Volume 2 (2012) // Issue 1 // e-ISSN 2224-3453

Neurology · Neurosurgery · Medical Oncology · Radiotherapy · Paediatric Neuro- oncology · Neuropathology · Neuroradiology · Neuroimaging · Nursing · Patient Issues

Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours Benit CP, Vecht CJ European Association of NeuroOncology Magazine 2012; 2 (1) 15-24

Homepage: www.kup.at/ journals/eano/index.html

OnlineOnline DatabaseDatabase FeaturingFeaturing Author,Author, KeyKey WordWord andand Full-TextFull-Text SearchSearch

THE EUROPEAN ASSOCIATION OF NEUROONCOLOGY Member of the Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours Christa P Bénit, Charles J Vecht

Abstract: Seizures are a common manifestation sionally develops during radiation. Although fects that are almost impossible to unravel, as in patients with brain tumours, and most patients this side effect is rare, it can be life-threaten- drugs and other therapies used can have aggra- need anticonvulsants. Apart from seizure control, ing. Many anti-epileptic drugs can have extra vating or counteracting effects on each other. the risk of side effects makes the proper choice toxic effects with existing organ dysfunction, Knowledge of individual anticonvulsants and an- of anticonvulsants a major concern. Toxicities like bone-marrow suppression or liver abnor- ticipation of toxicity including the recognition of not only exist as common side effects, but also malities, this applies particularly for PB, PHT, already existing co-morbidities all contribute to appear as drug-drug interactions, neurotoxici- CBZ, and VPA. better selection and dosing of anticonvulsants, ties, and other organ dysfunctions. Existing clinical or subclinical signs of brain including the choice of agents that do not inter- One reason for interactions is the use of the damage secondary to space-occupying tumoural act. Although this survey is not aimed at classical anti-epileptic drugs (AED), phenobarbital effects or the sequelae of previous neurosurgery, the proper drug choice, future studies need to (PB), phenytoin (PHT), and carbamazepine (CBZ). radio-, and chemotherapy enhance the chances show which anti-epileptic agents or combin- Large differences in dose regimens with concomi- of neurotoxicity. Besides, the intake of - ations would be the best match to achieve effec- tant chemotherapy reflect the potency of these ef- vulsants itself and their total number strongly tive seizure control together with good tolerabil- fects. Although valproic acid (VPA) can be benefi- contribute to cognitive dysfunction. As neuro- ity. Eur Assoc Neurooncol Mag 2012; 2 (1): cial to prevent tumour growth, it may lead to bone cognitive decline interferes with quality of life, 15–24. marrow suppression and other toxicities because such changes may substantially affect daily ac- of its enzyme-inhibiting properties. tivities of patients and their family members. Key words: brain tumour, seizure, anticonvul- Another noteworthy side effect are skin reac- The multitude of co-therapies applied with sant, drug interaction, toxicity, cognitive dys- tions, like erythema multiforme, which occa- brain tumours contributes to a myriad of side ef- function

 Introduction Seizures in patients with brain tumours can be classified as partial (simple or complex partial) or symptomatic, with or Epilepsy is common in patients with brain tumours, and sei- without secondary generalisation. Here we briefly mention zures constitute the presenting symptom in 30–50 % of pa- the main characteristics of anticonvulsants, and Table 1 de- tients with brain tumours [1]. Seizures can also affect patients picts an overview of the main mechanisms of action, the meta- with systemic cancer with brain or leptomeningeal metastases bolic pathways involved, their pharmacokinetic properties or by organ dysfunction or drug treatment, including chemo- and common toxicities, including idiosyncratic side effects therapy causing metabolic or toxic encephalopathies [2]. For occurring in cancer patients. these reasons, patients with seizures and cancer often need 2 anticonvulsants; /3 of patients with primary brain tumours use Phenobarbital (PB) is one of the oldest anticonvulsants and AEDs [3]. Unavoidably, this may lead to side effects either as still in use in many parts of the world. Major drawbacks are its general toxic effects, or more specifically related to the under- relatively strong sedating effect as well as its being a strong lying condition, for example the occurrence of interactions enzyme-inducer. Nevertheless, because of its action as a broad- with concomitantly administered chemotherapeutic agents. In spectrum anticonvulsant, it may still be applied in treatment- this review, we will comment on the general side effects of resistant seizures. In cancer patients, one should be aware of anticonvulsants, followed by a more extensive discussion on cognitive side effects, hepatic dysfunction, skin reactions in- side effects of anticonvulsants associated with brain tumours cluding Stevens-Johnson syndrome (SJS), and drug interac- or systemic cancer. tions.

