Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
Kwartaalbericht 3e kwartaal 2014 16 juli 2014 Contents Voorwoord 3 1. Observations 4 1.1. Doxycycline and skin discoloration 4 1.2. Statins and lichenoid drug eruption 10 1.3. Statins and muscle rupture 16 1.4. Tamsulosin and urinary incontinence 22 1.5. Prednisolone and hiccups 27 1.6. Atovaquone/ proguanil hydrochloride and psychotic disorder 31 1.7. Ciclosporin and posterior reversible encephalopathy syndrome 36 2. Publications 43 2 Voorwoord Waakzaamheid Klaar om op te stijgen van London City airport staan we gereed op de startbaan. Het toestel maakt snelheid en draait dan plotseling rechts de baan af. Vanuit het raampje zie ik een ander vliegtuig dezelfde baan naderen. Blijkbaar moest deze toch nog vrijgemaakt worden voor dit vliegtuig dat de landing al ingezet had. Een voorbeeld van een noodzakelijke correctie van een procedure die al in gang gezet was. Niet zozeer een voorbeeld van foutief handelen dus, maar juist van een normale voor iedereen acceptabele maatregel die de veiligheid vergroot. Een parallel met de geneesmiddelenbewaking is snel gelegd. Ook hier moeten we soms veiligheidsmaatregelen nemen die horen bij het continue proces van afwegen van de balans tussen baten en risico’s van geneesmiddelen en waar iedereen zijn voordeel mee doet; kleine correcties die erger kunnen voorkomen. Enkele voorbeelden van zaken die een mogelijke aanpassing in de voorlichting en waarschuwing van het gebruik van geneesmiddelen behoeven, treft u ook weer aan in dit kwartaalbericht. Farmacovigilance heeft tot doel het gebruik van geneesmiddelen veiliger te maken. Correcties en voorzorgsmaatregelen horen hier bij. “Viglantia” (lat.) betekent dan ook zowel waakzaamheid als zorg. Precies wat de verkeersleiding in Londen deed, waakzaam zijn en zorgen voor de veiligheid. Mijn medepassagiers hadden van het voorval niets gemerkt; ze zaten ontspannen te wachten op het vertrek. Eugène van Puijenbroek 3 1. Observations 1.1. Doxycycline and skin discoloration Introduction Doxycycline is an antibiotic belonging to the group of tetracyclines. It is active against gram positive and gram negative bacteria. It has been on the Dutch market at least since 1973 and the oral preparation is indicated for infections of the respiratory tract, infections of the urogenital tract, infections of the skin and soft tissues, Borrelia burgdorfori infections, infections of the gastrointestinal tract and eye infection in particular trachoma [1-4]. Photodermatitis or photosensitivity is mentioned as an adverse drug reaction in the SmPC of doxycycline. The SmPC does not mention skin discoloration or skin hyperpigmentation [1-4]. Melanocytes are derived embryonically from neural crest cells that migrate into the basal layer of the epidermis. In the skin, melanocytes continuously produce melanosomes, organelles that are transferred to keratinocytes. The melanosomes convert tyrosine to melanin, giving skin its color. Under the stimulus of hormones or irritation, the production of melanosomes increases, leading to hyperpigmentation. In response to sun exposure or idiopathically in some disorders, activation of melanocytes occurs and causes hyperpigmentation. The same melanocyte concentration is present in persons of all races who have normal skin. However, some races have larger melanosomes, giving their skin a darker colour [5]. Drug-induced pigmentation represents 10 to 20% of all cases of acquired hyperpigmentation [6]. Overall, several mechanisms for drug-induced pigmentation have so far been described [6,7] and these mechanisms are not mutually exclusive. Firstly, the accumulation of melanin, either free in the dermis or contained mainly within cutaneous cells particularly the dermal macrophages rather than in the basal layer of the epidermis. This melanin accumulation usually results from either hyperproduction by epidermal melanocytes specifically stimulated by the medication or in response to a nonspecific cutaneous inflammation linked to the drug itself. This mechanism is very often worsened by sun exposure. Secondly, the accumulation of the triggering medication itself, without any association to melanin, usually appearing as granules freely scattered among extracellular matrix elements or included within dermal macrophages that are unable to eliminate these foreign bodies. Thirdly, the synthesis of special pigments, such as lipofuscin, probably under the direct influence of the drug. Lastly, the deposition of iron, usually resulting from drug-induced damage to dermal vessels with leakage of red blood cells and subsequent lysis throughout the dermis [6]. Reports The Netherlands Pharmacovigilance Centre Lareb received 5 reports of skin discoloration/hyperpigmentation associated with the use of doxycycline in a period from 01-10-1999 until 21-06-2013. The reports are listed in table 1. Table 1. Reports of skin and nail discoloration associated with the use of doxycycline Patient, Sex, Age Drug Indication for use Concomitant medication Suspected adverse drug reaction Time to onset, Action with drug outcome 4 Patient, Sex, Age Drug Indication for use Concomitant medication Suspected adverse drug reaction Time to onset, Action with drug outcome A 156211 M, 71 years and older Specialist doctor doxycycline 3 dd 100 mg Chronic Q-fever and infected vessel prothesis acetylsalicylic skin acid, hyperpigmentation esomeprazole, levothyroxine sodium, nebivolol, atorvastatin 9 months discontinued not recovered B 156212 M, 71 years and older Specialist doctor doxycycline, about 4 months 200 mg daily, the next 6 months 300 mg daily Chronic Q-fever acetylsalicylic hyperpigmentation acid, omeprazole, skin ferrous fumarate, celecoxib, tamsulosine with dutasteride, prednisolone, azathioprine, hydroxychloroquine, simvastatin. 10 months discontinued not recovered C 102886 F, 61-70 years Pharmacist doxycycline 100mg start 2dd, furthermore once daily 100 mg Respiratory infection discoloration skin 2 days discontinued recovered D 51212 F, 31-40 years General Practitioner doxycycline 100mg perindopril 1dd Lower respiratory tract infection hyperpigmentation skin 2 weeks discontinued unknown E 60201 M, 61-70 years Pharmacist doxycycline 100mg 1dd Infection photosensitivity reaction, pigmentation abnormal 9 days after start, 2 days after cessation discontinued not recovered Additional information about the cases is described below: Cases A and B were reported by the same Specialist doctor. In case C it is described that the skin was black as coal and smooth, there was no itching or any other symptom. The patient has a light tinted skin color. The latency period described in this case is short, so possibly sun exposure is a causative factor. In case D the hyperpigmentation of the skin was located in the face. There were brown, not sharply defined, maculae on the forehead. The patient does not have a history of skin disorders or naevi and she is not pregnant. In case E the patient went on a holiday in the sun two days after doxycycline was withdrawn. During the course the patient stayed out of the sun but it was unknown to him that he should do so when the course had ended. He now has severe and lasting hyperpigmentation on his nose. In addition, Lareb received three reports of nail discoloration associated with the use of doxycycline. In two cases the patient also suffered from onycholysis. These cases are described in Quarterly Report 2013-2 ‘Doxycycline and photoonycholysis – an update’ [8] as Case A (Reportnumber 25961) and case J (Reportnumber 139748). Additional report 71069 from a pharmacist concerns a female aged 51-60 years years, with nail discolouration following administration of doxycycline (dose twice daily 100 mg) for Lyme’s disease with a latency of two months after start. Doxycycline was withdrawn. The patient outcome is unknown. 5 Because we could not be certain that the nail discoloration is not a result of the onycholysis in the first two cases, the focus of this signal was placed on discoloration of the skin and not the nails. Other sources of information SmPC Photodermatitis or photosensitivity is mentioned as an adverse drug reaction in the SmPC of doxycycline. The SmPC does not mention skin or nail hyperpigmentation [1-4]. Literature Hyperpigmentation of the oral cavity (teeth, mucosa, alveolar bone), skin, nails, eyes, thyroid and even bone has been reported due to minocycline intake [9-11]. Also for other tetracyclines hyperpigmentation of the skin has been described, albeit to a lesser extent than for minocycline [12-14]. Pichardo et al. [14] reported on a 44-year old man who had been treated with doxycycline for three years, 100 mg twice daily for chronic follicular conjunctivitis. For the last six months he suffered from progressive, symmetric blue-gray periocular discoloration. A biopsy from lesional skin showed granular deposits of a brown to black pigment in the superficial dermis. Eight months after cessation of doxycycline, the patient had almost completely recovered. Nail discoloration induced by doxycycline has also been described. Akcam et al. [15] report on an 11-year-old-boy with nail discoloration caused by doxycycline intake who was referred to their hospital for evaluation. The history revealed that, in April 2004, the patient had brucellosis that was treated with doxycycline 200 mg on the first day and 100 mg daily thereafter, combined with gentamicin for 10 days. Doxycycline therapy was stopped because he developed photosensitivity. The symptoms of brucellosis resolved, but brown discoloration of the nails developed after 15 days of doxycycline intake. Physical examination revealed painless brown discoloration of the fingernails. The oral cavity and teeth had no changes in colour. The laboratory findings revealed normal hematologic and biochemical results. The nail discoloration disappeared in one month. Databases Table 2. Reports of skin discolouration/hyperpigmention with doxycycline in the databases of the Netherlands Pharmacovigilance Centre Lareb and the WHO- and Eudravigilance (EMA) database [16,17]. Database Preferred Terms Number of reports ROR (95% CI) Lareb Skin hyperpigmentation 3 3.6 (1.1-11.4) Skin discolouration 1 - Pigmentation disorder 1 - Total 5 1.2 (0.5-3.0) Skin hyperpigmentation 17 3.6 (2.2-5.7) Skin discolouration 45 1.3 (1.0-1.8) Pigmentation disorder 14 3.9 (2.3-6.7) Total 76 1.8 (1.5-2.3) Skin hyperpigmentation 8 8.5 (4.2 – 17.1) Skin discolouration 19 3.6 (2.3 – 5.6) Pigmentation disorder 5 4.4 (1.8 – 10.6) WHO Eudravigilance 6 Database Preferred Terms Number of reports ROR (95% CI) Total 32 4.4 (3.1 – 6.2) Prescription data Table 3. Number of patients using doxycycline in the Netherlands between 2009 and 2013 [18]. Drug 2009 2010 2011 2012 2013 Doxycycline 965,820 933,230 878,230 819,780 739,860 Mechanism Skin hyperpigmentation induced by minocycline is a well-recognized side effect but has rarely been reported for other tetracyclines. Several types of minocyclineinduced hyperpigmentation of the skin have been distinguished based on distinct clinical features and their histopathologic correlates. The first type is characterized by blue-black pigmentation within areas of scars and previously inflamed skin; in the second type, blue-gray pigment spots develop in normal skin, especially on the shins and arms; and the third type is characterized by a muddy-brown generalized pigmentation, mostly accentuated in sun-exposed areas [6,19]. This third type is likely caused by increased melanin or melanin– minocycline complexes at the dermal–epidermal interface [9]. Patient E reported to Lareb the hyperpigmentation occurred after exposure to the sun, which is also described for the third type of minocycline induced skin discoloration. Based on a previously reported unusual case of chronic doxycycline abuse of twelve years in a psychotic patient [13], Böhm et al. [19] have investigated the nature of the observed pigment changes in the same patient. The histomorphologic and ultrastructural changes induced by doxycycline shared several features with cutaneous hyperpigmentation caused by minocycline. The biophysical findings further suggest a direct deposition of doxycycline, probably chelated with iron and/or calcium, within the lesional skin. The authors mention that in the present case a prolonged suprapharmacologic dosage of doxycycline was used, but that physicians should watch for pigment changes in patients receiving long-term therapy. Discussion and conclusion The association between doxycycline and skin hyperpigmentation has been reported to Lareb 5 times and is supported by a statistically significant disproportionality in the WHO- and Eudravigilance database, as well as cases in the literature [13,14] and a possible pharmacological mechanism [19]. In addition, Lareb received three reports of nail discoloration associated with doxycycline use. However, two of these patients also suffered from onycholysis and it’s unclear if this was also the cause of the discoloration. Nail discoloration induced by doxycycline has also been described [15]. Risk factors for tetracycline-induce pigmentary changes include the duration of treatment, the cumulative dose (risk high above 50g) the presence of previous skin alterations related to inflammation or excessive sun exposure or the concomitant intake of other pigmentation- inducing medications [6]. Böhm et al. [19] have speculated on the reason for the apparently much higher incidence of hyperpigmentation caused by minocycline compared with doxycycline. Minocycline is the classic antibiotic for patients with acne or rosacea and may be prescribed for several years. Doxycycline is more commonly used for acute bacterial infections is usually given for shorter periods. However, case A and B reported to Lareb used doxycycline chronically for months. 