Minimizing Drug Adverse Events by Informing About the Nocebo Effect-An Experimental Study

Yiqi Pan, Timm Kinitz, Marin Stapic, Yvonne Nestoriuc, Yiqi Pan, Timm Kinitz, Marin Stapic, Yvonne Nestoriuc

Abstract

Relevance: Informing patients about potential adverse events as part of the informed consent may facilitate the development of nocebo-driven drug adverse events (nocebo side effects). Objective: To investigate whether informing about the nocebo effect using a short information sheet can reduce nocebo side effects. Methods: A total of N = 44 participants with weekly headaches for at least 6 months were recruited using the cover story of a clinical trial for a headache medicine. In reality, all participants took a placebo pill and were randomized to the nocebo information group or the standard leaflet group. Participants were instructed to read the bogus medication leaflet entailing side effects information shortly before pill intake. The nocebo group additionally received an explanation about the nocebo effect as part of the leaflet. Questionnaires were completed at baseline, 2 min, and 4 days after the pill intake. We conducted general linear models with bootstrap sampling. Baseline symptoms were included as a covariate. Results: Most participants (70.5%) reported nocebo side effects at 2 min. Participants who received the nocebo information (n = 24) reported less nocebo symptoms than the control group (n = 20) (estimated difference: 3.3, BCa 95% CI [1.14; 5.15], p = 0.01, Cohen's d = 0.59). Baseline symptoms, perceived sensitivity to medicine, and side effect expectations each moderated the group effect (estimated difference in slope: 0.47, BCa 95% CI [0.19; 0.73], p = 0.001, d = 0.75; 1.07 [0.27; 1.61], p = 0.006, d = 0.73; 1.57 [0.38; 2.76], p = 0.02, d = 0.58). No group differences were found at 4-day follow-up. After revealing the actual aim of the study, 86% of the participants evaluated the nocebo information to be helpful in general. Conclusions: Results provide the first evidence that informing about the nocebo effect can reduce nocebo side effects.

Keywords: drug safety information; inert exposure; informed consent; nocebo effect; patient education; predictors; risk factors; side effects.

Figures

Figure 1
Figure 1
Moderators of the intervention. The panels (A to E) show the candidate moderators on the x-axis: baseline symptoms, monitoring cognitive coping style, trait-anxiety, perceived sensitivity to medicine, and side effect expectations. For each panel, the primary outcome nocebo side effects is shown on the y-axis.The relationship between each moderator and nocebo side effects by intervention group (nocebo information: n = 24; standard leaflet: n = 20) are presented using estimates of general linear models with bootstrap sampling, adjusted for baseline symptoms. Bias-corrected and accelerated 95% confidence intervals are portrayed as upper and lower boundaries. For interaction effects, log-likelihood tests comparing each model with the intercept-only model are shown in the upper left area. *p < 0.05, **p < 0.01.