Side effects of anticonvulsants are more common in patients Phenytoin (PHT) is a first-generation anticonvulsant which is with brain tumours (20–40 %) than in other types of epilepsy also employed in status epilepticus. It is a strong enzyme-in- 1 and lead in almost /4 to a discontinuation of therapy [4]. ducer and has been implied in many reports on interactions These effects include cognitive changes, abnormalities re- with co-administered agents, including chemotherapeutic lated to organ function, interactions with other drugs, most drugs (CTD). Besides, it shows non-linear pharmacokinetics notably chemotherapy, and other toxicities particularly asso- and has a relatively small therapeutic window. Today, it is ciated with brain tumours. mainly applied by the intravenous route in status epilepticus, and is felt to be less suitable for oral maintenance therapy. Interaction with other drugs and side effects like encepha- Received on December 30, 2011; accepted on January 8, 2012; Pre-Publishing lopathy, hepatitis, coagulation defects, and bone marrow hy- Online on January 24, 2012 poplasia are of concern in patients with brain tumours. From the Neuro-oncology Unit, Department of Neurology, Medical Center The Hague, The Netherlands Correspondence to: Charles J Vecht, MD, PhD, Neuro-oncology Unit, Depart- Carbamazepine (CBZ) is a still widely used first-generation ment of Neurology, Medical Center The Hague, POB 432, 2501 CK The Hague, anticonvulsant for partial seizures. It is a potent enzyme-in- The Netherlands; e-mail: [email protected] ducer and can strongly accelerate the metabolism of many