7 According to Böhm et al. [19] the apparently higher incidence of pigment changes in patients taking minocycline may also be the result of differences in the chemical structures of minocycline and doxycycline. Doxycycline has a hydroxyl group at position 5 and a methyl group at position 6, whereas minocycline carries a para-N, N-dimethylamino group at position 7 of the naphthacene carboxamide ring. These structural differences create a number of changes in the physicochemical properties of both tetracyclines. For instance, minocycline is twice as lipophilic as doxycycline and penetrates more easily in tissues. For minocycline it has been described that pigmentation of the skin and nails may require months to years to fade after discontinuation of the drug, and other sites may remain permanently discoloured [10]. For doxycycline Lareb has also received cases of patients who had not recovered from the hyperpigmentation at the moment of reporting. Based on the available information for this association, skin hyperpigmentation should be explicitly mentioned in the SmPC of doxycycline in addition to the photodermatitis/photosensitivity reactions. Skin discoloration should be mentioned in the SmPC of doxycycline References 1. Dutch SmPC Doxycycline 100 mg PCH, omhulde tabletten 100 mg. (version date: 12-4-2012, access date: 16-1-2013) http://db.cbg-meb.nl/IB-teksten/h09519.pdf. 2. Dutch SmPC Doxycycline Actavis Disper 100 mg, tabletten. (version date: 29-11-2006, access date: 16-1-2013) http://db.cbg-meb.nl/IB-teksten/h12871.pdf. 3. Dutch SmPC Doxycycline dispergeerbaar ratiopharm 100 mg, tabletten. (version date: 21-5-2012, access date: 16-1-2013) http://db.cbg-meb.nl/IB-teksten/h16491.pdf. 4. Dutch SmPC Efracea, capsules met gereguleerde afgifte, hard 40 mg. (version date: 21-7-0012, access date: 16-1-2013) http://db.cbg-meb.nl/IB-teksten/h33759.pdf. 5. Stulberg DL, Clark N, Tovey D. Common hyperpigmentation disorders in adults: Part II. Melanoma, seborrheic keratoses, acanthosis nigricans, melasma, diabetic dermopathy, tinea versicolor, and postinflammatory hyperpigmentation. Am.Fam.Physician 2003;68(10):1963-8. 6. Dereure O. Drug-induced skin pigmentation. Epidemiology, diagnosis and treatment. Am.J.Clin.Dermatol. 2001;2(4):253-62. 7. Butler, D. F. Drug-Induced Pigmentation. (version date: 28-9-2012, access date: 15-4-2014) http://emedicine.medscape.com/article/1069686-overview#a0104. 8. The Netherlands Pharmacovigilance Centre Lareb. Doxycycline and photo-onycholysis – an update. (version date: 1-6-2013, access date: 15-4-2014) http://www.lareb.nl/Signalen/KWB_2013_2_doxycycline_photo-onycholysis_WEB.aspx. 9. Tavares J, Leung WW. Discoloration of nail beds and skin from minocycline. CMAJ. 2011;183(2):224 10. Eisen D, Hakim MD. Minocycline-induced pigmentation. Incidence, prevention and management. Drug Saf 1998;18(6):431-40. 11. Pandit S, Hadden W. Black pigmentation of bone due to long-term minocycline use. Surgeon. 2004;2(4):236-7. 12. Hawfield W, Goodrich R, Warren S, Morrell D. Trauma-induced cutaneous pigmentation from tetracycline: a case report. Pediatr.Dermatol. 2004;21(2):164-6. 13. Westermann GW, Bohm M, Bonsmann G, Rahn KH, Kisters K. Chronic intoxication by doxycycline use for more than 12 years. J.Intern.Med. 1999;246(6):591-2. 14. Pichardo RO, Yeatts RP, Sangueza OP. Doxycycline-inducted Cutaneous Hyperpigmentation. [Abstract] American Journal of Dermatopathology: 2006;28(3):325 15. Akcam M, Artan R, Akcam FZ, Yilmaz A. Nail discoloration induced by doxycycline. Pediatr.Infect.Dis.J. 2005;24(9):845-6. 16. Uppsala Monitoring Centre. WHO Global Individual Case Safety Reports database (Vigibase). (version date: 2014, access date: 23-4-2014) https://tools.who-umc.org/webroot/ (access restricted). 17. European medicines Agency. Eudravigilance database. (version date: 2014, access date: 23-42014) http://bi.eudra.org (access restricted). 8 18. College for Health Insurances. GIP database. (version date: 9-6-2009, access date: 16-3-2011) http://www.gipdatabank.nl/index.asp?scherm=tabellenFrameSet&infoType=g&tabel=01basis&item=J01FF. 19. Bohm M, Schmidt PF, Lodding B, Uphoff H, Westermann G, Luger TA, Bonsmann G, Metze D. Cutaneous hyperpigmentation induced by doxycycline: histochemical and ultrastructural examination, laser microprobe mass analysis, and cathodoluminescence. Am.J.Dermatopathol. 2002;24(4):345-50. 9 1.2. Statins and lichenoid drug eruption Introduction Statins inhibit the enzyme HMG-CoA (3-hydroxy-methylglutaryl-coenzyme Areductase, which plays an important role in the synthesis of cholesterol by catalysing the conversion from HMG-CoA to mevalonate. Statins are indicated for hypercholesterolemia. They are effective in both the primary and the secondary prevention of ischemic heart diseases and stroke prevention. Among the statins simvastatin (Zocor®) was granted marketing authorization in the eighties, atorvastatin (Lipitor®), pravastatin (Selektine®) and fluvastatin (Lescol®) in the nineties, rosuvastatin (Crestor®) in 2002 and pitavastatin (Vezepra®, Livazo®) in 2010. Common adverse skin reactions in this group are urticaria, eczema, dermatitis, skin eruption, pruritus, alopecia and angioneurotic edema. Beside these, several serious skin reactions are described [1-7]. Lichen planus (LP) is an inflammatory, pruritic disease of the skin and mucous membranes, which can be either generalized or localized. It is characterized by distinctive purplish, flat-topped papules having a predilection for the trunk and flexor surfaces. On the surface often white stripes (Wickham’s striae) might be visible. The lesions may be discrete or coalesce to form plaques. Histologically, there is a "saw-tooth" pattern of epidermal hyperplasia and vacuolar alteration of the basal layer of the epidermis along with an intense upper dermal inflammatory infiltrate composed predominantly of T-cells. The etiology is unknown. It occurs in the general population at a rate of 0.9-1.2 % and oral lesions may be seen in 3070 % of these patients. It affects men and women almost equally and it is likely to start in middle age. It is diagnosed on clinical symptoms and biopsy can confirm the diagnosis. It is a self-limiting disease, but recovery might be slow an remission occurs in 1-2 years; oral lichen seems to follow a more chronic course, with a mean duration of 4.5 years [8,9]. Drug induced lichenoid eruptions (LDE)* can differ in clinical (and histological) aspects from lichen planus; next to lichenoid elements, LDE may be accompanied with papular, scaling and eczematous lesions. The predilection sites are rarely involved. On the other hand LDE produces lesions that might be clinically and histologically indistinguishable from idiopathic LP. The two conditions can be differentiated only by the time course of skin or mucous membrane involvement in relation to drug administration and by re-challenging with the suspected agent. Frequently the lichenoid eruptions occur a few months after starting the drug, but the latency may very between days to several years. Clearance of symptoms can occur within a few weeks after withdrawal of the drug; in some reports the healing period stretched from less than a week to many months. In some single case reports, symptoms cleared without discontinuation of the drug, in another study with oral LDE patients did not recover after withdrawal of the drug. Similar as in idiopathic lichen planus LDE results in hyperpigmentation, which regresses slowly or even can be irreversible. Histologic differences between LP an LDE are often subtle and not reliable. The dermal infiltrate (as well as peripheral blood) may contain eosinophils and plasma cells and may sometimes be distinguished from the infiltrate in LP [8,10]. * A reliable differentiation between lichenoid drug eruption an drug induced lichen planus cannot be made, therefore the term lichenoid drug eruptions (LDE) is used [8]. The current observation describes the association between statins and lichenoid drug eruptions in 13 patients. A previous report regarding this association in 4 patients has been sent to the Medical Evaluation Board in 2004 [11]. 10 Reports Lareb received 13 reports of lichen planus or lichenoid dermatitis associated with the use of statins, in a period from July 18, 1996 till April 3, 2014. The reports are listed in Table 1. In one patient (D) biopsy results confirmed the diagnosis. In four other cases (B,E,I,L) the diagnosis was confirmed by a dermatologist. Table 1. Reports of lichenoid drug eruptions associated with the use of statins Patient, Number, Sex, Age, Source Drug, daily dose Indication for use A 14687 F, 51-60 years Hospital Pharmacist simvastatin, 20mg daily pure hypercholesterolaemia B 35066 F, 51-60 years Pharmacist atorvastatin 20mg daily C 37816 M, 61-70 years General Practitioner Concomitant Medication Suspected adverse drug reaction Time to onset, Action with drug outcome dermatitis lichenoid unknown (”soon”) no change recovering carbasalate calcium ibuprofen lichen planus 12-14 weeks discontinued not yet recovered atorvastatin 20mg daily pure hypercholesterolaemia carbasalate calcium diclofenac nitroglycerin isosorbide dinitrate metoprolol dermatitis lichenoid 2 weeks no change unknown D 43561 M or F General Practitioner simvastatin 10mg daily diazepam diclofenac therapeutic response unexpected with drug substitution, dermatitis lichenoid unknown discontinued recovered E 48214 F, 51-60 years Specialist doctor simvastatin 20mg daily pure hypercholesterolaemia lichen planus 15 years no change unknown F 53337 F, 51-60 years Pharmacist atorvastatin 40mg daily hypercholesterolaemia ezetimibe 10mg hypercholesterolaemia furosemide, captopril acetylsalicylic acid esomeprazole metoprolol nitroclycerin transdermal betahistin lichen planus 6 years G 119392 F, 71 years and older Pharmacist simvastatin 20mg daily hypercholesterolaemia macrogol/ electrolytes vitamin B1, B6, B12, lamotrigine, miconazole vaginal oxazepam omeprazole, metoprolol telmisartan/hydrochlorothiazide sucralfate lichen planus 2-3 months no change not recovered H 129411 F, 51-60 years atorvastatin 20mg daily hypercholesterolaemia lichen planus 3 months discontinued recovering 4 months both discontinued recovering 11 Consumer I 133942 F, 51-60 years Consumer simvastatin 10 mg daily hypercholesterolaemia metformin irbesartan lichen planus < 2 years discontinued recovering J 136378 F, 61-70 years Pharmacist simvastatin 20mg daily hypercholesterolaemia omeprazole enalapril amlodipine lichen planus on lower legs and arms 18 months discontinued not yet recovered* K 146716 M, 61-70 years Pharmacist simvastatin 20 mg daily hypercholesteraemia dermatitis lichenoid 6-7 years discontinued unknown** L 148414 F, 61-70 years Specialist doctor (student) atorvastatin 20 mg daily hypercholesterolaemia enalapril triamterene/hydrochl orothiazide 50/25 metoprolol dermatitis lichenoid 1 week discontinued recovered simvastatin 20 mg daily hypercholesterolaemia bisoprolol fluticasone carbasalate calcium ranitidin lichen planus 1 year discontinued recovering M 172669 F, 51-60 years Pharmacist * Report sent at date of discontinuation **Patient switched to atorvastatin Other sources of information SmPC Lichenoid drug eruptions, including lichen planus, are not mentioned in the SmPCs of atorvastatin, fluvastatin, pravastatin, rosuvastatin, simvastatin and pitavastatin [1-7]. Literature Case reports of lichenoid drug eruptions have been described in association with simvastatin [12,13] pravastatin [14,15], fluvastatin and lovastatin [16]. Roger et al. described a case of a 57-year-old woman with a pruritic, erythematous eruption of polygonal papules on her wrists and elbows one month after starting simvastatin 10 mg once daily. She used no concomitant medication. Patient did not recover after treatment with a topical corticosteroid. Simvastatin has been withdrawn and the rash resolved four weeks later. The rash reassembled a lichenoid drug eruption and histopathology was also compatible with a lichenoid eruption [12]. Stoebner et al. described a 63-year-old man, who was treated with simvastatin for hypercholesterolemia. After one month he developed a pruriginous and bullous lichenoid eruption. Histological and direct immunofluorescent features were consistent with the diagnosis of lichen planus pemphigoides. After two months use, simvastatin was discontinued. He was treated with a topical class II corticosteroid during one week. Four weeks hereafter the itching had vanished and no new lesions had appeared. Three months later he had recovered with only some hyperpigmentated sequelae [13]. Pravastatin-induced LDE was first reported in a 75-year-old black patient who developed a lichenoid rash, on the extensor surfaces of the arms and dorsal 12 aspect of the hands, 3 weeks after starting pravastatin at a dose of 10 mg daily. Clinical examination revealed multiple scattered, polygonal, erythematous plaques and papules with a translucent, shiny scale and the diagnosis of a lichenoid drug eruption was confirmed on histopathological examination of a skin biopsy. Discontinuation of pravastatin led to resolution with mild postinflammatory hyperpigmentation in two weeks’ time. Upon re-challenge, the rash reappeared within 1 week [14]. A case of pravastatin-induced diffuse and numerous pigmented macules on the face and upper back was described in a 64 year old woman. Histopathological examination showed a lichenoid dermatitis. Three months before, pravastatin had been added to her treatment regime, because of an acute coronary syndrome. Cessation of pravastatin resulted in gradual fading of pigmentation over a 9 months period. All other chronic medications, including furosemide and candesartan – which both can cause also LDE- had been continued [15]. Sebök described a case of a 59-year old woman with bilateral pruritic, papulous eruption on the dorsal