Skip to main content
Log in

Biosimilars und der Nocebo-Effekt

Biosimilars and the nocebo effect

  • Hot Topics
  • Published:
Zeitschrift für Rheumatologie Aims and scope Submit manuscript

Zusammenfassung

In Deutschland sind seit vielen Jahren Biosimilars zugelassen, seit wenigen Jahren auch in der Rheumatologie. Zwar haben die Biosimilars, die wie die Referenzbiologika biotechnologisch hergestellten Produkte sind, inzwischen in einigen Regionen schon erheblich Marktanteile erreicht, es gibt aber bei Patienten und Ärzten immer noch viele Zweifler, die einen Qualitätsverlust befürchten – auch, wenn es dafür keinen Anhalt gibt. Ein Teil dieses Problems ist durch den Nocebo-Effekt zu erklären, der aber auch darüber hinaus eine erhebliche medizinische Bedeutung hat. Dieser Effekt wird in diesem Artikel beschrieben und erläutert. Psychosoziale und kontextbezogene Faktoren wie die Beziehung zwischen Patient und Arzt, frühere Behandlungserfahrungen und Behandlungserwartungen können die Wirksamkeit einer therapeutischen Intervention entweder verbessern oder beeinträchtigen. Diese Phänomene werden üblicherweise als Placebo- und Nocebo-Effekte bezeichnet. Da Placebo- und Nocebo-Effekte die Symptomentwicklung, die Häufigkeit unerwünschter Ereignisse und die Wirksamkeit der Behandlung beeinflussen können, ist es entscheidend, diese Effekte zu kennen und Strategien zur Prävention zu entwickeln, um die Behandlungsergebnisse zu optimieren. Während experimentelle Studien in den letzten Jahren wesentliche Fortschritte bei der Aufklärung der psychosozialen und neurobiologischen Mechanismen von Placebo-Effekten erzielt haben, sind die detaillierten Mechanismen von Nocebo-Effekten noch weitgehend unerforscht. Ein besseres Verständnis dieser Mechanismen verspricht die Entwicklung benutzerfreundlicher Strategien für die klinische Versorgung zur Verbesserung der Behandlungsergebnisse und der Patientenzufriedenheit.

Abstract

Biosimilars have been approved for use in Germany for many years and in the meantime also in rheumatology but only a few years ago. Biosimilars, which are biotechnologically manufactured products the same as reference biologicals, have actually now achieved a substantial proportion of the market in some regions but there are still doubters among patients and physicians who fear a loss of quality even if there is no evidence for this. A part of this problem can be explained by the nocebo effect but which furthermore also has a substantial medical importance. This effect is described and explained in this article. Psychosocial and context-related factors, such as the relationship between patient and physician, previous experience with treatment and treatment expectations can either improve or impair the efficacy of treatment interventions. These phenomena are commonly known as placebo and nocebo effects. As placebo and nocebo effects can influence the development of symptoms, the frequency of undesired events and the efficacy of treatment, it is decisive to know these effects and to develop strategies for prevention in order to optimize the treatment results. Although in recent years experimental studies have achieved substantial progress in the clarification of the psychosocial and neurobiological mechanisms of placebo effects, detailed mechanisms of nocebo effects are still widely unexplored. An improved understanding of these mechanisms promises the development of user-friendly strategies for the clinical care to improve treatment results and patient satisfaction.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Literatur

  1. Weise M (2019) From bioequivalence to biosimilars: How much do regulators dare? Z Evid Fortbild Qual Gesundhwes. https://doi.org/10.1016/j.zefq.2018.12.001

    Article  PubMed  Google Scholar 

  2. Krüger K (2018) Kompendium Biosimilars Bd. 3. Thieme, Stuttgart, S 20–24

    Google Scholar 

  3. Jørgensen KK, Olsen IC, Goll GL, NOR-SWITCH study group et al (2017) Switching from originator infliximab to biosimilar CT-P13 compared with maintained treatment with originator infliximab (NOR-SWITCH): a 52-week, randomised, double-blind, non-inferiority trial. Lancet 389(10086):2304–2316