EUR ASSOC NEUROONCOL MAG 2012; 2 (1) 15 For personal use only. Not to be reproduced without permission of Krause & Pachernegg GmbH. Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours : + 5–95 < 15 < None 38 CL Protein to GFR to 0.130 1/2 30 0.032 12– 0.044– 55 T 6.3 d 0.75 13– 0.027– 20–25 1.5– 20– 0.09 86 0.6 13 0.8 72 0.02 0.8– 5– 0.133 75 V 0.5– 24– 0.003– 85–9 5 0.8 10 0.7 0.3– 8– 2 26 0.65– 5–7 0.120– None 1.04 valproic acid; ZON: zonisamide. valproic : sodium; NMDA: N-Methyl-D-aspartate; Ca : sodium; NMDA: + (without 1.0– the blood, 0.5– 5–11 0.01 None droxylation, mainly renal 0.6– 19– 0.022– 9–17 mide; LTG: lamotrigine; LEV: levetiracetam; OXC: oxcarbazepine; OXC: levetiracetam; lamotrigine; LEV: mide; LTG: ), renal excretion (30 %)), renal excretion 1.8 70 0.019 ugation (60 %), renal amate; VGB: vigabatrin; VPA: amate; VGB: VPA: vigabatrin; droxylation, glucuronidation droxylation, Hepatic epoxidation, and excretion (l/kg) (h) (l/kg/h) binding (10 %) (10 acetylation (40 %) excretion metabolism 4.4 80 Hepatic oxidation, glucosidation,Hepatic oxidation, 0. 42– 46– 0.006– 45–60 hydroxylation, conjugationhydroxylation, 0.75 136 0.009 unchanged renal excretion (40 %) renal excretion unchanged No metabolism, renal excretion conjugation conjugation (70 %, unchanged) conj tion (20 % phase-1 reaction), renal excretion 1.3 60 0.084 to Metabolism 6–1.3 in hydrolysed Partially 1–4 Hepatic reduction and 4–12 hy Hepatic oxidation, 1 al bio- Time availability peak (%) (h) levels (%) - + -/Ca + - > 95 2–6 Hepatic acetylation, glucuronida- 1.2– 60– 0.015– 40–50 + -channel > 90 2–6 and Hepatic hydroxylation + activity conjugation 15 0.015 -channels + -channels 90 + -channels, 81–95 2–4 No metabolism, -channels, < 65 2–3 without Renal excretion - and Ca hanism of action Or + + + ltage-dependent > 95 1–3 Hepatic glucuronidation ltage-dependent 95 voltage-dependent > 95 4–6 Hy e-dependent Na ks NMDA receptors ks NMDA -channels -channels -channels -channels + + + + protein 2 (SV2A) dependent Na glutamate antagonist, blocks channels channels voltag Main mec bloc GABA receptor agonist GABA fects, receptor agonist GABA 90 : elimination half-life; Cl: clearance; DRESS: drug reactions with eosinophilia and systemic symptoms; Na reactions with eosinophilia and systemic Cl: clearance; DRESS: drug : elimination half-life; 1/2 ects (33 %,(33 ects transaminase inhibitor GABA 80–90 1–2excretionmetabolism,renal No 0.8 6–8 Similar None aplastic voltage-dependent Blocks 75–85 4–8 