side of her forearms, hands and on the volar surfaces of her wrists, on the soles and to a lesser extent on her trunk and thighs four weeks after starting fluvastatin. Topical steroids had only a temporary effect. The antihypertensive treatment, perindopril, was continued. After discontinuation of fluvastatin and treatment with mometason-furoate, the lesions cleared after three weeks. Two weeks later, lovastatin was introduced, because of an increase in cholesterol levels. The eruption recurred. Biopsy showed a lichenoid dermatitis. After stopping lovastatin, recovery was observed within three weeks, leaving postinflammatory hyperpigmented macules [16]. Databases Table 2. Reports of lichen planus/lichenoid dermatitis with statins in the databases of the Netherlands Pharmacovigilance Centre Lareb, the WHO- and Eudravigilance (EMA) database [17,18]. Database Preferred Terms Number of reports ROR (95% CI) Lareb Lichen planus 8 4.3 (2.3-10.4) Lichenoid dermatitis* 5 5.7 (2.2-15.1) WHO Lichen planus 91 5.3 (4.3-6.6) Eudravigilance Lichen planus 31 3.2 (2.3 -4.7) *Lichenoid dermatitis is the Lower Level Term; the corresponding MedDRA Preferred Term Lichenoid keratosis is not appropriate Prescription data Table 3. Number of patients using statins in the Netherlands between 2009 and 2013 [19]. Drug 2009 2010 2011 2012 2013 simvastatin 826,190 930,700 980,250 pravastatin 164,470 170,080 169,640 170,760 171,410 fluvastatin 23,113 22,181 21,667 21,474 20,937 1,044,000 1,084,000 13 Drug 2009 2010 2011 atorvastatin 394,750 375,810 rosuvastatin 179,010 Total 2012 2013 364,900 383,670 412,580 181,230 191,890 209,550 221,690 1.507.000 1.594.000 1.662.000 1.751.000 1.827.000 Mechanism The pathogenic mechanism of LDE is not well understood, a type IV allergy is sometimes involved. A dose dependency is suggested. Some drugs change surface antigens, whereas other drugs change enzyme systems. These aberrations may precipitate an immune response, in which cytotoxic CD8+ T cells are activated, which then cause epidermal damage [8,10]. For statins no specific mechanism is described. Discussion and conclusion Lareb has received 13 reports of lichen planus and lichenoid dermatitis in association with statins; in one case a lichenoid dermatitis occurred after drug substitution of simvastatin and in one case also ezetimibe was reported as suspected drug. In one patient (D) biopsy results confirmed the diagnosis. In four other cases (B,E,I,L) reporting was done by a dermatologist or patients had been referred to a dermatologist, therefore a reliable diagnosis was assumed. As is described by Ellgehausen it is difficult to distinguish LDE form idiopathic lichen planus, clinically as well as histologically [8]. The time course in relation to drug administration of the suspected agent might be of help, but may also vary between days and years. In the reported cases to Lareb, the latency in most patients was several weeks. According to Ellgehausen, the clearance of symptoms in LDE might vary from a week to many months or can be irreversible. Therefore idiopathic lichen planus cannot be ruled out easily. Of the thirteen cases reported to Lareb, five had recovered or were recovering after discontinuation of the statins at the moment of reporting; in one case also the suspected ezetimibe was discontinued. Four of these patients had also been treated with either topical corticosteroids, light treatment or acupuncture. Recovery took weeks to several months. In three of these patients several concomitant medications were used, which are known to be associated with lichenenoid eruptions as well, including bisoprolol, metoprolol, hydrochlorothiazide and/or enalapril/captopril. These medications were however continued during the recovering phase, which strengthens an association with the statin. The association was supported by the WHO- and Eudravigilance data and by several publications; in one of these even a positive re-challenge was observed after re-introduction of a statin. It is of importance to acknowledge the possible role of statins in a patient with lichenoid eruption, which might have a major influence on well-being. Discontinuation of these statins might result in a substantial improvement in symptoms. For this reason, it is suggested statins might have a causative role in the occurrence of lichenoid drug eruption. 14 Lichenoid drug eruptions should be mentioned in the SmPC of statins References 1. Dutch SmPC Lescol®. (version date: 1-11-2013, access date: 3-4-2014) http://db.cbg-meb.nl/IBteksten/h18719.pdf. 2. Dutch SmPC Zocor®. (version date: 31-12-2013, access date: 3-4-2014) http://db.cbg-meb.nl/IBteksten/h13193.pdf. 3. Dutch SmPC Lipitor®. (version date: 2-1-2014, access date: 3-4-2014) http://db.cbg-meb.nl/IBteksten/h21081.pdf. 4. Dutch SmPC Selektine®. (version date: 30-4-2013, access date: 3-4-2014) http://db.cbgmeb.nl/IB-teksten/h13755.pdf. 5. Dutch SmPC Crestor®. (version date: 23-12-2013, access date: 3-4-2014) http://db.cbg-meb.nl/IBteksten/h26872.pdf. 6. Dutch SmPC Vezepra®. (version date: 19-8-2012, access date: 22-4-2014) http://db.cbgmeb.nl/IB-teksten/h103422.pdf. 7. Dutch SmPC Livazo®. (version date: 19-8-2012, access date: 22-4-2014) http://db.cbg-meb.nl/IBteksten/h103768.pdf. 8. Ellgehausen P, Elsner P, Burg G. Drug-induced lichen planus. Clin.Dermatol. 1998;16(3):325-32. 9. Thompson DF, Skaehill PA. Drug-induced lichen planus. Pharmacotherapy 1994;14(5):561-71. 10. Mulder WMC, editor. Side effects in dermatology. Naarden: IMP; 2009. 11. Netherlands Pharmacovigilance Centre Lareb. HMG-CoA-reductase inhibitors and lichenoid eruption. (version date: 2004, access date: 22-4-2014) http://www.lareb.nl/Signalen/kwb_2004_2_stati2. 12. Roger D, Rolle F, Labrousse F, Brosset A, Bonnetblanc JM. Simvastatin-induced lichenoid drug eruption. Clin.Exp.Dermatol. 1994;19(1):88-9. 13. Stoebner PE, Michot C, Ligeron C, Durand L, Meynadier J, Meunier L. [Simvastatin-induced lichen planus pemphigoides]. Ann.Dermatol.Venereol. 2003;130(2 Pt 1):187-90. 14. Keough GC, Richardson TT, Grabski WJ. Pravastatin-induced lichenoid drug eruption. Cutis 1998;61(2):98-100. 15. Pua VS, Scolyer RA, Barnetson RS. Pravastatin-induced lichenoid drug eruption. Australas.J.Dermatol. 2006;47(1):57-9. 16. Sebok B, Toth M, Anga B, Harangi F, Schneider I. Lichenoid drug eruption with HMG-CoA reductase inhibitors (fluvastatin and lovastatin). Acta Derm.Venereol. 2004;84(3):229-30. 17. WHO Global Individual Case Safety Reports database (Vigibase). (version date: 2014, access date: 23-4-2014) https://tools.who-umc.org/webroot/ (access restricted). 18. Eudravigilance database. (version date: 2014, access date: 23-4-2014) http://bi.eudra.org (access restricted). 19. College for health insurances. GIP database. (version date: 7-3-2014, access date: 15-4-2014) http://www.gipdatabank.nl/. 15 1.3. Statins and muscle rupture Introduction Statins inhibit the enzyme 3-hydroxy-methylglutaryl-co-enzyme A-reductase (HMG-CoA-reductase), which plays an essential role in the synthesis of cholesterol by catalysing the conversion from HMG-CoA to mevalonate [1]. Statins are indicated for hypercholesterolemia. They are effective in both the primary and the secondary prevention of ischemic heart diseases and stroke prevention [2-4]. Over the past decades, several statins have been granted marketing authorization in the Netherlands, including simvastatin (Zocor®), pravastatin (Selektine®), fluvastatin (Lescol®), atorvastatin (Lipitor®), rosuvastatin (Crestor®) and more recently pitavastatin (Vezepra®, Livazo®) [1]. One of the most important and well-known ADRs of statins are the musculoskeletal ADRs; including myalgia, muscle cramp, myopathy, myositis and rhabdomyolysis [1,5-11]. It is important to differentiate myalgia from myopathy and myositis. Although myopathy and myositis may cause myalgia, most individuals with myalgia have neither [12]. The risk for myopathy is increased by higher doses, predisposing factors (renal failure, hypothyroidism, personal or family history of hereditary muscular disorders, history of muscular toxicity caused by a statin or fibrate, history of liver disease and/or alcohol abuse, female sex, and age >65 years) and in combination with other drugs, in particular fibrates [1]. A muscle rupture is a contraction-induced injury in which muscle fibres tear. It mostly occurs as a result of a powerful eccentric contraction or overstretching of the muscle and is therefore a typical injury during explosive movements, such as sprinting, lunging or jumping [13]. Historically, acute muscle injuries have been classified as strains (grade I), partial tears (grade II) and complete tears (grade III) [14]. Spontaneous muscle ruptures that occur without intense muscle contraction are very rare. Reports The Netherlands Pharmacovigilance Centre Lareb received 11 reports on muscle rupture associated with the use of statins, in the period from 22 February 2006 until 12 January 2014. The reports are listed in Table 1. Table 1. Reports of muscle rupture associated with the use of statins. Patient, Number, Sex, Age, Source Drug, daily dose Indication for use Concomitant Medication Suspected adverse drug reaction Time to onset, Action with drug, outcome A, 166606, M, 61-70 years, physician simvastatin, 1dd 40mg, secondary prevention CVA baclofen, clopidogrel, lisinopril, valsartan, tamsulosin, hydrochlorothiazide, venlafaxine, nifedipine muscle rupture 15 month, withdrawn, recovering B, 158137, F, 61-70 years, physician simvastatin, 1dd 40mg, hypercholesterola emia salmeterol/fluticas one, calcium carbonate/colecalc iferol, fluticasone, zolpidem, pramipexole, muscle rupture 8 months, withdrawn, unknown 16 omeprazole, oxazepam, ibuprofen C, 154672, F, 51-60 years, physician rosuvastatin, 1dd 10mg, hypercholesterola emia pramipexole muscle rupture 7 months, withdrawn, recovering D, 152410, M, 41-50 years, consumer atorvastatin, 1dd 20mg, cardiovascular disorder metoprolol muscle rupture, back pain, myalgia, liver enzyme abnormal, stools abnormal 1 month, withdrawn, recovered E, 115957, M, 61-70 years, pharmacist atorvastatin, 1dd 10mg, cardiac arrhythmia phenprocoumon, digoxin, metoprolol muscle rupture 5 years, continued, recovered F, 128372, F, 71 years and older, consumer pravastatin, 1dd 20mg, hypertension ezetimib muscle rupture, myalgia 4 months, withdrawn, unknown G, 86755, M, 51-60 years, nurse pravastatin, 1dd 10mg, hypercholesterola emia muscle rupture, tendon rupture within 1 month, withdrawn, recovered with sequel H, 83259, M, 61-70 years, physician rosuvastatin, 1dd 10mg, hypercholesterola emia ofloxacine, 1dd 400mg, prostatitis muscle rupture, drug interaction 2 years / 3 days, continued / withdrawn, unknown I, 70663, M, physician fluvastatin 20mg muscle rupture 7 years, withdrawn, unknown J, 58917, M, 41-50 years, specialist doctor rosuvastatin 1dd 40mg, hypercholesterola emia muscle rupture months, withdrawn, recovered K, 55786, M, 61-70 years, specialist doctor fluvastatin, hypercholesterola emia, diltiazem, coronary artery disease muscle rupture, tendon rupture 5 years, withdrawn, unknown omeprazole, metoprolol, ramipril, lercanidipine, hydrochlorothiazide/valsartan, povidone acetylsalicylic acid, quinapril Case A describes a patient with first a rupture of his left biceps and second a rupture of his right biceps. The patient was a highly trained athlete in the period before the ruptures occurred. Case B describes a partial rupture of the biceps muscle. Case C describes a rupture of the calf muscle. Within several months the patient experienced three spontaneous muscle ruptures of the calf in both legs. The muscle rupture did not occur during exercise or lunging, but during normal daily activities. The earlier muscle ruptures were misdiagnosed as thrombophlebitis. This report involves the third rupture, which was a rupture of the medial head of the gastrocnemius muscle. This was confirmed by a physiotherapist. In the period of the occurrence of the first muscle rupture, creatine kinase (CK) level was 123 U/L. Four months later the CK level was 190 U/L. Again one month later, at the 17 moment of the third muscle rupture, CK level was 156 U/L. CK levels after the withdrawal of rosuvastatin were unknown. Case D describes a muscle rupture during exercise. During earlier treatment with simvastatin the patient was suffering from musculoskeletal pain and had increased liver enzymes. Case E describes a rupture of the hamstring. The patient twice experienced a hamstring injury in the past. It is unknown if the patient was treated with statins during the occurrence of the previous hamstring injuries. Case F describes a spontaneous rupture of the right biceps and rupture of a knee ligament. Case G describes a patient who was treated with atorvastatin prior to the treatment with pravastatin. Case H describes a rupture of the right calf muscle that occurred spontaneously while standing. In this case it cannot ruled out that the muscle rupture is caused by the treatment with ofloxacine. Case I describes that the deterioration continued after withdrawal of fluvastatin. Later the situation stabilised. Case J describes an obese patient (BMI: 32.8 kg/m 2) who experienced three muscle ruptures during treatment with rosuvastatin. The patient suffered from myalgia during earlier treatment with atorvastatin. The patient was physically active. At the moment atorvastatin was withdrawn and rosuvastatin was started CK levels were 212 U/L. At the moment of the first muscle rupture the CK level was 310 U/L. In the month that rosuvastatin was withdrawn the CK level was 121 U/L. Case K describes a patient with a rupture of muscles and tendons of both biceps and both quadriceps. CK levels were 88 U/L, which is within the normal range of 20-200 U/L for men, nine months after the withdrawal of fluvastatin. In 2 cases (B and C) the reporter explicitly stated that the rupture was not a rupture of the tendon. In two cases (D and J) exercise could have played a role in the occurrence of the muscle rupture. In five cases (B, E, G, I and K) it was unknown if the muscle rupture occurred during exercise or during normal daily activities. In four cases (A, C, F and H) it was mentioned that the muscle rupture occurred spontaneously during normal daily activities. It is remarkable that only two reports (D and F) mentioned myalgia as an ADR. Other sources of information SmPC The SmPCs of statins available on the Dutch market do not mention muscle rupture as an ADR [5-9]. However they do mention, except for fluvastatin [7] and pitavastatin [10,11], tendon rupture as a possible ADR [5,6,8,9]. The US SPCs of statins available on the US market do not mention muscle rupture as a possible ADR [15-21]. Only the US SmPC of atorvastatin mentions tendon rupture as a possible ADR [16]. Literature Myotoxicity can occur during treatment with statins. Possible ADRs that are described in literature are myalgia, muscle cramps, myositis, rhabdomyolysis and increased serum levels of creatine kinase. Tendon ruptures are also described [22,23]. Mansi et al. reported for the first time that statin use was associated with increased risk of dislocation/strain/sprain and maybe osteoarthritis [22]. An association between treatment with statins and muscle rupture has not been described in literature. 