gitlin LN, Kales HC, Lyketsos CG. Nonpharmacologic management of behavioral symptoms in dementia. JAMA. 2012;308(19):2020–2029. doi: 10.1001/jama.2012.36918.
    1. Kress HG. Clinical update on the pharmacology, efficacy and safety of transdermal buprenorphine. Eur J Pain. 2009;13(3):219–230. doi: 10.1016/j.ejpain.2008.04.011.
    1. Al-Tawil N, Odar-Cederlof I, Berggren AC, Johnson HE, Persson J. Pharmacokinetics of transdermal buprenorphine patch in the elderly. Eur J Clin Pharmacol. 2013;69(2):143–149. doi: 10.1007/s00228-012-1320-8.
    1. Karlsson J, Soderstrom A, Augustini BG, Berggren AC. Is buprenorphine transdermal patch equally safe and effective in younger and elderly patients with osteoarthritis-related pain? Results of an age-group controlled study. Curr Med Res Opin. 2014;30(4):575–587. doi: 10.1185/03007995.2013.873714.
    1. Sandvik R, Selbaek G, Kirkevold O, Husebo BS, Aarsland D. Analgesic prescribing patterns in Norwegian nursing homes from 2000 to 2011: trend analyses of four data samples. Age Ageing. 2016;45(1):54–60. doi: 10.1093/ageing/afv184.
    1. Jensen-Dahm C, Gasse C, Astrup A, Mortensen PB, Waldemar G. Frequent use of opioids in patients with dementia and nursing home residents: a study of the entire elderly population of Denmark. Alzheimers Dement. 2015;11(6):691–699. doi: 10.1016/j.jalz.2014.06.013.
    1. Etchells E, Darzins P, Silberfeld M, Singer PA, McKenny J, Naglie G, et al. Assessment of patient capacity to consent to treatment. J Gen Intern Med. 1999;14(1):27–34. doi: 10.1046/j.1525-1497.1999.00277.x.

Source: PubMed

3
Iratkozz fel