ness, headache, ness, nausea,headache, of voltage- inactivation Slow 95 > 2–4 Hepatic demethylation, ache, blood dyscrasia, ataxia blood dyscrasia, ache, Na anaemia, hepatotoxicity, Na gum hyperplasia, lupus-likegum hyperplasia, Na toxicity, weight loss, renal weight toxicity, receptor agonist, GABA excretion 1.0 25 0.036 diplopia, blurred vision, cognitive diplopia, blurred hyponatraemia, SJS/TEN hyponatraemia, drowsiness loss weight seizures skin reactions dysfunction, irritabiity psychosis suppression, hyponatraemia reactions, hirsutism calculi drowsiness syndrome cystic ovarian renal cuniculi cognition, ataxia, mood change, neutropenia, aplastic anaemia, glutaminergic tremor, weight gain, hair loss, weight tremor, SJS/TEN aplastic anaemia, insom nia, conductance often irreversible), fatigue, irreversible), often diplopia, SJS, bone marrowdiplopia, SJS, Na Somnolence, dizziness, ataxiaCa to Binds : volume of distribution; T of distribution; : volume d : potassium; GFR: glomerular filtration rate; CBZ: carbamazepine; CZP: clonazepam; FBM: felbamate; GBP: gabapentin; LCM: lacosa FBM: felbamate; CZP: clonazepam; : potassium; filtration rate; CBZ: carbamazepine; GFR: glomerular + Mechanisms of action of AEDs (in alphabetical order) and pharmacokinetic characteristics. Based on [5–8]. Based AEDs (in alphabetical order) and pharmacokinetic characteristics. of action Mechanisms dosage range side effects (mg/day) (mg/l) LEV 1000–3000 3–30 Somnolence, asthenia, Binding to synaptic vesicle95 > 0. AED: antiepileptic drug; V AED: antiepileptic drug; FBM 1200–3600 30–100GBP 900–3600 Hepatic SJSdisturbance, 2–20LCM 200–400 and Na NMDA 10–20 of gain, worsening Weight Dizzi Blocks Ca CZP 0.5–4 0.02–0.08 ef Sedation, cognitive LTG 200–600 1–15 head-TEN, DRESS, Rash, SJS, Blocks vo PB 30–180 15–40 impairedcity, Rash, hepatotoxi receptor agonist, GABA 80–100 1–3 VPA 500–2500 50–100ZON thrombo- and Hepatotoxicity, 200–600GABA affect 20–30 Uncertain,may 95 > Somnolence, ataxia, dizziness,Na Blocks 1–10 glucuronidation, oxidation, Hepatic 0.15 8– 0.006– 8 CBZ 400–1600 4–12 Leukopenia, Table 1. 1. Table AED Usual Therapeutic Common/important PGB 150–600 2–8 TPM: topir epidermal necrolysis; TEN: toxic syndrome; PGB: pregabalin; SJS: Stevens-Johnson phenytoin; PB: phenobarbitone; PHT: PHT 150–400 10–20 hepatitis, SJS, Blood dyscrasia, vo Blocks calcium; K TPM 100–500 2–20 Impaired cognition, hepato- Blocks Na VGB 200–300 0.8–36 field def Visual OXC 900–2400 10–35 Somnolence, headache, Blocks