References

    1. Bingel U, Wanigasekera V, Wiech K, Ni Mhuircheartaigh R, Lee MC, Ploner M, et al. The effect of treatment expectation on drug efficacy: imaging the analgesic benefit of the opioid remifentanil. Sci Transl Med (2011) 3(70):70ra14. 10.1126/scitranslmed.3001244
    1. Aslaksen PM, Zwarg ML, Eilertsen HI, Gorecka MM, Bjorkedal E. Opposite effects of the same drug: reversal of topical analgesia by nocebo information. Pain (2015) 156(1):39–46. 10.1016/j.pain.0000000000000004
    1. Barsky AJ, Saintfort R, Rogers MP, Borus JF. Nonspecific medication side effects and the nocebo phenomenon. JAMA (2002) 287(5):622–7. 10.1001/jama.287.5.622
    1. Neukirch N, Colagiuri B. The placebo effect, sleep difficulty, and side effects: a balanced placebo model. J Behav Med (2015) 38(2):273–83. 10.1007/s10865-014-9590-5
    1. Kaptchuk TJ, Stason WB, Davis RB, Legedza AR, Schnyer RN, Kerr CE, et al. Sham device v inert pill: randomised controlled trial of two placebo treatments. BMJ (2006) 332 :391–7. 10.1136/bmj.38726.603310.55
    1. Link J, Haggard R, Kelly K, Forrer D. Placebo/nocebo symptom reporting in a sham herbal supplement trial. Eval Health Prof (2006) 29(4):394–406. 10.1177/0163278706293403
    1. Amanzio M, Corazzini LL, Vase L, Benedetti F. A systematic review of adverse events in placebo groups of anti-migraine clinical trials. Pain (2009) 146(3):261–9. 10.1016/j.pain.2009.07.010
    1. Mitsikostas DD. Nocebo in headache. Curr Opin Neurol (2016) 29(3):331–6. 10.1097/WCO.0000000000000313
    1. Mitsikostas DD, Mantonakis LI, Chalarakis NG. Nocebo is the enemy, not placebo. A meta-analysis of reported side effects after placebo treatment in headaches. Cephalalgia (2011) 31(5):550–61. 10.1177/0333102410391485
    1. Cepeda MS, Lobanov V, Berlin JA. Use of to estimate condition-specific nocebo effects and other factors affecting outcomes of analgesic trials. J Pain (2013) 14(4):405–11. 10.1016/j.jpain.2012.12.011
    1. Rief W, Nestoriuc Y, Weiss S, Welzel E, Barsky AJ, Hofmann SG. Meta-analysis of the placebo response in antidepressant trials. J Affect Disord (2009) 118(1–3):1–8. 10.1016/j.jad.2009.01.029
    1. Sohl SJ, Schnur JB, Montgomery GH. A meta-analysis of the relationship between response expectancies and cancer treatment-related side effects. J Pain Symptom Manage (2009) 38(5):775–84. 10.1016/j.jpainsymman.2009.01.008
    1. Nestoriuc Y, von Blanckenburg P, Schuricht F, Barsky AJ, Hadji P, Albert US, et al. Is it best to expect the worst? Influence of patients’ side-effect expectations on endocrine treatment outcome in a 2-year prospective clinical cohort study. Ann Oncol (2016) 27(10):1909–15. 10.1093/annonc/mdw266
    1. Colagiuri B, Dhillon H, Butow PN, Jansen J, Cox K, Jacquet J. Does assessing patients’ expectancies about chemotherapy side effects influence their occurrence? J Pain Symptom Manage (2013) 46(2):275–81. 10.1016/j.jpainsymman.2012.07.013
    1. Jose J, AlHajri L. Potential negative impact of informing patients about medication side effects: a systematic review. Int J Clin Pharm (2018) 40(4):806–22. 10.1007/s11096-018-0716-7
    1. Myers MG, Cairns JA, Singer J. The consent form as a possible cause of side-effects. Clin Pharmacol Ther (1987) 42(3):250–3. 10.1038/clpt.1987.142
    1. Cocco G. Erectile dysfunction after therapy with metoprolol: the Hawthorne effect. Cardiology (2009) 112(3):174–7. 10.1159/000147951
    1. Mondaini N, Gontero P, Giubilei G, Lombardi G, Cai T, Gavazzi A, et al. Finasteride 5 mg and sexual side effects: how many of these are related to a nocebo phenomenon? J Sex Med (2007) 4(6):1708–12. 10.1111/j.1743-6109.2007.00563.x