    PubMed  Google Scholar 

  4. Braun J, Lorenz HM, Müller-Ladner U et al (2018) Revised version of the statement by the DGRh on biosimilars-update 2017. Z Rheumatol 77(1):81–90

    CAS  PubMed  Google Scholar 

  5. Kay J, Schoels MM, Dörner T, Task Force on the Use of Biosimilars to Treat Rheumatological Diseases, Task Force on the Use of Biosimilars to Treat Rheumatological Diseases (2018) Consensus-based recommendations for the use of biosimilars to treat rheumatological diseases. Ann Rheum Dis 77(2):165–174 (Epub 2017 Sep 2)

    CAS  PubMed  Google Scholar 

  6. Smolen JS, Landewé R, Bijlsma J et al (2017) EULAR recommendations for the management of rheumatoid arthritis with synthetic and biological disease-modifying antirheumatic drugs: 2016 update. Ann Rheum Dis 76(6):960–977 (Epub 2017 Mar 6)

    PubMed  Google Scholar 

  7. Fiehn C, Holle J, Iking-Konert C et al (2018) S2e guideline: treatment of rheumatoid arthritis with disease-modifying drugs. Z Rheumatol 77(Suppl 2):35–53

    CAS  PubMed  Google Scholar 

  8. Kardas P, Lewek P, Matyjaszczyk M (2013) Determinants of patient adherence: a review of systematic reviews. Front Pharmacol 4:91

    PubMed  PubMed Central  Google Scholar 

  9. Howick J, Hoffmann T (2018) How placebo characteristics can influence estimates of intervention effects in trials. Cmaj 190(30):E908–E911

    PubMed  PubMed Central  Google Scholar 

  10. Häuser W et al (2012) Nocebo phenomena in medicine: their relevance in everyday clinical practice. Dtsch Arztebl Int 109(26):459–465 (Epub 2012 Jun 29). https://doi.org/10.3238/arztebl.2012.0459

    Article  PubMed  PubMed Central  Google Scholar 

  11. Jütte R (2019) Placeboforschung: Selbst eingebildete Pillen können wirken. DÄB 116(31–32):1181–1182

    Google Scholar 

  12. Pouillon L et al (2018) The nocebo effect: a clinical challenge in the era of biosimilars. Expert Rev Clin Immunol 14(9):739–749 (Epub 2018 Aug 30)

    CAS  PubMed  Google Scholar 

  13. Kristensen LE, Alten R, Puig L et al (2018) Non-pharmacological effects in switching medication: the Nocebo effect in switching from originator to biosimilar agent. BioDrugs 32(5):397–404

    PubMed  PubMed Central  Google Scholar 

  14. Odinet JS, Day CE, Cruz JL, Heindel GA (2018) The biosimilar Nocebo effect ? A systematic review of double-blinded versus open-label studies. J Manag Care Spec Pharm 24(10):952–959

    PubMed  Google Scholar 

  15. Petrie KJ, Rief W (2019) Psychobiological mechanisms of placebo and Nocebo effects: pathways to improve treatments and reduce side effects. Annu Rev Psychol 70:599–625 (Epub 2018 Aug 15)

    PubMed  Google Scholar 

  16. Evers AWM, Colloca L, Blease C et al (2018) Implications of placebo and Nocebo effects for clinical practice: expert consensus. Psychother Psychosom 87(4):204–210 (Epub 2018 Jun 12)

    PubMed  Google Scholar 

  17. Kennedy WP (1961) The Nocebo reaction. Med World 95:203–205

    CAS  PubMed  Google Scholar 

  18. Kessner S, Wiech K, Forkmann K, Ploner M, Bingel U (2013) The effect of treatment history on therapeutic outcome: an experimental approach. JAMA Intern Med 173:1468–1469