16 EUR ASSOC NEUROONCOL MAG 2012; 2 (1) Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours other drugs. Relevant side effects include drug interactions, sence of interactions with other agents. It is being excreted by hepatotoxicity, skin reactions including Stevens-Johnson syn- the kidney, and in the presence of renal dysfunction, dose ad- drome (SJS), leukopenia, bone marrow dyscrasia, and hy- justment is necessary. Side effects include irritability and psy- ponatraemia. chosis; blood dyscrasia is rare.

Valproic acid (VPA) is a broad-spectrum anticonvulsant for Pregabalin (PGB) is a third-generation anticonvulsant that both generalized and partial epilepsy with rather mild toxic- does not show pharmacokinetic interactions with enzyme-in- ity. Recent reports indicate that it may also have anti-tumour ducing or -inhibiting drugs. To date, there is little experience effects in glioblastoma multiforme and other types of cancer, with pregabaline as an anticonvulsant, including its applica- possibly secondary to its action as a histone-deacetylase in- tion in brain tumour patients. In patients with renal dysfunc- hibitor [9–11]. In children and adults with high serum levels, tion, dose adjustment is necessary. one should be aware of a disturbed haemostasis; monitoring of liver functions, platelet counts, and coagulation parameters Lacosamide (LCM) selectively enhances slow sodium chan- is advisable. The impact of these abnormalities is uncertain as nel inactivation. Because of the absence of drug interactions, clinical studies have not indicated enhanced bleeding tenden- lacosamide seems a promising AED in cancer. Presently, it is cies following neurosurgery. Nevertheless, in patients with registered for use as an add-on AED. Side effects include cog- cancer, one should be aware of drug-drug interactions and nitive dysfunction and skin reactions. side effects including hepatotoxicity, disturbed haemostasis, bone marrow suppression, and skin reactions, ie, rash, SJS, or Zonisamide (ZON) is a broad-spectrum anticonvulsant and is toxic epidermal necrolysis (TEN). metabolised by conjugation with glucuronic acid. It does not inhibit or induce hepatic co-enzymes, however, other enzy- Clonazepam (CZP) is a benzodiazepine, and a broad-spec- me-inducing drugs, like PHT and PHB, can enhance its me- trum and effective anticonvulsant. Because its use may easily tabolism. Side effects like weight loss and cognitive distur- lead to sedation and tolerance, its application is generally re- bances are of concern in patients with brain tumours. stricted to the abrogation of acute seizures, in status epilepti- cus, or as a last resort therapy when other antiepileptic drugs have failed.  Drug Interactions Lamotrigine (LTG) represents a first-choice anticonvulsant Drug interactions are characterized as either pharmacokinetic for symptomatic partial epilepsy. However, although lamo- (when uptake, distribution, metabolism, or excretion is af- trigine by itself has only weak inducing and inhibiting activi- fected) or pharmacodynamic (when target organs or receptor ties of the P-450 co-enzyme system of the liver, LTG is sus- sites are affected). Pharmacokinetic drug-drug interaction ceptible to induction by concomitantly given drugs. A draw- _occurs when ≥ 2 drugs are administered simultaneously and back of lamotrigine is that the initiation of therapy requires a one drug modifies the metabolism of the other. In this way, long time period of dose increments before therapeutic ranges serum concentrations of co-administered drugs can become are reached. One should be aware of side effects in cancer reduced or elevated, leading to either ineffective therapy or patients such as skin reactions (SJS, TEN), bone marrow tox- drug toxicity. The most common interactions between anti- icity, and drug interactions. epileptic and cytostatic drugs are of a pharmacokinetic nature, and as a rule may occur when both are being metabolized by a Oxcarbazepine (OXC) has a close structural similarity to CBZ corresponding co-enzyme of the P450 CYP system, by epox- but it is better tolerated and shows fewer drug interactions. In ide oxidation, or by glucuronidation in the liver [16, 17]. Al- patients with cancer, one should be aware of hyponatraemia, though the CYP system consists of approximately 60 different skin reactions (SJS, TEN), and sometimes bone marrow sup- iso-enzymes, 5 of them (CYPs 3A4, 2D6, 2C9, 2C19, and pression. 1A2) are responsible for the metabolism of 95 % of all drugs, of which the CYPs 3A4, 2C9, and 2C19 (Table 2) are particu- Topiramate (TPM) is a second-generation and effective broad- larly important in relation to potential interactions of AEDs spectrum AED with no obvious drug interactions, although its [19, 20]. An overview of the different groups of chemothera- tolerability is lower than for many of the other newer AEDs. peutic agents and the interactions they may have with anti- In cancer patients, one should be aware of cognitive dysfunc- convulsants, most notably with enzyme-inducing AEDs, is tion, hepatic abnormalities, and blood dyscrasia (rare). given in Table 3. Gabapentin (GBP) is one of the second-generation anticon- vulsants, it is generally well-tolerated and shows no interac-  Compromised Activity of CTDs by Enzyme- tion with other drugs. However, it has a limited efficacy and sometimes a worsening of seizures may happen. In patients Inducing AEDs with renal dysfunction, dose adjustment is necessary. Alkylating Agents Felbamate (FBM) can lead to serious adverse events, like he- Cyclophosphamide is an alkylating agent and is commonly patic abnormalities or aplastic anaemia; its use is mainly re- administered in the treatment of malignant lymphomas, stricted to intractable types of epilepsy. leukaemias, neuroblastoma, retinoblastomas, and carcinomas of the ovary, breast, endometrium, and lung, usually in combin- Levetiracetam (LEV) has good anti-seizure efficacy, includ- ation with other chemotherapeutic drugs [25]. By itself, cy- ing in brain tumours [12–15]. Its major advantage is the ab- clophosphamide is inactive and requires enzymatic bioactiv-