    1. Silvestri A, Galetta P, Cerquetani E, Marazzi G, Patrizi R, Fini M, et al. Report of erectile dysfunction after therapy with beta-blockers is related to patient knowledge of side effects and is reversed by placebo. Eur Heart J (2003) 24(21):1928–32. 10.1016/j.ehj.2003.08.016
    1. Howland JS, Baker MG, Poe T. Does patient education cause side effects? A controlled trial. J Fam Pract (1990) 31(1):62–4.
    1. Lamb GC, Green SS, Heron J. Can physicians warn patients of potential side effects without fear of causing those side effects? Arch Intern Med (1994) 154(23):2753–6. 10.1001/archinte.1994.00420230150018
    1. Morris LA, Kanouse DE. Informing patients about drug side-effects. J Behav Med (1982) 5(3):363–73. 10.1007/BF00846163
    1. Mahr A, Golmard C, Pham E, Iordache L, Deville L, Faure P. Types, frequencies, and burden of nonspecific adverse events of drugs: analysis of randomized placebo-controlled clinical trials. Pharmacoepidemiol Drug Saf (2017) 26(7):731–41. 10.1002/pds.4169
    1. Iuga AO, McGuire MJ. Adherence and health care costs. Risk Manag Healthc Policy (2014) 7:35–44. 10.2147/RMHP.S19801
    1. Stark RG, John J, Leidl R. Health care use and costs of adverse drug events emerging from outpatient treatment in Germany: a modelling approach. BMC Health Serv Res (2011) 11:9. 10.1186/1472-6963-11-9
    1. Wells RE, Kaptchuk TJ. To tell the truth, the whole truth, may do patients harm: the problem of the nocebo effect for informed consent. Am J Bioeth (2012) 12(3):22–9. 10.1080/15265161.2011.652798
    1. Colloca L, Miller FG. The nocebo effect and its relevance for clinical practice. Psychosom Med (2011) 73(7):598–603. 10.1097/PSY.0b013e3182294a50
    1. Bingel U. Placebo Competence Team. Avoiding nocebo effects to optimize treatment outcome. JAMA (2014) 312(7):693–4. 10.1001/jama.2014.8342
    1. Tversky A, Kahneman D. The framing of decisions and the psychology of choice. Science (1981) 211 :453–8. 10.1126/science.7455683
    1. O’Connor AM, Pennie RA, Dales RE. Framing effects on expectations, decisions, and side effects experienced: the case of influenza immunization. J Clin Epidemiol (1996) 49(11):1271–6. 10.1016/S0895-4356(96)00177-1
    1. Wilhelm M, Rief W, Doering BK. Decreasing the burden of side effects through positive message framing: an experimental proof-of-concept study. Int J Behav Med (2018) 25(4):381–9. 10.1007/s12529-018-9726-z
    1. Heisig SR, Shedden-Mora MC, Hidalgo P, Nestoriuc Y. Framing and personalizing informed consent to prevent negative expectations: an experimental pilot study. Health Psychol (2015) 34(10):1033–7. 10.1037/hea0000217
    1. Faasse K, Huynh A, Pearson S, Geers AL, Helfer SG, Colagiuri B. The influence of side effect information framing on nocebo effects. Ann Behav Med (2019) 53(7):621–9. 10.1093/abm/kay071
    1. Caplandies FC, Colagiuri B, Helfer SG, Geers AL. Effect type but not attribute framing alters nocebo headaches in an experimental paradigm. Psychol Consciousness Theor Res Pract (2017) 4(3):259–73. 10.1037/cns0000130
    1. Webster RK, Weinman J, Rubin GJ. Positively framed risk information in patient information leaflets reduces side effect reporting: a double-blind randomized controlled trial. Ann Behav Med (2018) 52(11):920–9. 10.1093/abm/kax064
    1. Seminowicz DA, Mikulis DJ, Davis KD. Cognitive modulation of pain-related brain responses depends on behavioral strategy. Pain (2004) 112(1–2):48–58. 10.1016/j.pain.2004.07.027
    1. Brown RJ. Psychological mechanisms of medically unexplained symptoms: an integrative conceptual model. Psychol Bull (2004) 130(5):793–812. 10.1037/0033-2909.130.5.793