    PubMed  Google Scholar 

  19. Gupta A, Thompson D, Whitehouse A, Collier T, Dahlof B et al (2017) Adverse events associated with unblinded, but not with blinded, statin therapy in the Anglo-Scandinavian Cardiac Outcomes Trial-Lipid-Lowering Arm (ASCOT-LLA): a randomised double-blind placebo-controlled trial and its nonrandomized non-blind extension phase. Lancet 389(10088):2473–2481

    CAS  PubMed  Google Scholar 

  20. Mahr A, Golmard C, Pham E, Iordache L, Deville L, Faure P (2017) Types, frequencies, and burden of nonspecific adverse events of drugs: analysis of randomized placebo-controlled clinical trials. Pharmacoepidemiol Drug Saf 26:731–741

    CAS  PubMed  Google Scholar 

  21. Clark PI, Leaverton PE (1994) Scientific and ethical issues in the use of the placebo control in clinical trials. Annu Rev Public Health 15:19–38

    CAS  PubMed  Google Scholar 

  22. Barsky AJ, Saintfort R, Rogers MP, Borus FJ (2002) Nonspecific medication side effects and the nocebo phenomenon. Jama 287:622–627

    PubMed  Google Scholar 

  23. Petrie KJ, Sivertsen B, Hysing M, Broadbent E, Moss-Morris R et al (2001) Thoroughly modern worries: the relationship of worries about modernity to reported symptoms, health, and medical care utilization. J Psychosom Res 51:395–401

    CAS  PubMed  Google Scholar 

  24. Rief W, Glaesmer H, Baehr V, Broadbent E et al (2012) The relationship of modern health worries to depression, symptom reporting and quality of life in a general population survey. J Psychosom Res 72:318–320

    PubMed  Google Scholar 

  25. Rubin GJ, Hillert L, Nieto-Hernandez R et al (2011) Do people with idiopathic environmental intolerance attributed to electromagnetic fields display physiological effects when exposed to electromagnetic fields? A systematic review of provocation studies. Bioelectromagnetics 32:593–609

    PubMed  Google Scholar 

  26. Chapman S, George SA, Waller K, Cakic V (2013) The pattern of complaints about Australian wind farms does not match the establishment and distribution of turbines: support for the psychogenic, “communicated disease” hypothesis. Plos One 8:e76584

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Crichton F, Chapman S, Cundy T, Petrie KJ (2014) The link between health complaints and wind turbines: support for the Nocebo expectations hypothesis. Front Public Health 2:220

    PubMed  PubMed Central  Google Scholar 

  28. Lionetti E, Pulvirenti A, Vallorani M, Catassi G, Verma AK et al (2017) Re-challenge studies in non-celiac gluten sensitivity: a systematic review and meta-analysis. Front Physiol 8:621

    PubMed  PubMed Central  Google Scholar 

  29. Freeman S, Yu R, Egorova N, Chen X, Kirsch I, Claggett B, Kaptchuk TJ, Gollub RL, Kong J (2015) Distinct neural representations of placebo and Nocebo effects. Neuroimage 112:197–207 (Epub 2015 Mar 14)

    PubMed  Google Scholar 

  30. Kirsch I (1985) Response expectancy as a determinant of experience and behaviour. Am Psychol 40:1189–1202

    Google Scholar 

  31. Myers MG, Cairns JA, Singer J (1987) The consent form as a possible cause of side effects. Clin Pharmacol Ther 42:250–253

    CAS  PubMed  Google Scholar 

  32. Nestoriuc Y, Orav EJ, Liang M, Horne R, Barsky AJ (2010) Prediction of nonspecific side effects in rheumatoid arthritis patients by beliefs about medicines. Arthritis Care Res 62:791–799

    Google Scholar 

  33. Nestoriuc Y, von Blanckenburg P, Schuricht F, Barsky AJ, Hadji P et al (2016) 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 27:1909–1915