18 Databases Table 2. Total reports of muscle rupture associated with statins in the databases of Lareb [24], WHO [25] and EMA [26]. Drug Number of reports Combined ROR (95% CI) statins Lareb: 11 WHO: 107 Eudravigilance: 118 24.1 (10.6 – 54.6) 11.7 (9.5 – 14.4) 13.1 (10.7 – 16.0) Prescription data Table 3. Total number of patients using statins in the Netherlands between 2009 and 2013 [27]. statines 2009 1,507,000 2010 1,594,000 2011 1,662,000 2012 1,751,000 2013 1,827,000 Mechanism It is plausible that myotoxicity can predispose muscles to tear. Many hypothesis have been proposed to explain the myotoxicity caused by statin use. Statins can weaken the integrity of skeletal muscles by reducing the cholesterol content in cell membranes. Another theory explains the myotoxicity by a reduction in the availability of the isoprenoid cometabolites farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (G-PP), causing a reduction in the prenylation of small guanosine triphosphate-binding proteins, such as Rac, Rho, and Ras, which is thought to result in apoptosis of muscle cells. Statins catalyzes the synthesis of mevalonate. Mevalonate is an important precursor of cholesterol, but also of ubiquinone (coenzyme Q), dolichol, and isopentenyl adenosine. Deficiencies in these products may affect the membrane of the myocyte adversely, predisposing the cell to myotoxic consequences. In addition, statins induce a sustained increase in cytosolic Ca2+ levels, which could lead to muscle dysfunction and dysregulation. Statin use can also lead to secondary carnitine deficiency that clinically may manifest as myositis and/or myalgia. Some evidence suggests that statins can inhibit lactic acid efflux from myocytes and thereby induce damage to muscle cells. Alterations of protein synthesis and protein degradation have also been implicated in statin-induced myotoxicity [28]. Draeger et al. performed a study to further investigate the mechanism that mediated statin-induced skeletal muscle damage. Skeletal muscle biopsies from statin users who were asymptomatic were examined and compared with skeletal muscle biopsies from non-statin-users, using both electron microscopy and biochemical approaches. The study shows a clear evidence of skeletal muscle damage in statin-users, with a characteristic pattern that includes breakdown of the T-tubular system and subsarcolemmal rupture. These characteristic structural abnormalities were reproduced by extraction of cholesterol from skeletal muscle fibers in vitro. The authors hypothesize that statin-induced cholesterol lowering contributes to myocyte damage, regardless of whether the patient is symptomatic. This could explain why only two casus describe myalgia as an ADR. The sophisticated membrane architecture with its unique lipid/protein segregation of the skeletal muscle accounts for the vulnerability of skeletal muscle sarcolemma [29]. Discussion and conclusion Lareb received 11 reports of muscle rupture associated with the use of statins. The association showed strong significant disproportionality in the Lareb, WHO and Eudravigilance database. Despite that the association has not been 19 described in literature, it is plausible that statin-induced myotoxicity can predispose muscles to tear. Myotoxicity is a well-known ADR of statins [22]. Our data suggests that statin-induced muscle rupture can occur without intense muscle contraction and without the presence of myalgia. The association of muscle rupture with the use of statins is a new signal. Further investigation of the marketing authorization holder and other national centres is needed to evaluate the signal References 1. KNMP. Informatorium Medicamentorum. (version date: 2014, access date: 14-4-2014) http://kennisbank.knmp.nl/index.asp#IMG1030. 2. Taylor F, Huffman MD, Macedo AF, Moore TH, Burke M, Davey SG, Ward K, Ebrahim S. Statins for the primary prevention of cardiovascular disease. Cochrane.Database.Syst.Rev. 2013;1:CD004816 3. Ward S, Lloyd JM, Pandor A, Holmes M, Ara R, Ryan A, Yeo W, Payne N. A systematic review and economic evaluation of statins for the prevention of coronary events. Health Technol.Assess. 2007;11(14):1-iv 4. Elley CR. ACP Journal Club. Review: Statins reduce mortality and major vascular events in patients with no history of CV disease. Ann.Intern.Med. 2013;159(2):JC2 5. SPC Zocor®. (version date: 31-12-2013, access date: 14-4-2014) http://db.cbg-meb.nl/IBteksten/h13194.pdf. 6. SPC Lipitor®. (version date: 2-1-2014, access date: 14-4-2014) http://db.cbg-meb.nl/IBteksten/h21082.pdf. 7. SPC Lescol®. (version date: 1-11-2013, access date: 14-4-2014) http://db.cbg-meb.nl/IBteksten/h18720.pdf. 8. SPC Selektine®. (version date: 30-4-2013, access date: 14-4-2014) http://db.cbg-meb.nl/IBteksten/h20665.pdf. 9. SPC Crestor®. (version date: 23-12-2013, access date: 14-4-2014) http://db.cbg-meb.nl/IBteksten/h26873.pdf. 10. SPC Livazo®. (version date: 19-8-2012, access date: 16-4-2014) http://db.cbg-meb.nl/IBteksten/h103768.pdf. 11. SPC Vezepra®. (version date: 19-8-2012, access date: 16-4-2014) http://db.cbg-meb.nl/IBteksten/h103447.pdf. 12. Shmerling, R. H. Approach to the patient with myalgia. (version date: 22-1-2014, access date: 155-2014) http://www.uptodate.com/contents/approach-to-the-patient-with-myalgia. 13. Garrett WE, Jr. Muscle strain injuries. Am.J.Sports Med. 1996;24(6 Suppl):S2-S8 14. Chan O, Del BA, Best TM, Maffulli N. Acute muscle strain injuries: a proposed new classification system. Knee.Surg.Sports Traumatol.Arthrosc. 2012;20(11):2356-62. 15. US SPC Zocor®. (version date: 2014, access date: 16-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/019766s091lbl.pdf. 16. US SPC Lipitor®. (version date: 2012, access date: 16-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/020702s062s063lbl.pdf. 17. US SPC Lescol®. (version date: 2012, access date: 16-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/020261s048,021192s021lbl.pdf. 18. US SPC Crestor®. (version date: 2013, access date: 16-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/021366s028s029lbl.pdf. 19. US SPC Pravachol®. (version date: 2012, access date: 16-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/019898s063s064lbl.pdf. 20. US SPC Livalo®. (version date: 2013, access date: 16-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/022363s012lbl.pdf. 21. US SPC Altoprev®. (version date: 2012, access date: 16-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2012/021316s028lbl.pdf. 22. Mansi I, Frei CR, Pugh MJ, Makris U, Mortensen EM. Statins and musculoskeletal conditions, arthropathies, and injuries. JAMA Intern.Med. 2013;173(14):1-10. 23. Sathasivam S, Lecky B. Statin induced myopathy. BMJ 2008;337:a2286 24. Lareb database. (version date: 2014, access date: 22-4-2014) http://www.lareb.nl/Bijwerkingen/Zoek-op-geneesmiddel. 25. WHO Database. (version date: 2014, access date: 22-4-2014) https://tools.who-umc.org/webroot/ (access restricted). 20 26. Eudravigilance database. (version date: 2014, access date: 23-4-2014) http://bi.eudra.org (access restricted). 27. GIP database – Drug Information System of the Dutch Health Care insurance Board. (version date: 7-32014, access date: 18-4-2014) http://www.gipdatabank.nl/databank.asp?tabel=01basis&geg=gebr&item=C10A. 28. Taha DA, De Moor CH, Barrett DA, Gershkovich P. Translational insight into statin-induced muscle toxicity: from cell culture to clinical studies. Transl.Res. 2014; 29. Draeger A, Monastyrskaya K, Mohaupt M, Hoppeler H, Savolainen H, Allemann C, Babiychuk EB. Statin therapy induces ultrastructural damage in skeletal muscle in patients without myalgia. J.Pathol. 2006;210(1):94-102. 21 1.4. Tamsulosin and urinary incontinence Introduction Tamsulosin hydrochloride (Omnic®) is an antagonist of α1-adrenoceptors in the prostate. Tamsulosin is indicated for the treatment of the signs and symptoms of benign prostatic hyperplasia (BPH). It is also used off-label for the treatment of nephrolithiasis in women [1]. Tamsulosin has been approved for the Dutch market since April 1995 [2]. The symptoms associated with benign prostatic hyperplasia (BPH) are related to bladder outlet obstruction, which is comprised of two underlying components: static and dynamic. The static component is related to an increase in prostate size caused, in part, by a proliferation of smooth muscle cells in the prostatic stroma. The dynamic component is a function of an increase in smooth muscle tone in the prostate and bladder neck leading to constriction of the bladder outlet. Smooth muscle tone is mediated by the sympathetic nervous stimulation of alpha1 adrenoceptors, which are abundant in the prostate, prostatic capsule, prostatic urethra, and bladder neck. Blockade of these adrenoceptors can cause smooth muscles in the bladder neck and prostate to relax, resulting in an improvement in urine flow rate and a reduction in symptoms of BPH. In clinical practice tamsulosin is also used off-label for the treatment of nephrolithiasis. Tamsulosin exhibits selectivity for α1-receptors in the human prostate. At least three discrete α1-adrenoceptor subtypes have been identified: α1A, α1B, and α1D; their distribution differs between human organs and tissue. Approximately 70% of the α1-adrenoreceptors in the human prostate are of the α1A subtype [3]. Other selective α1-antagonists for the treatment of BPH on the Dutch market are alfuzosin (Xatral®), doxazosin (Cardura®), silodosin (Silodyx®) and terazosin (Hytrin®). Urinary incontinence often has an identifiable cause in younger persons. In older persons a multifactorial syndrome is more likely. Neuro-urinary pathology, agerelated factors, comorbid conditions, medications, and functional and cognitive impairments may play a role in the older population [4]. The current observation describes the association between tamsulosin and urinary incontinence. No reports for incontinence associated with other α1adrenoceptor antagonists were received by Lareb. Therefore, a possible class effect was not investigated. Reports On April 11th 2014, the database of the Netherlands Pharmacovigilance Centre Lareb contained eleven reports of incontinence associated with the use of tamsulosin. The reports are listed in table 1. Table 1. Reports of urinary incontinence associated with the use of tamsulosin Patient, Number, Sex, Age, Source Drug, daily dose Indication for use A 24881 M, 61-70 years Pharmacist B 41722 M, 61-70 years General Concomitant Medication Suspected adverse drug reaction Time to onset, Action with drug outcome tamsulosin 0.4mg 1dd hyperplasia of prostate urinary incontinence, rash not reported unknown not reported tamsulosin 0.4mg 1dd not reported urinary incontinence 3 days discontinued recovered positive 22 Practitioner rechallenge C 44261 M, 61-70 years Consumer tamsulosin 0.4mg 1dd hyperplasia of prostate allopurinol urinary incontinence not reported unknown not recovered D 52650 M, 71 years and older Pharmacist tamsulosin 0.4mg 1dd hydrochlorothiazide with quinapril pyridoxine calcium levothyroxine carbasalate calcium folic acid dihydrotachysterol urinary incontinence, therapeutic response unexpected with drug substitution not reported no change unknown E 72129 F, 51-60 years Pharmacist tamsulosin 0.4mg 1dd kidney stone diclofenac allopurinol zopiclone incontinence 3 days no change recovered positive rechallenge F 107476 M, 51-60 years Pharmacist tamsulosin 0.4mg 1dd benign prostatic hyperplasia incontinence of urine 2 months discontinued not recovered G 129137 M, 51-60 years Consumer dutasteride/tamsulosin 0,5/0,4mg 1dd benign prostatic hyperplasia Incontinence, ejaculation failure 2 months no change not recovered H 145200 F, 51-60 years Specialist doctor tamsulosin 0.4mg 1dd stone urinary bladder hydrochlorothiazide incontinence of urine several days discontinued recovered I 147585 M, 61-70 years Pharmacist tamsulosin 0.4mg 1dd benign neoplasm of prostate losartan incontinence, headache, tremor, urticaria, hot flush, palpitations, rhinorrhea 1 day discontinued recovered J 160773 M, 61-70 years Pharmacist tamsulosin 0.4mg 1dd lower abdominal pain leuprorelin urinary incontinence 5 days discontinued recovered K 168760 M, 71 years and older Pharmacist tamsulosin 0.4mg 1dd benign prostatic hyperplasia colecalciferol urea mometasone urinary incontinence, fecal incontinence, therapeutic response unexpected with drug substitution 4.5 years discontinued recovered Additional information about the cases is described below: In case A, the complaints were reported as urge incontinence. In case B, both a positive de- and rechallenge were reported. Additionally, the patient ceased his excessive consumption of alcohol (gamma-GT value of 280 U/l) after starting tamsulosin. 