    1. Crichton F, Petrie KJ. Health complaints and wind turbines: the efficacy of explaining the nocebo response to reduce symptom reporting. Environ Res (2015) 140:449–55. 10.1016/j.envres.2015.04.016
    1. Quidde J, Pan Y, Salm M, Hendi A, Nilsson S, Oechsle K, et al. Preventing adverse events of chemotherapy by educating patients about the nocebo effect (RENNO study)— study protocol of a randomized controlled trial with gastrointestinal cancer patients. BMC Cancer (2018) 18(1):916. 10.1186/s12885-018-4814-7
    1. Vambheim SM, Flaten MA. A systematic review of sex differences in the placebo and the nocebo effect. J Pain Res (2017) 10:1831–9. 10.2147/JPR.S134745
    1. Faasse K, Grey A, Horne R, Petrie KJ. High perceived sensitivity to medicines is associated with higher medical care utilisation, increased symptom reporting and greater information-seeking about medication. Pharmacoepidemiol Drug Saf (2015) 24(6):592–9. 10.1002/pds.3751
    1. Symon A, Williams B, Adelasoye QA, Cheyne H. Nocebo and the potential harm of ‘high risk’ labelling: a scoping review. J Adv Nurs (2015) 71(7):1518–29. 10.1111/jan.12637
    1. Webster RK, Weinman J, Rubin GJ. A systematic review of factors that contribute to nocebo effects. Health Psychol (2016) 35(12):1334–55. 10.1037/hea0000416
    1. Planes S, Villier C, Mallaret M. The nocebo effect of drugs. Pharmacol Res Perspect (2016) 4(2):e00208. 10.1002/prp2.208
    1. Muris P, van Zuuren F. Monitoring, medical fears and physical symptoms. Br J Clin Psychol (1992) 31(Pt 3):360–2. 10.1111/j.2044-8260.1992.tb01006.x
    1. Rodriguez-Raecke R, Doganci B, Breimhorst M, Stankewitz A, Buchel C, Birklein F, et al. Insular cortex activity is associated with effects of negative expectation on nociceptive long-term habituation. J Neurosci (2010) 30(34):11363–8. 10.1523/JNEUROSCI.2197-10.2010
    1. Ellerbrock I, Wiehler A, Arndt M, May A. Nocebo context modulates long-term habituation to heat pain and influences functional connectivity of the operculum. Pain (2015) 156(11):2222–33. 10.1097/j.pain.0000000000000297
    1. Zimmermann T, Heinrichs N. Seite an Seite—eine Krebserkrankung in der Partnerschaft gemeinsam bewältigen. Ein Ratgeber für Paare. Göttingen: Hogrefe; (2008).
    1. Rief W, Barsky AJ, Glombiewski JA, Nestoriuc Y, Glaesmer H, Braehler E. Assessing general side effects in clinical trials: reference data from the general population. Pharmacoepidemiol Drug Saf (2011) 20(4):405–15. 10.1002/pds.2067
    1. Reuter U, del Rio MS, Carpay JA, Boes CJ, Silberstein SD, Program GHM. Placebo adverse events in headache trials: headache as an adverse event of placebo. Cephalalgia (2003) 23(7):496–503. 10.1046/j.1468-2982.2003.00530.x
    1. Horne R, Faasse K, Cooper V, Diefenbach MA, Leventhal H, Leventhal E, et al. The perceived sensitivity to medicines (PSM) scale: an evaluation of validity and reliability. Br J Health Psychol (2013) 18(1):18–30. 10.1111/j.2044-8287.2012.02071.x
    1. Laux L, Glanzmann PS CD. State Trait Angstinventar (STAI). Weinheim: Beltz Testgesellschaft; (1981).
    1. van Zuuren FJ, de Groot KI, Mulder NL, Muris P. Coping with medical threat: an evaluation of the threatening medical situations inventory (TMSI). Pers Individ Dif (1996) 21(1):21–31. 10.1016/0191-8869(96)00029-3
    1. European Medicines Agency Guideline on adjustment for baseline covariates in clinical trials (2015) [06–06–2019]. Available from: .
    1. Field A, Miles J, Field Z. Discovering Statistics using R. Thousand Oaks, California: SAGE Publications Ltd; (2012). 992 p.
    1. Brookes ST, Whitely E, Egger M, Smith GD, Mulheran PA, Peters TJ. Subgroup analyses in randomized trials: risks of subgroup-specific analyses; power and sample size for the interaction test. J Clin Epidemiol (2004) 57(3):229–36. 10.1016/j.jclinepi.2003.08.009