    CAS  PubMed  Google Scholar 

  34. Webster RK, Weinman J, Rubin GJ (2018) Medicine related beliefs predict attribution of symptoms to a sham medicine: a prospective study. Br J Health Psychol 23:436–454

    PubMed  PubMed Central  Google Scholar 

  35. Petrie KJ, Moss-Morris R, Grey C, Shaw M (2004) The relationship of negative affect and perceived sensitivity to symptom reporting following vaccination. Br J Health Psychol 9:101–111

    PubMed  Google Scholar 

  36. Faasse K, Grey A, Horne R, Petrie KJ (2015) High perceived sensitivity to medicines is associated with higher medical care utilisation, increased symptom reporting and greater information-seeking about medication. Pharmacoepidemiol Drug Saf 24:592–599

    PubMed  Google Scholar 

  37. Horne R, Faasse K, Cooper V et al (2013) Personal sensitivity to medicines scale. Br J Health Psychol 18:18–30

    PubMed  Google Scholar 

  38. Rief W, Nestoriuc Y, Weiss S et al (2009) Meta-analysis of the placebo response in antidepressant trials. J Affect Disord 118:1–8

    CAS  PubMed  Google Scholar 

  39. Amanzio M, Corazzini LL, Vase L, Benedetti F (2009) A systematic review of adverse events in placebo groups of anti-migraine clinical trials. Pain 146:261–269

    CAS  PubMed  Google Scholar 

  40. Colgan S, Faasse K, Martin LR, Stephens MH, Grey A et al (2015) Perceptions of generic medication in the general population, doctors and pharmacists: a systematic review. Bmj Open 5:e8915

    PubMed  PubMed Central  Google Scholar 

  41. Faasse K, Cundy T, Gamble G, Petrie KJ (2013) The effect of an apparent change to a branded or generic medication on drug effectiveness and side effects. Psychosom Med 75:90–96

    CAS  PubMed  Google Scholar 

  42. Boone NW, Liu L, Romberg-Camps MJ et al (2018) The Nocebo effect challenges the non-medical infliximab switch in practice. Eur J Clin Pharmacol 74:655–661

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Weissenfeld J, Stock S, Lungen M, Gerber A (2010) The Nocebo effect: a reason for patients’ non-adherence to generic substitution? Pharmazie 65:451–456

    CAS  PubMed  Google Scholar 

  44. Bartley H, Faasse K, Horne R, Petrie KJ (2016) You can’t always get what you want: the influence of choice on Nocebo and placebo responding. Ann Behav Med 50:445–451

    PubMed  Google Scholar 

  45. Vögtle E, Barke A, Kröner-Herwig B (2013) Nocebo hyperalgesia induced by social observational learning. Pain 154:1427–1433

    PubMed  Google Scholar 

  46. Faasse K, Petrie KJ (2016) From me to you: the effect of social modelling on treatment outcomes. Curr Dir Psychol Sci 25:438–443

    Google Scholar 

  47. Lorber W, Mazzoni G, Kirsch I (2007) Illness by suggestion: expectancy, modeling, and gender in the production of psychosomatic symptoms. Ann Behav Med 33:112–116

    PubMed  Google Scholar 

  48. Witthöft M, Rubin GJ (2013) Are media warnings about the adverse health effects of modern life self-fulfilling? An experimental study on idiopathic environmental intolerance attributed to electromagnetic fields (IEIEMF). J Psychosom Res 74:206–212

    PubMed  Google Scholar 

  49. Bräscher AK, Raymaekers K, Van den Bergh O, Witthöft M (2017) Are media reports able to cause somatic symptoms attributed to WiFi radiation? An experimental test of the negative expectation hypothesis. Environ Res 156:265–271