23 In case C, the patient experienced the complaints related to stress incontinence (blowing his nose). In case D, the complaints arose when the patient was switched from capsules to tablets. In case E, the patient experienced similar problems in the past. In case F, the patient had not recovered at the time of reporting, which was 6 days after withdrawal of tamsulosin. In case G, the patient switched to separate preparations of dutasteride and alfuzosin and experienced the same complaints. The patient previously used tamsulosin without dutasteride and experienced the ejaculation disorder. Incontinence was not reported for this episode of tamsulosin use. In case I, the patient was treated with cetirizine for his urticaria, and recovered from all complaints two days after withdrawal of tamsulosin. In case J, the patient had a history of prostate cancer for which he underwent surgery. He previously used tamsulosin without experiencing any complaints. In case K, the patients switched from the Ranbaxy brand (which he had been using for approximately 2 years) to the Mylan brand and his complaints disappeared quickly. Other sources of information SmPC Urinary incontinence is not mentioned in the SmPC of tamsulosin [2]. Literature Urinary incontinence has been described as an adverse effect in women using tamsulosin. In a prospective study with 106 patients with voiding dysfunction, three patients developed de novo stress incontinence and one patient experienced an aggravation of an existing stress incontinence [5]. Furthermore, a study with α1-adrenoceptors other than tamsulosin (prazosin, terazosin and doxazosin) showed a statistically significant higher percentage of urinary incontinence in patients using α1-adrenoceptors compared to matched controls [6]. Studies investigating this association in male patients could not be found. Databases Table 2. Reports of (urinary) incontinence with tamsulosin (with or without dutasteride) in the databases of the Netherlands Pharmacovigilance Centre Lareb and the WHO [7] and Eudravigilance (EMA) database [8]. Database Preferred Terms Number of reports ROR (95% CI) Lareb Urinary incontinence 8 7.2 (3.5 – 14.6) Incontinence 3 18.3 (5.6 – 59.3) Total 11 8.7 (4.7 – 15.9) Urinary incontinence 103 5.1 (4.2 – 6.1) Incontinence 30 9.3 (6.5 – 13.3) Total 133 5.7 (4.8 – 6.7) Urinary incontinence 31 3.5 (2.5 – 5.0) Incontinence 12 4.5 (2.6 – 8.0) Total 43 3.8 (2.8 – 5.1) WHO Eudravigilance 24 Prescription data Table 3. Number of patients using tamsulosin in the Netherlands between 2009 and 2013 [9]. Drug 2009 2010 2011 2012 2013 Tamsulosin 172,950 186,450 194,660 199,250 205,890 Tamsulosine / dutasteride - 5,723 19,438 28,787 34,019 Mechanism Tamsulosin binds selectively and competitively to the postsynaptic α1adrenoreceptors, in particular to the subtype α1A and α1D [2]. It is known that α1adrenergic receptors are present in both the detrusor muscle [10] and the bladder sphincter [11]. The antagonistic effect of tamsulosin on the α1-adrenergic receptors of the bladder could therefore theoretically result in both urinary retention and incontinence, depending on the exact affinity of the drug for each receptor subtype and their ratios in sphincter and detrusor. A functional urodynamic study however, showed that tamsulosin had a significant relaxing effect on the resting urethral tone, suggesting a pharmacological treatment for urinary retention [12]. Urinary incontinence could therefore be seen as a possible detrimental effect of this pharmacological mechanism. Discussion and conclusion Lareb received eleven cases of urinary incontinence associated with the use of tamsulosin. The reports concern nine males and two females. In five cases a positive dechallenge was reported and in two cases a positive rechallenge, which support the causality of this association. However, there was also one negative dechallenge. In one case the complaints arose after switching to another brand and in one case after switching to another formulation (tablets to capsules of the same brand). This association was disproportionality present in the Lareb, WHO and Eudravigilance databases, has been described in literature and seems pharmacologically plausible. In addition to the previously described literature, a case report of a woman experiencing stress urinary incontinence after starting doxazosin was found [13]. This case shows similarities with our case C who also experienced stress incontinence, indicating that the complaints are possibly not due to confounding by indication (BPH). Although the articles found in the literature are limited to studies in female patients, our data suggest that male patients could also experience these symptoms. Although BPH could be considered as a possible confounder, these patients often experience overflow incontinence. This is however not mentioned in any of the described cases. Moreover, in two cases (E,H) this can be ruled out since the patients are female, whereas two other cases (A,C) specifically mention urge and stress incontinence respectively. Age however, could be a possible confounding factor in this association. Urinary incontinence should be mentioned in the SmPC of tamsulosin References 25 1. van LJ, van Koningsbruggen PJ, Boukes FS, Goudswaard AN. [Summary of the practice guideline 'Urolithiasis' (first revision) from the Dutch College of General Practitioners]. Ned.Tijdschr.Geneeskd. 2008;152(45):2448-51. 2. Dutch SPC Omnic®. (version date: 2014, access date: 11-4-2014) http://db.cbg-meb.nl/IBteksten/h17931.pdf. 3. US SPC Flomax®. (version date: 2011, access date: 11-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2011/020579s027lbl.pdf. 4. UpToDate. UpToDate. (version date: 2012, access date: 11-4-2014) http://www.uptodate.com/. 5. Lee KS, Han DH, Lee YS, Choo MS, Yoo TK, Park HJ, Yoon H, Jeong H, Lee SJ, Kim H, et al. Efficacy and safety of tamsulosin for the treatment of non-neurogenic voiding dysfunction in females: a 8-week prospective study. J.Korean Med.Sci. 2010;25(1):117-22. 6. Marshall HJ, Beevers DG. Alpha-adrenoceptor blocking drugs and female urinary incontinence: prevalence and reversibility. Br.J.Clin.Pharmacol. 1996;42(4):507-9. 7. WHO Global Individual Case Safety Reports database (Vigibase). (version date: 2014, access date: 23-4-2014) https://tools.who-umc.org/webroot/ (access restricted). 8. Eudravigilance database. (version date: 2014, access date: 23-4-2014) http://bi.eudra.org (access restricted). 9. College for Health Insurances. GIP database. (version date: 9-6-2009, access date: 16-3-2011) http://www.gipdatabank.nl/index.asp?scherm=tabellenFrameSet&infoType=g&tabel=01basis&item=J01FF. 10. Malloy BJ, Price DT, Price RR, Bienstock AM, Dole MK, Funk BL, Rudner XL, Richardson CD, Donatucci CF, Schwinn DA. Alpha1-adrenergic receptor subtypes in human detrusor. J.Urol. 1998;160(3 Pt 1):937-43. 11. KNMP kennisbank. (version date: 2014, access date: 11-4-2014) http://www.kennisbank.knmp.nl/index.asp#home. 12. Reitz A, Haferkamp A, Kyburz T, Knapp PA, Wefer B, Schurch B. The effect of tamsulosin on the resting tone and the contractile behaviour of the female urethra: a functional urodynamic study in healthy women. Eur.Urol. 2004;46(2):235-40. 13. Menefee SA, Chesson R, Wall LL. Stress urinary incontinence due to prescription medications: alpha-blockers and angiotensin converting enzyme inhibitors. Obstet.Gynecol. 1998;91(5 Pt 2):853-4. The Marketing Authorization Holder of tamsulosin (Astellas) has informed Lareb on 07-10-2014 of the following: After careful evaluation of the signal report tamsulosin and urinary incontinence, the Marketing Authorization Holder, is of the opinion that there is no sufficient evidence to support a causal relationship between tamsulosin and urinary incontinence. An addition of a warning for urinary incontinence to the SmPC is therefore not warranted, according to Astellas. 26 1.5. Prednisolone and hiccups Introduction Prednisolone is a corticosteroid drug with predominant glucocorticoid and low mineralocorticoid activity which is used to treat a variety of inflammatory and autoimmune condition [1]. Prednisolone has been marketed internationally since 1955. Prednisolone is the active metabolite of the drug prednisone and is preferred especially in patients with hepatic failure, as these individuals are unable to metabolise prednisone into prednisolone [2]. Prednisolone irreversibly binds with glucocorticoid receptors (GR) alpha and beta for which they have a high affinity. AlphaGR and BetaGR are found in virtually all tissues with variable numbers between 3000 and 10000 per cell, depending on the tissue involved. Prednisolone can activate and influence biochemical behaviour of most cells. The steroid/receptor complexes dimerise and interact with cellular DNA in the nucleus, binding to steroid-response elements and modifying gene transcription. They induce synthesis of some proteins, and inhibit synthesis of others [3]. A hiccup is an involuntary, intermittent, spasmodic contraction of the diaphragm and intercostal muscles. There are numerous causes of hiccups. Hiccups are usually caused by gastric distention from overeating or carbonated beverages. Other causes include gastrointestinal disorders, thoracic disorders or cardiac disorders. Hiccups may also be drug-related. In rare cases, hiccups can be a manifestation of severe underlying disease (eg, malignancy, multiple sclerosis) [4]. Reports The Netherlands Pharmacovigilance Centre Lareb received 3 reports of hiccups associated with the use of prednisolone, in a period from July 11th 2013 to November 1st 2013. Case A (157126) This non-serious spontaneous report from a pharmacist concerns a male aged 71 years and older, with hiccups following administration of prednisolone tablets 20 mg twice daily for asthma with a latency of 1 day after start. The hiccups lasted for 4 days. Prednisolone was withdrawn. The patient recovered. Concomitant medications were levothyroxine sodium, salbutamol/ipratropium, salmeterol/fluticasone, alfacalcidol, fenoterol/ipratropium, tiotropium bromide, formoterol/beclometasone, indacaterol, montelukast, pravastatin, candesartan, diltiazem, carbasalate calcium, pantoprazole, lercanidipine. Start- and stopdates of the concomitant medications were not reported, follow-up questions did not lead to more information. Case B (160866) This report from a pharmacist concerns a male aged 61-70 years years, with hiccups following administration of prednisolone capsules 30 mg once daily for exacerbation of a lung disease and pregabaline 150 mg twice daily for neuropathic pain with a latency of 2 days after start of prednisolone and 1 year after start of pregabaline. The dose for prednisolone and pregabaline was not changed. The patient recovered after 5 days. Concomitant medications were formoterol/budesonide, alfuzosin, amlodipine, metformin, omeprazole and allopurinol. Follow-up questions about the duration of treatment with prednisolone did not lead to more information. 27 Case C (162118) This non-serious spontaneous report from a specialist doctor concerns a male of unknown age, with hiccups following oral administration of prednisolone (dose unknown) for Bechterew's disease with a latency of 1 day after start. The drug prednisolone was withdrawn. The patient recovered almost immediately. After restart of intramuscular prednisolone some time later, the hiccups appeared again within 24 hours. After withdrawal the patient recovered. Concomitant medication was not reported. Other sources of information SmPC Hiccups are not mentioned in the SmPC of prednisolone [5], however the SmPC’s of dexamethasone [6], betamethasone [7] and methylprednisolone [8] do mention hiccups. Literature Although not widely reported, several articles associate hiccups with corticosteroid treatment, however not specifically with prednisolone. Peacock [9] describes a patient who was given preoperative dexamethasone and developed hiccups before anaesthesia and surgery commenced. He at no time was in distress, and the surgical procedure was completed without complication. By the second postsurgical day his hiccups were resolved completely. Other authors report annoying hiccups following intra-articular corticosteroid injection of betamethasone acetate/betamethasone sodium phosphate at the knee joint [10]. A case series presents five patients who developed hiccups after receiving dexamethasone for chemotherapy-induced nausea and vomiting. However, switching dexamethasone to an equipotent dosage of either methylprednisolone or prednisolone resolved the hiccups [11]. This was also seen in a study of 40 cancer patients. Dexamethasone-induced hiccups in these patients could be controlled by replacing dexamethasone with methylprednisolone. After readministration of dexamethasone 74% had recurrence of hiccups [12]. Databases On April 4th 2014, the database of the Netherlands Pharmacovigilance Centre Lareb contained 3 reports of hiccups associated with the use of prednisolone. Hiccups might possibly be a class-effect of corticosteroids, therefore other corticosteroids were taken into account as well. Table 1. Reports of hiccups associated with the use of glucocorticoids in the Lareb, WHO and Eudravigilance database [13,14]. Database Drug Number of reports ROR (95% CI) Lareb Prednisolone 3 5.9 (1.9-18.7) Betamethasone 1 N.A. Dexamethasone 10 42.9 (22.2-82.8) Methylprednisolone 2 N.A. Prednisolone 27 3.0 (2.0-4.3) Betamethasone 50 26.1 (19.7-34.6) Cortisone 2 N.A. Dexamethasone 268 30.5 (27.0-34.6) Hydrocortisone 7 2.8 (1.4-6.0) Methylprednisolone 