    1. van Hoorn R, Tummers M, Booth A, Gerhardus A, Rehfuess E, Hind D, et al. The development of CHAMP: a checklist for the appraisal of moderators and predictors. BMC Med Res Methodol (2017) 17(1):173. 10.1186/s12874-017-0451-0
    1. Feingold A. Effect sizes for growth-modeling analysis for controlled clinical trials in the same metric as for classical analysis. Psychol Methods (2009) 14(1):43–53. 10.1037/a0014699
    1. Colloca L, Petrovic P, Wager TD, Ingvar M, Benedetti F. How the number of learning trials affects placebo and nocebo responses. Pain (2010) 151(2):430–9. 10.1016/j.pain.2010.08.007
    1. Berna C, Kirsch I, Zion SR, Lee YC, Jensen KB, Sadler P, et al. Side effects can enhance treatment response through expectancy effects: an experimental analgesic randomized controlled trial. Pain (2017) 158(6):1014–20. 10.1097/j.pain.0000000000000870
    1. Barsky AJ. The iatrogenic potential of the physician’s words. JAMA (2017) 318(24):2425–6. 10.1001/jama.2017.16216
    1. Corsi N, Colloca L. Placebo and nocebo effects: the advantage of measuring expectations and psychological factors. Front Psychol (2017) 8:308. 10.3389/fpsyg.2017.00308
    1. Corsi N, Emadi Andani M, Tinazzi M, Fiorio M. Changes in perception of treatment efficacy are associated to the magnitude of the nocebo effect and to personality traits. Sci Rep (2016) 6:30671. 10.1038/srep30671
    1. Liccardi G, Senna G, Russo M, Bonadonna P, Crivellaro M, Dama A, et al. Evaluation of the nocebo effect during oral challenge in patients with adverse drug reactions. J Investig Allergol Clin Immunol (2004) 14(2):104–7
    1. Heller MK, Chapman SC, Horne R. Beliefs about medication predict the misattribution of a common symptom as a medication side effect—evidence from an analogue online study. J Psychosom Res (2015) 79(6):519–29. 10.1016/j.jpsychores.2015.10.003
    1. Henningsen P, Gundel H, Kop WJ, Lowe B, Martin A, Rief W, et al. Persistent physical symptoms as perceptual dysregulation: a neuropsychobehavioral model and its clinical implications. Psychosom Med (2018) 80(5):422–31. 10.1097/PSY.0000000000000588
    1. Wiech K. Deconstructing the sensation of pain: the influence of cognitive processes on pain perception. Science (2016) 354 :584–7. 10.1126/science.aaf8933
    1. Malone CD, Bhowmick A, Wachholtz AB. Migraine: treatments, comorbidities, and quality of life, in the USA. J Pain Res (2015) 8:537–47. 10.2147/JPR.S88207
    1. Papanikolaou PN, Churchill R, Wahlbeck K, Ioannidis JP. Safety reporting in randomized trials of mental health interventions. Am J Psychiatry (2004) 161(9):1692–7. 10.1176/appi.ajp.161.9.1692
    1. Zorzela L, Golder S, Liu Y, Pilkington K, Hartling L, Joffe A, et al. Quality of reporting in systematic reviews of adverse events: systematic review. BMJ (2014) 348:f7668. 10.1136/bmj.f7668
    1. Roewer N, Thiel H. Anästhesiologische Pharmakotherapie. 3 ed. Stuttgart: Thieme; (2014).
    1. Derry S, Wiffen PJ, Moore RA. Aspirin for acute treatment of episodic tension-type headache in adults. Cochrane Database Syst Rev (2017) 1:CD011888. 10.1002/14651858.CD011888.pub2
    1. Rabbie R, Derry S, Moore RA. Ibuprofen with or without an antiemetic for acute migraine headaches in adults. Cochrane Database Syst Rev (2013) (4), CD008039. 10.1002/14651858.CD008039.pub3
    1. Reicherts P, Gerdes AB, Pauli P, Wieser MJ. Psychological placebo and nocebo effects on pain rely on expectation and previous experience. J Pain (2016) 17(2):203–14. 10.1016/j.jpain.2015.10.010

Source: PubMed

3
Abonnieren