    PubMed  Google Scholar 

  50. Faasse K, Cundy T, Petrie KJ (2010) Thyroxine: anatomy of a health scare. BMJ 340:20–21

    Google Scholar 

  51. Faasse K, Gamble G, Cundy T, Petrie KJ (2012) Impact of television coverage on the number and type of symptoms reported during a health scare: a retrospective pre-post observational study. Bmj Open 2:e1607

    PubMed  PubMed Central  Google Scholar 

  52. Roscoe JA, Morrow GR, Aapro MS, Molassiotis A, Olver I (2011) Anticipatory nausea and vomiting. Support Care Cancer 19:1533–1538

    PubMed  Google Scholar 

  53. Liccardi G, Senna G, Russo M et al (2004) Evaluation of the nocebo effect during oral challenge in patients with adverse drug reactions. J Investig Allergol Clin Immunol 14:104–107

    CAS  PubMed  Google Scholar 

  54. van den Bergh O, Stegen K, Van Diest I, Raes C, Stulens P et al (1999) Acquisition and extinction of somatic symptoms in response to odours: a Pavlovian paradigm relevant to multiple chemical sensitivity. Occup Environ Med 56:295–301

    PubMed  PubMed Central  Google Scholar 

  55. Stewart-Williams S, Podd J (2004) The placebo effect: dissolving the expectancy versus conditioning debate. Psychol Bull 130:324–340

    PubMed  Google Scholar 

  56. Petersen GL, Finnerup NB, Colloca L, Amanzio M, Price DD et al (2014) The magnitude of the Nocebo effects in pain: a meta-analysis. Pain 155:1426–1434

    PubMed  PubMed Central  Google Scholar 

  57. Petrie KJ, Faasse K, Crichton F, Grey A (2014) How common are symptoms? Evidence from a New Zealand national telephone survey. Bmj Open 4(6):e5374

    PubMed  PubMed Central  Google Scholar 

  58. Tan K, Petrie KJ, Faasse K, Bolland MJ, Grey A (2014) Unhelpful information about adverse drug reactions. Bmj 349:g5019

    PubMed  Google Scholar 

  59. Rief W, Avorn J, Barsky AJ (2006) Medication-attributed adverse effects in placebo groups: implications for assessment of adverse effects. Arch Intern Med 166:155–160

    PubMed  Google Scholar 

  60. Faasse K, Petrie KJ (2013) The Nocebo effect: patient expectations and medication side effects. Postgrad Med J 89:540–546

    PubMed  Google Scholar 

  61. Feldman PJ, Cohen S, Doyle W, Skoner DP, Gwaltner JM (1999) The impact of personality on the reporting of unfounded symptoms and illness. Journal of Personality and Social Psychology 77:370–378

    CAS  PubMed  Google Scholar 

  62. Watson D, Pennebaker JW (1989) Health complaints, stress, and distress: exploring the central role of negative affectivity. Psychol Rev 96:234–254

    CAS  PubMed  Google Scholar 

  63. Wells RE, Kaptchuk TJ (2012) To tell the truth, the whole truth, may do patients harm: the problem of the nocebo effect for informed consent. Am J Bioeth 12:22–29

    PubMed  PubMed Central  Google Scholar 

  64. Miller FG (2012) Clarifying the Nocebo effect and its ethical implications. Am J Bioeth 12(3):30–39

    CAS  PubMed  Google Scholar 

  65. Bromwich D (2012) Plenty to worry about: consent, control and anxiety. Am J Bioeth 12(3):35–36

    PubMed  Google Scholar 

  66. Bingel U (2014) Avoiding nocebo effects to optimize treatment outcome. Jama 312:693–694

    CAS  PubMed  Google Scholar 

  67. O’Connor AM, Pennie RA, Dales RE (1996) Framing effects on expectations, decisions and side effects experienced: the case of influenza immunization. J Clin Epidemiol 49:1271–1276

    PubMed  Google Scholar 

  68. Crichton F, Petrie KJ (2015) Health complaints and wind turbines: the efficacy of explaining the Nocebo response to reduce symptom reporting. Environ Res 140:449–455