48 6.2 (4.6-8.2) WHO 28 Database Drug Number of reports ROR (95% CI) Prednisone 17 1.4 (0.9-2.3) Triamcinolone 12 3.3 (1.9-5.8) 7 1.3 (0.6 – 2.6) Betamethasone 10 12.1 (6.5 – 22.6) Cortisone 2 N.A. Dexamethasone 38 11.2 (8.1 – 15.4) Hydrocortisone 1 N.A. Methylprednisolone 11 3.5 (1.9 – 6.4) Prednisone 4 0.8 (0.3 – 2.2) Triamcinolone 2 N.A. Eudravigilance Prednisolone Prescription data Table 3. Number of patients using glucocorticoids (with ATC code H) in the Netherlands between 2009 and 2013 [15]. Drug 2009 2010 2011 2012 2013 412,080 427,550 452,730 472,340 488,330 31,541 34,069 18,280 20,007 23,803 2,269 2,181 1,787 1,475 1,385 Dexamethasone 34,918 35,015 46,520 42,169 44,692 Hydrocortisone 6,502 7,235 8,024 7,984 8,611 Methylprednisolone 11,615 11,831 11,913 11,589 11,832 Prednisone 39,707 34,979 33,030 29,778 25,635 170,910 173,930 179,380 182,940 190,310 Prednisolone Betamethasone Cortisone Triamcinolone Mechanism The mechanism by which corticosteroids induce hiccups has not been fully elucidated yet. Hiccups have been classified as a neurologic reaction triggered by a multitude of factors. However, it has been proposed that corticosteroids lower the threshold for synaptic transmission in the midbrain and directly stimulate the hiccup reflex arc [16]. Discussion and conclusion The Netherlands Pharmacovigilance Centre Lareb received 3 reports of hiccups associated with the use of prednisolone. Two positive dechallenges and one positive rechallenge were reported. The association of prednisolone and hiccups is supported by a statistically significant disproportionality in the database of Lareb and the WHO. This is also the case for other glucocorticoids. The literature mentions hiccups predominantly with the use of dexamethasone and corticosteroid rotation with (methyl)prednisolone is thought to reduce the hiccups. The mechanism behind this is not clear. Other glucocorticoids such as betamethasone are also described to induce hiccups. Therefore, the more potent corticosteroids are reported in the literature to induce hiccups. Possibly the relatively high doses of prednisolone administered in the patients described in the cases reported to Lareb contribute to 29 the occurrence of the hiccups. Several SmPCs of glucocorticoids already mention hiccups as an adverse event, however the SmPC of prednisolone does not. Hiccups should be mentioned in the SmPC of prednisolone References 1. Dutch SmPC Di-Adreson-F®. (version date: 2011, access date: 4-4-2014) http://db.cbg-meb.nl/IBteksten/h00093.pdf. 2. KNMP/Winap. Informatorium Medicamentorum. (version date: 1-10-2012, access date: 4-4-0014) http://kennisbank.knmp.nl/index.asp#IMS324. 3. Rang HP, Dale MM, Ritter JM, et al. Hunter I, editors.5 ed. 2003;The pituitary and the adrenal cortex. p. 413-5. 4. Lembo, A. J. Overview of hiccups. (version date: 7-11-2013, access date: Up to Date®. 5. Dutch SmPC prednisolon. (version date: 2012, access date: 4-4-2014) http://db.cbg-meb.nl/IBteksten/h50969.pdf. 6. Dutch SmPC Oradexon®. (version date: 2014, access date: 4-4-2014) http://db.cbg-meb.nl/IBteksten/h00113.pdf. 7. Dutch SmPC Celestone®. (version date: 2010, access date: 4-4-2014) http://db.cbg-meb.nl/IBteksten/h01834.pdf. 8. Dutch SmPC Depo-Medrol®. (version date: 2013, access date: 4-4-2014) http://db.cbg-meb.nl/IBteksten/h00605.pdf. 9. Peacock ME. Transient hiccups associated with oral dexamethasone. Case.Rep.Dent. 2013;2013:426178 10. Habib G, Artul S, Hakim G. Annoying Hiccups following Intra-Articular Corticosteroid Injection of Betamethasone Acetate/Betamethasone Sodium Phosphate at the Knee Joint. Case.Rep.Rheumatol. 2013;2013:829620 11. Kang JH, Hui D, Kim MJ, Kim HG, Kang MH, Lee GW, Bruera E. Corticosteroid rotation to alleviate dexamethasone-induced hiccup: a case series at a single institution. J.Pain Symptom.Manage. 2012;43(3):625-30. 12. Lee GW, Oh SY, Kang MH, Kang JH, Park SH, Hwang IG, Yi SY, Choi YJ, Ji JH, Lee HY, et al. Treatment of dexamethasone-induced hiccup in chemotherapy patients by methylprednisolone rotation. Oncologist. 2013;18(11):1229-34. 13. WHO database Vigimine. (version date: 2014, access date: 4-4-2014) https://tools.whoumc.org/webroot/ (access restricted). 14. Eudravigilance database. (version date: 2014, access date: 23-4-2014) http://bi.eudra.org (access restricted). 15. GIPdatabase - Drug Information System of the Dutch Health Care Insurance Board. (version date: 2013, access date: 4-4-2014) http://www.gipdatabank.nl. 16. Dickerman RD, Jaikumar S. The hiccup reflex arc and persistent hiccups with high-dose anabolic steroids: is the brainstem the steroid-responsive locus? Clin.Neuropharmacol. 2001;24(1):62-4. 30 1.6. Atovaquone/ proguanil hydrochloride and psychotic disorder Introduction Atovaquone/ proguanil hydrochloride (generic, Malarone® and Malarone® junior) is indicated for the prophylaxis of Plasmodium falciparum malaria in travelers (Malarone® junior: 11-40 kg) and for the treatment of acute and uncomplicated Plasmodium falciparum malaria (Malarone® junior: 5-11 kg) [1-7]. It is recommended for the prophylaxis and the treatment of Plasmodium falciparum in the area where the pathogen is resistant to other antimalarials [1-7]. Atovaquone/ proguanil interferes with two different pathways, both are involved in the biosynthesis of pyrimidines required for the nucleic acid replication of the malaria parasite. Atovaquone inhibits the mitochondrial electron transport and disrupts the mitochondrial membrane potential. The active metabolite of proguanil, cycloguanil, inhibits dihydrofolate reductase and enhances the effect of atovaquone on the mitochondrial membrane potential [1,2]. Atovaquone/ proguanil hydrochloride was granted marketing authorization in the Netherlands on 25 July 2000 (Malarone®) [1] and on 3 March 2003 (Malarone junior®) [2]. The term psychotic disorder has historically received a number of different definitions, none of which has achieved universal acceptance. The narrowest definition of psychotic disorder is restricted to symptoms like delusions or prominent hallucinations. A broader definition concerns a mental disorder that resulted in “impairment that grossly interferes with the capacity to meet ordinary demands of life”. The term has been defined as a loss of ego boundaries or a gross impairment in reality testing [8]. Reports The Netherlands Pharmacovigilance Centre Lareb received four reports of psychotic disorder associated with atovaquone/ proguanil, in a period from June 1st 1995 to April 22th 2014, see Table 1. Additional information regarding the cases is described below. Table 1. Reports of psychotic disorder associated with the use of atovaquone/ proguanil. Patient, Sex, Age, source Drug Indication for use Concomitant medication Suspected adverse drug reaction Time to onset, Action with drug outcome A 153871 M, 21-70 years Psychiatrist Atovaquone/ proguanil tablet Malaria prophylaxis Short psychotic disorder 2 days discontinued recovered in 3 months B 36146 M, 41-50 years Community health professional Atovaquone/ proguanil tablet Malaria prophylaxis C 56444 F 41-50 years Community health professional Atovaquone/ proguanil tablet Malaria prophylaxis Auditory hallucinations that ultimately resulted in a psychosis 12 days Discontinued Unknown D 54661 F, unknown Physician Atovaquone/ proguanil tablet Drug use for unknown indication Psychosis Unknown Unknown Recovered Chlorthalidone 25 Strong psychosis with mg and cetirizine severe suicidal 10 mg. characteristics, irritability, ulcer hands, and paraesthesia of the mouth 1 day discontinued recovered in 2 days 31 In all four cases, it is unknown when the consumers started the use of atovaquone/ proguanil at home or abroad. Casus A: The patient was hospitalized in the psychiatric ward and treated with olanzapine. Another influencing factor mentioned was sleep deprivation. The destination for holiday and medical history were unknown. Casus B: The community health professional reported about himself. The patient was not able to continue atovaquone/ proguanil therapy and evacuation from Tanzania was necessary. Cetirizine was started 1 month before the start of atovaquone/ proguanil and withdrawn on the same date as atovaquone/ proguanil. There was no psychiatric history and he had never had atovaquone/ proguanil therapy before. Casus C: The destination was unknown. The patient was admitted at a psychiatric hospital and diagnosed with Schizophrenia. There is no information on further treatment. Other influencing factor was a stressful period before administration of atovaquone/ proguanil. Casus D: Due to psychosis the patient had returned back home from Africa. Other sources of information SmPC All the SmPCs of atovaquone/ proguanil mention abnormal dreams, depression, anxiety, hallucinations, panic attack and nightmares as possible adverse drug reactions (ADRs) [1-4,6,7,9]. The SmPCs of brand Malarone® and Malarone® junior also mention psychiatric disorders [1,2]. Psychotic disorder is not explicitly mentioned in the SmPCs of atovaquone/ proguanil [1-4,6,7,9]. The US SmPC of the FDA mentions that psychotic events (such as hallucinations) have been identified during postmarketing use of atovaquone/ proguanil. However, a causal relationship has not been established [10]. Literature A search on Pubmed revealed no specific information of atovaquone/ proguanil induced psychotic disorder or hallucinations. One systematic review and metaanalysis, written by Halima Nakato et al. [11], described the occurrence of neuropsychiatric ADRs of atovaquone/ proguanil compared to chloroquine/ proguanil or mefloquine. Ten randomized controlled trials were included. Of these trials, three meta-analysis were included in which patients could report ADRs. No significant difference was found (RR = 0.74; 95% CI = 0.5 - 1.1; I2 = 86.7%) in the reporting of neuropsychiatric ADRs. One of the included trials [12] described the neuropsychiatric ADRs of atovaquone/ proguanil compared to mefloquine in a randomized double blind study. The atovaquone/ proguanil group (n=493) experienced lower neuropsychiatric ADRs than the mefloquine group (n=483) (14% versus 29%, P = 0.001). Among the neuropsychiatric ADRs reported were dreaming, insomnia, anxiety and depression. No hallucination or psychotic disorder were reported. The study was funded by Glaxo Smith Kline. Databases Table 2. Number of reported cases of psychotic disorder associated with the use of atovaquone/ proguanil in the database of Lareb, the WHO and Eudravigilance on April 23rd [13-15]. Drug Number of reports ROR (95% CI) Atovaquone/ proguanil Lareb: 4 4.4 (95% CI 1.6 – 12.0) WHO: 27 6.0 (95% CI 4.1 – 8.8) 32 Drug Number of reports ROR (95% CI) Eudravigilance: 29 9.0 (95% CI; 6.2 – 12.9) Prescription data Prescription data are retrieved from the GIP database [16], were most of the reimbursed prescriptions are stored. Because atovaquone/ proguanil is not covered by most of the basic insurance, the numbers of patients in the GIP database are not a real reflection of the users of atovaquone/ proguanil. Therefore, prescription data are unknown. Mechanism No possible mechanism explaining this association could be found in the literature. Atovaquone is a highly lipophilic compound. Intake with fat can increase the Area Under the Curve (AUC) by a factor 2 to 3 and the maximum concentration (Cmax) by a factor 5 compared to fasting. The bioavailability of atovaquone shows considerable interindividual variability of 45%. It is advised to administrate atovaquone/ proguanil with food or a dairy drink to increase the degree of absorption [1]. Proguanil is not dependent on the food intake [1]. Theoretically, the possible high fat content of a travelers diet in addition to the variable interindividual availability makes specific travelers group at risk to a higher concentration of atovaquone. It is plausible that a higher blood concentration increases the risk of ADRs. However, in susceptible air travel passengers, a jet lag may be sufficient to exacerbate affective illness and result in psychiatric morbidity [17]. No information could be found in literature about the passage of the drug through the blood-brain barrier. Discussion and conclusion The Netherlands Pharmacovigilance Centre Lareb received four reports of psychotic disorder associated with the use of atovaquone/ proguanil. All reports were assessed as serious. In case A and C, the consumer was hospitalized in the psychiatric ward. Patient C was diagnosed with Schizophrenia at the age of 44. The onset of Schizophrenia typically occurs between the late teens and the mid30s, but a late-onset (e.g., after age 45 years) may also occur. Woman are more likely to have a later-onset [8]. In case B and D, evacuation to home country was necessary. The latency period ranged from 1 day to 12 days. Patient B had no psychiatric history before and had a positive dechallenge. The patient recovered 2 days after atovaquone/ proguanil and cetirizine were withdrawn. The SmPC of cetirizine describes also hallucinations and confusion with an incidence of 0.001-0.01% [18]. However, because cetirizine started a month earlier than atovaquone/ proguanil and the psychotic disorder occurred 1 day after the start of atovaquone/ proguanil, it is more likely that the patient recovered due to the withdrawal of atovaquone/ proguanil. In reports A and C, sleep deprivation and stress were mentioned as a possible influencing factor. Most travellers experience sleep deprivation, stress, jet lag, circadian rhythm disruptions, dietary changes, alcohol consumption, or illicit drug use while traveling, which can contribute to a psychosic disorder [19]. In cases A and B, the psychotic symptoms started within 2 days. In case C, the psychotic symptoms started after 12 days and in case D the latency time is unknown. Because malaria incubation period is about 8 to 25 days [20], and none of the reported cases report fever or other malaria related symptoms, it is unlikely that malaria infection induced the psychotic symptoms. 