EUR ASSOC NEUROONCOL MAG 2012; 2 (1) 17 Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours

Table 2. Mechanisms of metabolism of AEDs and their effects on the P450 hepatic CYP system. Based on [16, 18].

AED Metabolism Substrate (CYP) Inducer (CYP) Inhibitor (CYP) CBZ Cytochrome P450 1A2, 2C9, 2C19, 3A4 1A2 (s), 2B6 (s), 2C9 (s), 2C19 (s), 3A4 (s) – 3A4 (s) CZP Cytochrome P450 3A4 – – FBM Cytochrome P450 2E1 2C19, 3A4 (w) 2C19 GBP Not metabolised – – – LCM Partial cytochrome P450 2C19 – – LTG UDPGT glucuronidation – UDGPT (w) – LEV Non-hepatic hydrolysis – – – OXC UDPGT glucuronidation, limited – 2C19 (w), 3A4 (w) 2C19 (w) cytochrome P450 metabolism PB Cytochrome P450 2C9, 2C19, 2E1 1A2 (s), 2B6 (s), 2C8 (s), 2C9/C19 (s), 3A4 (s) – PHT Cytochrome P450 2C8, 2C9, 2C19 1A2, 2B6 (s), 2C9/19 (s), 3A4 (s) – PGB Not metabolised – – – TPM Mainly renal excretion, limited – 2C19 (w), 3A4 (w) 2C19 (w) cytochrome P450 metabolism, UDPGT glucuronidation VGB Not metabolised – – – VPA Cytochrome P450, UDPGT 2A6, 2B6, 2C9, 2C19 – 2C9, EH, UDPGT glucuronidation, β-oxidation ZON Cytochrome P450 2C19, 3A4, 3A5 – – AEDs: see Table 1; s: strong; w: weak; CYP: cytochrome P450 enzyme; UDPGT: uridine diphosphate glucuronyltransferase ation in the liver and various CYP iso-enzymes have been Antimetabolites demonstrated to be involved, including 3A4/5 and 2C9/19 Methotrexate is an antimetabolite that acts as a folate antago- [25]. PB, PHT, and CBZ can induce the clearance of cyclo- nist, it is widely applied in different types of cancer, including phosphamide via its action on enzyme-inducing iso-enzymes leukaemia, lymphomas, and breast cancer. Methotrexate in- [62]. As a faster metabolism may also lead to an increased hibits dihydrofolate reductase, leading to reduced synthesis of exposure to active metabolites, this combination can also re- DNA, RNA, and proteins [67]. Its elimination is mainly renal, sult in toxicity [22]. Lower plasma concentrations of ifosfa- and 80–90 % is unchanged excreted in urine [68]. In a retros- mide have also been observed in combination with PHT, pective study, 40 of 716 children with acute lymphoblastic probably due to enhanced activity of CYP2B6 [24]. leukaemia receiving PB, PHT, or CBZ had a lower plasma clearance of methotrexate and teniposide, together with a Busulphan is an alkylating agent that is applied with autolo- worse event-free survival, more haematological and CNS re- gous bone marrow transplantation. Concomitant prescription lapses, and a shorter survival [26]. In a small retrospective of AEDs is recommended because busulphan gives rise to study, blood levels of methotrexate were lower in patients re- epileptic seizures. However, administration of PHT may re- ceiving EIAEDs as compared to either the non-EIAED topira- sult in reduced steady-state levels of busulphan [63]. mate or not receiving any anticonvulsant [27]. The exact mechanism of interaction between the AEDs methotrexate Antimitotic Cytostatics and EIAEDs is not known. Vinca alkaloids, including vinblastine, vincristine, and vindesine, are largely metabolized by 3A4/5 iso-enzymes [64, Topo-Isomerase Inhibitors 65]. Systemic clearance (Cl) of vincristine is 63 % higher, the Etoposide and teniposide are used in non-small-cell lung car- elimination half-life (T1/2) 35 % shorter, and the total area- cinoma (NSCLC) and acute myeloid leukaemia, (non-) Hodg- under-the-plasma-concentration-time curve (AUC) 43 % smaller kin’s lymphoma, acute lymphoblastic leukaemia, and in au- in patients receiving PTH or CBZ, probably as a result of an tologous bone marrow transplantation, and are mainly me- accelerated metabolism of vincristine [39]. tabolized by CYP 3A4 [64]. The Cl of etiposide is about 77 % faster in patients on concomitantly administered EIAEDs Taxanes [38]. Concomitant use of PB or PTH leads to a 2–3-fold in- Paclitaxel is widely used in different types of cancer, includ- crease of Cl of teniposide, which may thus compromise its ing lung, breast, and ovarian carcinoma and acts by stabi- efficacy [37]. lisation of tubulin polymerization. Its metabolism takes place through oxidative metabolism mediated by 3A4/5 and 2C8 The camptothecin analogue topotecan is mostly used in re- [66]. In combination with enzyme-inducing AEDs, the maxi- fractory small-cell lung cancer and ovarian cancer, and co- mum tolerated dose (MTD) of paclitaxel is 43–50 % higher administration of PHT leads to a 47-% increase of Cl and and depends to some extent on the way of administration [34, fewer haematological side effects [69]. For 9-aminocampto- 35]. tecin, plasma concentration levels are 3× lower with concomi-

18 EUR ASSOC NEUROONCOL MAG 2012; 2 (1) Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours

Table 3. Interactions between AEDs and CTDs

Table 3a. AED that reduce CTD efficacy Group CTD Type of CTD AED that reduces CTD Effect Reference Alkylating agents Busulphan PHT Cl 15 % ↑, AUC 16 % ↓, [21]** ↓ T1/2 23 % Cyclophos- PB, PHT AUC 67 % ↓, biotransformation [22]* [23]* phamide 200–300 % ↑ Ifosfamide PHT AUC ↓ (ns), Cl ↑ (ns) [24]* Thiotepa PHT AUC 29 % ↓ [25]*