    CAS  PubMed  Google Scholar 

  69. Scherlinger M, Langlois E, Germain V, Schaeverbeke T (2018) Acceptance rate and sociological factors involved in the switch from originator to biosimilar etanercept (SB4). Semin Arthritis Rheum. https://doi.org/10.1016/j.semarthrit.2018.07.005

    Article  PubMed  Google Scholar 

  70. Gasteiger C, Jones ASK, Kleinstäuber M et al (2019) Arthritis Care Res (Hoboken). https://doi.org/10.1002/acr.24012

    Article  Google Scholar 

  71. Rief W, Shedden-Mora MC, Laferton JA et al (2017) Preoperative optimization of patient expectations improves long-term outcome in heart surgery patients: results of the randomized controlled PSY-HEART trial. BMC Med 15(1):4

    PubMed  PubMed Central  Google Scholar 

  72. Carvalho C, Caetano JM, Cunha L et al (2016) Open-label placebo treatment in chronic low back pain: a randomized controlled trial. Pain 157:2766–2772

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Kirchhof J, Petrakova L, Brinkhoff A et al (2018) Learned immunosuppressive placebo responses in renal transplant patients. PNAS 115(16):4223–4227

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Irwin MR, Cole SW (2011) Reciprocal regulation of the neural and innate immune systems. Nat Rev Immunol 11:625–632

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Pacheco-López G, Engler H, Niemi MB, Schedlowski M (2006) Expectations and associations that heal: Immunomodulatory placebo effects and its neurobiology. Brain Behav Immun 20:430–446

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to U. Kiltz.

Ethics declarations

Interessenkonflikt

J. Braun hat Honorare für Vorträge, Advisory boards, bezahlte Beratungen und finanzielle Unterstützung für Studien von Abbvie (Abbott), Amgen, Baxter, Biogen, BMS, Boehringer, Celgene, Celltrion, Centocor, Chugai, Hexal, Janssen, Lilly, Medac, MSD (Schering-Plough), Mylan, Mundipharma, Novartis, Pfizer (Wyeth, Hospira), Roche, Sanofi-Aventis und UCB erhalten. U. Kiltz hat Honorare für Vorträge, Advisory boards, bezahlte Beratungen und finanzielle Unterstützung für Studien von AbbVie, Biocad, Chugai, Eli Lilly, Grünenthal, Janssen, MSD, Novartis, onkowissen.de, Pfizer, Roche und UCB sowie Forschungsunterstützung (unrestricted grant) von Abbvie, Biogen, Novartis und Pfizer erhalten. X. Baraliakos hat Beraterhonorare, Forschungsunterstützung und Bildungszuschüsse von AbbVie, BMS, Celgene, Chugai, Galapagos, Janssen, Lilly, MSD, Novartis, Mylan, Pfizer, Sandoz und UCB erhalten. B. Buehring hat Honorare für Vorträge, Advisory boards, bezahlte Beratungen von GE/Lunar, Kinemed, Janssen, UCB, Lilly, AbbVie und Gilead erhalten. I. Andreica hat Berater- und/oder Referentenhonorare von den Firmen AbbVie, Chugai, Eli Lilly, MSD, Novartis und Pfizer erhalten. D. Kiefer hat Honorare für Vorträge und advisory boards von Abbvie, Chugai/Roche, Janssen, Merck, Novartis, Sandoz und UCB erhalten. S. Tsiami gibt an, dass kein Interessenkonflikt besteht.

Additional information

Redaktion

U. Müller-Ladner, Bad Nauheim

U. Lange, Bad Nauheim

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Braun, J., Tsiami, S., Buehring, B. et al. Biosimilars und der Nocebo-Effekt. Z Rheumatol 79, 267–275 (2020). https://doi.org/10.1007/s00393-019-00729-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00393-019-00729-7

Schlüsselwörter

Keywords

Navigation