33 The lifetime prevalence for any psychotic disorder is 3% [21] Psychotic disorder with the use of atovaquone/ proguanil is not widely described in literature. Halima Nakato et al [11] found neuropsychiatric ADRs with the use of atovaquone/ proguanil, however this difference was not significant compared to mefloquine. The US SmPC of the FDA mentions that psychotic events have been identified during postmarketing use of atovaquone/ proguanil [10]. Lareb received four reports of psychotic disorder with the use of atovaquone/ proguanil. Although the definition for psychotic disorder is broad and there is a high background incidence, literature is available which describes the occurrence of neuropsychiatric disorder with the use of atovaquone/ proguanil. This association is further strengthened by the disproportional RORs in the Lareb database, Eudravigilance database and WHO database. Because atovaquone/ proguanil is an alternative drug therapy especially when mefloquine is contraindicated in patients with a recent history psychiatric disorders [1], it is important to further investigate if atovaquone/ proguanil can also cause psychotic disorder. Further investigation of the information of the marketing authorization holders and other national centers is needed to evaluate the signal References 1. Dutch SmPC Malarone® 250 mg /100 mg Filmomhulde Tabletten. (version date: 1-10-2013, access date: 18-4-2014) http://db.cbg-meb.nl/IB-teksten/h25386.pdf. 2. Dutch SmPC Malarone Junior® 62,5 mg /25 mg Filmomhulde Tabletten . (version date: 18-102013, access date: 18-4-2014) http://db.cbg-meb.nl/IB-teksten/h28319.pdf. 3. Dutch SmPC Atovaquon/Proguanilhydrochloride Glenmark 250 mg /100 mg Filmomhulde Tabletten. (version date: 7-3-2011, access date: 18-4-2014) http://db.cbg-meb.nl/IBteksten/h105808.pdf. 4. Dutch SmPC Atovaquon/Proguanil HCL Mylan 250 mg /100 mg Filmomhulde Tabletten. (version date: 30-8-2014, access date: 18-4-2014) http://db.cbg-meb.nl/IB-teksten/h109086.pdf. 5. Dutch SmPC Atovaquon/Proguanil HCL Teva 250 mg/100 mg, filmomhulde tabletten. (version date: 23-9-2013, access date: 18-4-2014) http://db.cbg-meb.nl/IB-teksten/h111742.pdf. 6. Dutch SmPC Atovaquon/Proguanil HCL Ratiopharm 250 mg/100 mg, filmomhulde tabletten. (version date: 23-9-2013, access date: 18-4-2014) http://db.cbg-meb.nl/IB-teksten/h111747.pdf. 7. Dutch SmPC Atovaquon/Proguanil HCL Sandoz 250 mg/100 mg, filmomhulde tabletten. (version date: 28-6-2013, access date: 18-4-2014) http://db.cbg-meb.nl/IB-teksten/h111785.pdf. 8. American Psychiatric Association. DSM-IV. 4 ed. 1994. 273p. 9. Dutch SmPC Atovaquon/Proguanil HCL Teva 250 mg/100 mg, filmomhulde tabletten. (version date: 23-9-2013, access date: 18-4-2014) http://db.cbg-meb.nl/IB-teksten/h111742.pdf. 10. FDA SmPC Malarone®. (version date: 7-2-2013, access date: 18-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/021078s022lbl.pdf. 11. Nakato H, Vivancos R, Hunter PR. A systematic review and meta-analysis of the effectiveness and safety of atovaquone proguanil (Malarone) for chemoprophylaxis against malaria. J.Antimicrob.Chemother. 2007;60(5):929-36. 12. Overbosch D, Schilthuis H, Bienzle U, Behrens RH, Kain KC, Clarke PD, Toovey S, Knobloch J, Nothdurft HD, Shaw D, et al. Atovaquone-proguanil versus mefloquine for malaria prophylaxis in nonimmune travelers: results from a randomized, double-blind study. Clin.Infect.Dis. 2001;33(7):1015-21. 13. WHO database . (version date: 2014, access date: 23-4-2014) https://tools.who-umc.org/webroot/. 14. Eudravigilance database . (version date: 2014, access date: 23-4-2014) http://bi.eudra.org. 15. Netherlands Pharmacovigilance Centrum Lareb Database. (version date: 2014, access date: 23-42014) http://www.lareb.nl/Bijwerkingen/Zoek-op-geneesmiddel. 16. College for Health Insurances. GIP-database. (version date: 7-3-2014, access date: 18-4-2014) http://www.gipdatabank.nl/. 17. Srinivasan V, Singh J, Pandi-Perumal SR, Brown GM, Spence DW, Cardinali DP. Jet lag, circadian rhythm sleep disturbances, and depression: the role of melatonin and its analogs. Adv.Ther. 2010;27(11):796-813. 34 18. Dutch SmPC Zyrtec® 10 mg filmomhulde tabletten . (version date: 3-10-2012, access date: 18-42014) http://db.cbg-meb.nl/IB-teksten/h13010.pdf. 19. Meier CR, Wilcock K, Jick SS. The risk of severe depression, psychosis or panic attacks with prophylactic antimalarials. Drug Saf 2004;27(3):203-13. 20. Mandell. Gerald L, editor. Principles and practice of infectious diseases. 7 ed. 2010. 3447p. 21. Perala J, Suvisaari J, Saarni SI, Kuoppasalmi K, Isometsa E, Pirkola S, Partonen T, TuulioHenriksson A, Hintikka J, Kieseppa T, et al. Lifetime prevalence of psychotic and bipolar I disorders in a general population. Arch.Gen.Psychiatry 2007;64(1):19-28. 35 1.7. Ciclosporin and posterior reversible encephalopathy syndrome Introduction Ciclosporin (Neoral®) is indicated after organ transplantation to prevent the rejection of a transplanted solid organ, where ciclosporin can be used as monotherapy or in combination with low doses of corticosteroids or other immunosuppressive medication. Furthermore ciclosporin is indicated after bone marrow transplantation for the prophylaxis of graft rejection, and for the prophylaxis or treatment of "graft-versus-host" (GVH) reaction. In addition ciclosporin is indicated for very severe psoriasis when other therapies had not been effective, very severe therapy-resistant atopical dermatitis in adults, steroidresistant nephrotic syndrome as a result of glomerular pathology, and very severe rheumatoid arthritis in adults when other therapies had not been effective [1]. Ciclosporin is a cyclic polypeptide consisting of 11 amino acids. It is a potent immunosuppressive agent. Preclinical studies showed that ciclosporin inhibits the development of cell responses, including the rejection of allogenic grafts, skin hypersensitivity of the delayed type, " graft versus host " reactions and T celldependent antibody formation. At cellular level, ciclosporin inhibits the production and release of lymphokine, including interleukin-2. It appears that ciclosporin blocks the resting lymphocytes in the G0 or G1 phase of the cell cycle and inhibits the antigen induced release of lymphokines from activated T-cells. The available data suggest that ciclosporin has a specific and reversible effect on lymphocytes. In contrast to cytotoxic drugs, ciclosporin has no clinically relevant effect on hematopoiesis [1]. Ciclosporin was granted marketing authorization in the Netherlands in 1983 [2]. Posterior reversible encephalopathy syndrome (PRES), also referred to as reversible posterior leukoencephalopathy syndrome (RPLS), is a clinical radiographic syndrome of heterogeneous etiologies that are grouped together because of similar findings on neuroimaging studies. The clinical syndrome is characterized by headaches, altered consciousness, visual disturbances and seizures. The syndrome is not always reversible, and it is often not confined to either the white matter or the posterior regions of the brain [3]. The incidence of PRES is unknown and all age groups appear susceptible. PRES has been described in a number of medical conditions, with hypertensive encephalopathy, eclampsia, and the use of cytotoxic and immunosuppressant drugs being the most common. Another risk factor is renal disease [3]. It is important to recognize and treat this condition promptly, in preventing the permanent damage that can occur in this otherwise typically reversible condition [3]. Reports From 1 August 2013 1999 until 20 February 2014 the Netherlands Pharmacovigilance Centre Lareb received two reports of PRES as the reported reaction, associated with ciclosporin. Of encephalopathy as the reported reaction, associated with ciclosporin, Lareb received six reports in the period from 29 April 1999 until 6 December 2012. The two reports with PRES as the coded reaction, are described here: Case A (158347) This serious spontaneous report from a specialist doctor concerns a male aged 71 years and older, with progressive renal failure following administration of 36 ciclosporin for nephrotic syndrome with a latency of 1 month after start. The dose for ciclosporin was reduced, followed by improved renal function. About 1 year and 3 months after start of ciclosporin the patient was hospitalized because of atrial fibrillation which spontaneously converted to sinusrithm after rate control. During this hospitalization the patient experienced progressive hypertension, despite elevation of the antihypertensive medication. Then the patient experienced unwellness and a convulsion wherefore admission to the intensive care unit, eventually designated or as occipital ischaemic CVA, or as PRES (differential diagnosis: caused by moxifloxacin/ciclosporin or hypertension). The drug ciclosporin was withdrawn. The patient recovered. Concomitant medications were prednisolone, moxifloxacin, rifampicin. The medical history indicates renal failure due to focal segmental glomerulosclerosis. Furthermore the medical history indicates total hip replacement. For wound infection the patient used moxifloxacin and rifampicin. The past drug therapy indicates risedronic acid with aggravated hip pain and alendronic acid with back pain. Case B (168399) This serious (hospitalisation) spontaneous report from a pharmacist concerns a female aged 51-60 years, with reversible posterior leukoencephalopathy syndrome (RPLS/PRES) resulting in seizures, following oral administration of ciclosporin, in a dosage of twice a day 275 mg, for immunosuppression in graft versus host disease after donor lymphocyte infusion, with a latency of 12 days after start. The patient was hospitalized for graft versus host disease after donor lymphocyte infusion. The patient was hospitalized for 5 weeks. CT en MRI 11 days after start of ciclosporin indicated RPLS. The CT scan 19 days after start of ciclosporin showed subarachnoidal bleedings and diffuse white matter abnormalities, and the patient had seizures. The drug ciclosporin was withdrawn and replaced by sirolimus. The seizures were treated with levatiracetam. The patient was recovering at the time the report was submitted to Lareb. Concomitant medications were pantoprazole, alendronic acid, posaconazole, levofloxacin, valaciclovir, calcium / colecalciferol, prednisone, levothyroxine sodium. The medical history indicates multiple myeloma stage IIIa, allogeneic stem cell transplantation, donor lymphocyte infusion in the month before start of the reaction. The patient used ciclosporin in the past without a similar reaction. In Case A the diagnosis PRES was considered possible, since the differential diagnosis also comprised ischaemic CVA. In addition, the patient also experienced hypertension, which made have played a role in the possible PRES. The report did not contain information whether a cerebral imaging was performed. In Case B the symptomatology of the patient and the image of PRES on the scans, were described. In this case a change in blood pressure was not described. The latencies of the two cases were very different: about 1 year and 3 months, and 12 days respectively. One patient recovered and the other patient was recovering at the time the report was submitted to Lareb, after withdrawal of ciclosporin. The six reports received by Lareb, with encephalopathy as the reported reaction are listed in Table 1. In three of these cases other possible causes of encephopathy were described: Case 34796 concerned progressive multifocal leukoencephalopathy. Case 64031 concerned hyperammonemia in a mycobacterium genavense infection, where the patient experienced septic shock, ARDS, hepatic failure and encephalopathy among other symptoms. Case 159238 concerned encephalopathy, hyponatremia, hyperammonemia, and fever, and lumbar puncture revealed infection. 