Antimetabolites Methotrexate PB, PHT, CBZ Cl ↑ (ns), Cpl 90 % ↓ [26]*** [27]* ↓ ↑ Antimitotic agents 9-Amino- PB, PHT, CBZ Cpl 67 % , MTD 109 % , [28]*** [29]** camptotecin Cl 122 % ↑ Irinotecan PB, PHT, CBZ, PRM, OXC MTD 160–250 % ↑, Cl 61 % ↑ [30]*** [31]*** [32]*** Topotecan PHT Cl 47 % ↑ [33]*** Paclitaxel EIAEDs (ns) MTD 43–57 % ↑, MTD 50 % ↑, [34]*** [35]*** [36]*** Css 55 % ↓ Teniposide PB, PHT Cl 146 % ↑ [37]* Etoposide AEDs (ns) Cl 77 % ↑ [38]* Vincristine PHT, CBZ Cl 63 % ↑, AUC 43 % ↓ [39]** Proteinkinase inhibitors Bortezomib EIAEDs (ns) MTD 47 % ↑ [40]**

Enzastaurin EIAEDs (ns) Cpl 80 % ↓ [41]** Erlotinib EIAEDs (ns) AUC 55 % ↓, MTD 150–333 % ↑ [42]*** [43]** Gefitinib PB, PHT, CBZ, PRM, OXC AUC 60 % ↓, MTD 300 % ↑ [44]**

Imatinib PHT, CBZ, PRM, OXC, TPM Cpl 66–68 % ↓, MTD 50 % ↑ [45]*** [46]***

Sorafenib EIAEDs (ns) Cpl ↓ [47]** Tipifarnib PB, PHT, CBZ AUC 83 % ↓, MTD 100 % ↑ [48]***

Vatalinib PB, PHT, CBZ, PRM, OXC Cl 200 % ↑, Cmax 67 % ↓ [49]**

Temsirolimus PB, PHT, CBZ, OXC Cmax 73 % ↓, MTD 47 % ↑, Cpl 33 % ↓ [50]*** [51]** Sirolimus PB, PHT, CBZ, OXC AUC 39 % ↓, MTD 100 % ↑ [52]*** [53]** Everolimus EIAEDs (ns) Lower C (ns) [50]*** [51]**

Table 3b. Increase of toxicity CTD AED that increases toxicity of CTD Result Reference CCNU (Lomustine) VPA Frequency of haematological toxicity 60 % ↑ [9]*** AED CTD that increase toxicity of AED Effect Reference ↑ PHT 5-Fluorouracil Cpl (PHT) 170–225 % [54]* [55]* Capecitabine Cpl (PHT) 168–171 % ↑ [55]* [56]*

Doxifluridrine Cpl (PHT) 400 % [57]*

Erlotinib Cpl (PHT) 322–406 % ↑ [58]*

Tamoxifen Cpl (PHT) 44 % [59]* VPA Fotemustine/Cisplatin Frequency of haematological toxicity 300 % ↑ [60]**** PHT, PB, CBZ, VPA Procarbazine Frequency of skin hypersensitivity reactions 390 % ↑ [61]**** (as combination therapy) CTD: chemotherapeutic drug; AED: antiepileptic drug (abbreviations: see Table 1); Cl: clearance; AUC: area-under-the-concentration-time curve; T1/2: elimination half-life; ns: not specified; C: concentration; EIAED: enzyme-inducing antiepileptic drug; Cpl: plasma concentration; MTD: maximum tolerated dose; PRM: primidone; TD: tolerated dose; Css: steady-state concentration; Cmax: peak plasma concentration. case report (1–4 patients); ** small series (4–19 patients); *** large series (20–49 patients); **** large prospective series (> 50 patients) tant EIAEDs [28]. Co-medication with EIAEDs results in an ways, including tyrosine-kinase inhibitors and angiogenic in- increase in clearance of 27–57 % and a decrease in bioavai- hibitors. The metabolism of these drugs is often influenced by lability of 75 % of irinotecan [28]. concomitant administration of anticonvulsants.

Other Cytostatic Drugs Erlotinib is an inhibitor of the epidermal growth factor re- Ixabepilone is a novel microtubule-targeting agent with anti- ceptor (EGFR) and is used in NSCLC, and mainly metabo- tumour activity against ovarian, colon, cervical, gastric, breast, lized via the iso-enzymes 3A4, 3A5, and to a lesser extent by melanoma, NSCLC, and non-Hodgkin’s lymphoma. Together 1A2 [43]. The MTD for erlotinib is 200 mg/day without en- with EIAEDs, the MTD is 41 % and the Cl 50 % higher, prob- zyme induction and more than 2–3× higher (450–650 mg/ ably by induction of 3A4 [70]. day) together with an EIAED [42, 43].