37 Table 1. Reports of encephalopathy associated with the use of ciclosporin Patient, Number, Sex, Age, Source Drug, daily dose Indication for use Concomitant Medication Suspected adverse drug reaction Time to onset, Action with drug outcome A 46582 F, 21-30 years Pharmaceutical Company tacrolimus 4mg per day lung transplant, ciclosporin 100mg 2dd lung transplant mycophenolate mofetil, prednisone, levonorgestrel/ethinyle stradiol headache, vision blurred, encephalopathy, somnolence, hemianopia, blood pressure increased, nausea not reported discontinued recovered B 24478 M, 61-70 years Specialist doctor ciclosporin 2dd 175 mg kidney transplant amitriptyline, acetylsalic acid, omeprazole, isradipine, prednisone, furosemide encephalopathy 2 years discontinued recovering C 34796 M, Pharmaceutical Company ciclosporin dose unknown lung transplant cataract, encephalopathy, death nos not reported unknown fatal D 64031 F, 61-70 years ciclosporin dose unknown renal transplant prednisolone, mycophenolate mofetil disseminated intravascular coagulation, renal failure, body temperature increased, lymphopenia, glasgow coma scale abnormal, septic shock, diarrhea, encephalopathy, normocytic anemia, acute respiratory distress syndrome, hyperammonemia, neutrophilia, hemoglobin low, palpitations, tachycardia, mycobacterial infection, consciousness decreased, spleen enlarged, myocardial infarction, hepatic failure, abdominal tenderness, lymphadenopathy 214 days unknown fatal E 159238 F, non specified blood stem cell transplant chronic lymphocytic leukaemia, ciclosporin dose unknown, fludarabine dose unknown chronic lymphocytic mycophenolic acid encephalopathy 38 days no change recovered 38 leukaemia, non specified drug prophylaxis, mycophenolate mofetil dose unknown chronic lymphocytic leukemia F 84712 M, 2-4 years Specialist doctor ciclosporin 2 dd 90 mg graft versus host disease co-trimoxazole encephalopathy 18 months discontinued recovered with sequelae Other sources of information SmPC There is a variation between SmPCs of ciclosporin within The Netherlands concerning the way this adverse drug reaction is mentioned. The Dutch SmPC of generic ciclosporin mentions as an uncommon (between 1/100 and 1/1,000) occurring adverse drug reaction: "Signs of encephalopathy such as convulsions, confusion, disorientation, decreased reactivity, irritability, insomnia, visual disturbances, cortical blindness, coma, paresis, cerebellar ataxia". Furthermore the Dutch SmPC of ciclosporin mentiones that with JCvirus associated progressive multifocal leukoencephalopathy (PML), has been observed in patients treated with cyclosporine. PRES is not specifically mentioned in the The Dutch SmPC of generic ciclosporin [4]. The SmPC of ciclosporin Neoral® mentiones as an uncommon (between 1/100 and 1/1,000) occurring adverse drug reaction: “Encephalopathy including Posterior Reversible Encefalopathiesyndrome (PRES), signs and symptoms such as convulsions, confusion, disorientation, decreased responsiveness, agitation, insomnia, visual disturbances, cortical blindness, coma, paresis, cerebellar ataxia” [1]. The US SmPC of the FDA mentions that encephalopathy, including Posterior Reversible Encephalopathy Syndrome (PRES), has been described both in postmarketing reports and in the literature. It is mentioned that manifestations include impaired consciousness, convulsions, visual disturbances (including blindness), loss of motor function, movement disorders and psychiatric disturbances. Furthermore it is describes that in many cases, changes in the white matter have been detected using imaging techniques and pathologic specimens. The US SmPC also describes that predisposing factors such as hypertension, hypomagnesemia, hypocholesterolemia, high-dose corticosteroids, high ciclosporin blood concentrations, and graft-versushost disease have been noted in many but not all of the reported cases. The SmPC mentions that the changes in most cases have been reversible upon discontinuation of ciclosporin, and in some cases improvement was noted after reduction of dose. Furthermore the SmPC adds that it appears that patients receiving liver transplant are more susceptible to encephalopathy than those receiving kidney transplant [5]. Literature In 1996 PRES was first described as an entity. It was described that PRES has diverse causes, immunosuppressive drugs being one of the common precipitants. In four of the cases the use of cyclosporine was described [6]. Afterwards many more cases have been published reporting PRES associated with the use of cyclosporine. 39 An article by Teive et al described eight patients who received ciclosporin A after allogeneic bone marrow transplantation or as treatment for severe aplastic anemia who developed PRES. In six of these patients neurological dysfunction occurred preceded by or concomitant with high blood pressure and some degree of acute renal failure. When lowering the dose or withdrawal of ciclosporin the symptoms and neuroimaging abnormalities improved [7]. Two patients after pulmonary transplantation for cystic fibrosis, developed arterial hypertension, headache, visual trouble and generalized seizures with diffuse cortical and subcortical lesions predominantly in posterior regions. Disappearance of the symptoms after withdrawal of ciclosporin confirmed the diagnosis of cyclosporine-related PRES [8]. A 27-year old patient with collapsing focal glomerulosclerosis developed abrupt elevation of blood pressure and neurological symptoms 3 weeks after start of ciclosporin. An MRI showed scan lesions suggestive of PRES. Two months after withdrawal of ciclosporin the MRI had normalized [9]. PRES related to ciclosporin has also been described in two cases after heart transplantation. A 68-year old woman developed arterial hypertension, headache, visual disturbances, and generalized seizures at day 14 after start of ciclosporin and mycophenolate mofetil and prednisone, and lesions on MRI were visible. A 19-year old man developed acute headache and generalized seizures on day 44 after start of ciclosporin and prednisone, with lesions on MRI. Ciclosporin concentrations were therapeutic. Both patients recovered. In the first case ciclosporin had to be withdrawn to reverse the symptoms [10]. A 35-year old woman with SLE was described who developed PRES during ciclosporin use. Althought this patient also had transient elevated blood pressure 3 days before development of the neurological symptoms, the authors estimated that ciclosporin had a causative role in the development of PRES, based on elevated serum ciclosporin level and dramatic neurologic recovery after withdrawal of ciclosporin [11]. In a study of 660 renal pediatric patients 11 patients (8 males, 3 females, age 315 years) experienced PRES, in which in one patient high trough level of ciclosporin was measured and ciclosporin toxicity was considered a contributory factor [12]. Databases Table 2. Reports of posterior reversible encephalopathy syndrome associated with ciclosporine in the Lareb, WHO and Eudravigilance database Database MedDRA PT Number of reports ROR (95% CI) Lareb Posterior reversible encephalopathy syndrome 2 - WHO “ 158 68.1 (57.5 - 80.7) Eudravigilance “ 279 32.4 (28.4 – 36.8) Prescription data Table 3. Number of patients using ciclosporin in The Netherlands between 2009 and 2013 [13]. Drug 2009 2010 2011 2012 2013 Ciclosporin 7,629 7,407 7,055 6,826 6,737 Mechanism 40 The pathogenesis of PRES remains unclear, but it appears to be related to disordered cerebral autoregulation and endothelial dysfunction. Different mechanisms might be etiologically important in different clinical situations. Ciclosporin is associated with the neurologic deficits of PRES. After renal toxicity, neurotoxicity is the most serious side effect with ciclosporin, which affects 25 percent to 59 percent of transplant patients. Hypomagnesemia, hypocholesterolemia, the vasoactive agent endothelin and hypertension have been implicated in facilitating ciclosporin neurotoxicity. Ciclosporin may exacerbate hypertension by inhibiting nitric oxide production. The symptoms of ciclosporin neurotoxicity resemble mitochondrial encephalopathy indicating an underlying mechanism of mitochondrial dysfunction [3]. In a previous study [14] in sixteen patients with neurologic injury attributed to cyclosporine therapy, the clinical and radiologic findings in patients showing the neurotoxic effects of ciclosporin appeared to be identical to those with hypertensive encephalopathy. In this study the only major factor associated with the neurotoxic effects of ciclosporin in all patients was systemic hypertension, although microangiopathic hemolytic anemia, thrombocytopenia, and hypoalbuminemia were common. Discussion and conclusion The Netherlands Pharmacovigilance Centre Lareb received a report of possible PRES and one of PRES, associated with the use of ciclosporin. Lareb also received three reports of encephalopathy in which no other causes of encephalopathy were described, so which might have indicated PRES. In the WHO database there are 158 cases present of PRES associated with ciclosporine, with a ROR of 68.1 (95% CI 57.5 - 80.7). In the Eudravigilance database there is the large number of 279 cases of PRES associated with cyclosporine present, with a ROR of 32.4 (95% CI 28.4 - 36.8). The FDA SmPC of ciclosporine also mentiones PRES in ciclosporin and provides an extensive explanation. The FDA SmPC mentions that encephalopathy, including PRES, has been described both in post-marketing reports and in the literature and it describes it’s manifestations. The FDA SmPC also describes that changes in most cases have been reversible upon discontinuation of ciclosporin, and in some cases improvement was noted after reduction of dose [5]. Also in the literature many cases of PRES in ciclosporin have been described. In the Dutch SmPCs there is a variation on mentioning PRES between different ciclosporin products. Because of the possible reversibility of the serious condition of PRES, it is important that PRES is specifically mentioned in the SmPCs of ciclosporin. As reported on the website of the European Medicines Agency (EMA), the EMA completed a review of Sandimmun and Sandimmun Neoral on 27 June 2013, and the Agency’s Committee for Medicinal Products for Human Use (CHMP) concluded that there was a need to harmonise the prescribing information for Sandimmun® and Sandimmun Neoral® in the European Union (EU) [15]. In the Sandimmun Article-30 referral-Annex III PRES is specifically mentioned [16]. Posterior Reversible Encephalopathy Syndrome (PRES), should be mentioned in the SmPC of all ciclosporin products 41 References 1. Dutch SmPC Neoral® 25 mg capsules, 100 mg capsules. (version date: 4-11-2013, access date: 22-4-2014) http://db.cbg-meb.nl/IB-teksten/h17496.pdf. 2. Dutch SmPC Sandimmune® concentraat voor oplossing voor intraveneuze infusie 50 mg/ml. (version date: 4-11-2013, access date: 29-4-2014) http://db.cbg-meb.nl/IB-teksten/h09846.pdf. 3. UpToDate. (version date: 2012, access date: 3-4-2014) http://www.uptodate.com/contents/reversible-posterior-leukoencephalopathysyndrome?source=search_result&search=reversible+posterior+leukoencephalopathy+syndrome& selectedTitle=1%7E150. 4. Dutch SmPC ciclosporine Actavis 25 mg capsules, 50 mg capsules, 100 mg capsules. (version date: 30-5-2013, access date: 22-4-2014) http://db.cbg-meb.nl/IB-teksten/h34434.pdf. 5. FDA. (version date: 5-2-2013, access date: 10-4-2014) http://www.accessdata.fda.gov/drugsatfda_docs/label/2013/050715s033,050716s034lbl.pdf. 6. Hinchey J, Chaves C, Appignani B, Breen J, Pao L, Wang A, Pessin MS, Lamy C, Mas JL, Caplan LR. A reversible posterior leukoencephalopathy syndrome. N Engl J Med. 1996;334(8):494-500. 7. Teive HA, Brandi IV, Camargo CH, Bittencourt MA, Bonfim CM, Friedrich ML, de Medeiros CR, Werneck LC, Pasquini R. Reversible posterior leucoencephalopathy syndrome associated with bone marrow transplantation. Arq Neuropsiquiatr. 2001;59(3-B):784-9. 8. Lepoivre T, Treilhaud M, Auffray-Calvier E, Rigal JC, Blanloeil Y. [Posterior reversible encephalopathy syndrome: about 2 cases related to the cyclosporine]. Ann Fr Anesth Reanim. 2003;22(5):466-9. 9. de Oliveira RA, Fechine LM, Neto FC, Nicodemus JM, Silva GBJr, Silva LS. Posterior reversible encephalopathy syndrome (PRES) induced by cyclosporine use in a patient with collapsing focal glomeruloesclerosis. Int Urol Nephrol. 2008;40(4):1095-8. 10. Dzudie A, Boissonnat P, Roussoulieres A, Cakmak, Mosbah K, Bejui FT, Obadia JF, Sebbaq L. Cyclosporine-related posterior reversible encephalopathy syndrome after heart transplantation: should we withdraw or reduce cyclosporine?: case reports. Transplant Proc. 2009;41(2):716-20. 11. Lai TK, Wong TC, Wong WC, Chin AC, Chan RY, Huang HY. A reversible cause of blindness that should not be forgotten: cyclosporine-induced posterior reversible encephalopathy syndrome. Hong Kong Med J. 2009;15(2):153-4. 12. Gera DN, Patil SB, Iyer A, Kute VB, Gandhi S, Kumar D, Trivedi HL. Posterior reversible encephalopathy syndrome in children with kidney disease. Indian J Nephrol. 2014;24(1):28-34. 13. College for Health Insurances. GIP database. (version date: 7-3-2014, access date: 10-4-2014) http://www.gipdatabank.nl/. 14. Schwartz RB, Bravo SM, Klufas RA, Hsu L, Barnes PD, Robson CD, Antin JH. Cyclosporine neurotoxicity and its relationship to hypertensive encephalopathy: CT and MR findings in 16 cases. AJR Am J Roentgenol. 1995;165(3):627 15. European Medicines Agency. Referrals. Sandimmun and associated names. (version date: 20-122013, access date: 10-7-2014) http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/referrals/Sandimmun_and _associated_names/human_referral_000346.jsp&mid=WC0b01ac05805c516f. 16. Annex III . Summary of product characteristics, labelling and package leaflet. Sandimmun and associated names. (version date: 20-12-2013, access date: 10-7-2014) http://www.ema.europa.eu/docs/en_GB/document_library/Referrals_document/Sandimmun_30/W C500144898.pdf. 42 2. Publications (1) Kant A, Rümke HC. Bijwerkingen na vaccinatie: vergeet ze niet te melden! Tijdschrift voor infectieziekten 2014;9(3):71-6. (2) Steenvoorden G, Oers A van. Signaleren en melden van bijwerkingenwanneer en waarom melden verpleegkundig specialisten? Dé verpleegkundig specialist 2014;9(1):30-1. (3) Winkel B ten. Borstvoeding en geneesmiddelgebruik, focus op antidepressiva. Praktische pediatrie 2014;(2):105-8. (4) Puijenbroek EP van, Grootheest AC van. Organization of pharmacovigilance in The Netherlands. Mann's Pharmacovigilance (eds E. B. Andrews and N. Moore), John Wiley & Sons, Ltd, Oxford, UK. 13e. 2014. (5) Oosterhuis I, Harmark LVD, Puijenbroek EP van. Experiences with the use of Varenicline in daily practice in teh Netherlands: a prospective, observational cohort study. Drug Safety 2014;(37):449-57. (6) Hunsel FPAM van, Kant A, Puijenbroek EP van. Trombose en embolie bij gebruik van Diane-35, analyse van meldingen bij Lareb. Nederlands Tijdschrift voor Geneeskunde 2014 May 1;158(0):A6651. (7) Rolfes L, Wilkes S, Hunsel FPAM van, Puijenbroek EP van, Grootheest AC van. Important information regarding reporting of adverse drug reactions; a qualitative study. Int J Pharm Pract 2014;22(3):231-3. (8) Passier A. Domperidon lijkt effectief ter stimulatie van borstvoeding. Pharm Weekbld 2014;(149):22-3. (9) Weber-Schoendorfer C, Chambers C, Wacker E, et al. Pregnancy outcome after methotrexate treatment for rheumatic disease prior to or during early pregnancy:a prospective multicenter cohort study. Arthritis Rheumatol 2014;66(5):1101-10. (10) Ekhart C, Puijenbroek EP van. Blijvende seksuele functiestoornissen na staken va een SSRI? Tijdschrift voor Psychiatrie 2014;56(5):336-40. 43