Proteinkinase Inhibitors Another tyrosine-kinase receptor inhibitor is the small mol- Research into the development of gliomas and other carcino- ecule gefitinib that is used in NSCLC and is metabolized by mas has improved the understanding of specific cellular, mo- 3A4, 3A5 co-enzymes, and the non-inducible CYP2D6. For lecular, and genetic mechanisms that lead to cancer growth patients using an EIAED, the peak dose of gefitinib is –45 %, and progression. To date, many agents can target these path- the AUC –60 % and the MTD 4× higher (1000 mg/d) as com-

EUR ASSOC NEUROONCOL MAG 2012; 2 (1) 19 Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours pared to an MTD of 250 mg/day without EIAEDs [44]. cause drug-drug interactions because of enzyme-inducing ef- Imatinib, another selective inhibitor of tyrosine-kinase, is ad- fects on the 3A4 iso-enzyme [72]. Vice versa, they are suscep- ministered in chronic myeloid leukaemia and with gastro- tible to agents affecting 3A4; PB, PHT, and CBZ enhance the intestinal stromal tumours, and largely metabolized by clearance of dexamethasone 2–4× [5, 73, 74]. Hydrocortisone CYP3A4 [46]. Mean trough levels of imatinib are 2.9× lower is metabolized via the hepatic 11-B-hydroxysteroid dehydro- with an MTD of 1200 mg/day for patients on EIAEDs as com- genase system, which is also susceptible to enzyme induction. pared to 800 mg/day normally [45, 46]. For sorafenib, which PB, PHT, and CBZ induce the metabolism of prednisolone is mostly used in advanced renal cell carcinoma and hepato- with enhanced clearances of 79 %, 41 % and 77 %, respec- cellular carcinoma, the peak concentration (Cmax) is –54 % tively, and the metabolism of methylprednisolone more than and the AUC –63 % for patients on EIAEDs [47]. 3.4, 1.5, and 2×, respectively [75]. Vice versa, dexamethasone given together with PHT may lead to an increased clearance Tipifarnib is currently under investigation, mostly in blood and of PHT and therefore with a lesser anti-epileptic activity. In- breast cancers. A phase-I trial of tipifarnib in patients with re- creased seizure frequency has indeed been observed with co- current glioma using EIAEDs showed that the MTD is 2× therapy of dexamethasone with 38–50 % lower serum PHT higher, and the AUC approximately 2× lower than for patients levels [76–78]. When dexamethasone is discontinued, PHT not on EIAEDs. It is unclear to what extent either glucuroni- concentrations can easily rise to toxic levels [79]. dation or oxidative metabolism by P450 co-enzymes is respon- sible for this decrease in systemic exposure [48]. These observations emphasize that it is equally important to apply dose adjustment not only at the time of initiating medi- Bortezomib is applied in multiple myeloma and in recurrent cation but also when interacting co-therapy is discontinued. mantle-cell lymphoma. The use of EIAEDs can lead to an ac- However, also increased levels of PHT have been observed celeration of metabolism requiring 2× higher doses of with co-medication of dexamethasone. Apparently, the use of bortezomib [40]. dexamethasone can lead to unpredictable interactions with PHT, including either enzyme-inducing or -inhibiting effects. In enzastaurin, a VEGFR inhibitor, which is investigated in B-cell lymphoma, chronic lymphatic leukaemia, and solid tu- From these observations, one may conclude that simultaneous mours including glioblastoma, simultaneous EIAED use use of enzyme-inducing anticonvulsants and corticosteroids leads to approximately –80 % AUC [41]. should rather be avoided, or, when given simultaneously, that plasma concentrations of anticonvulsants should be monitored. Temsirolimus is an ester of the immunosuppressive agent sirolimus and inhibits the mammalian target of rapamycin. It  Idiosyncratic and Skin Reactions is mainly used in metastatic renal cell carcinoma and under investigation in glioblastoma multiforme. Sirolimus is an im- Bone marrow suppression and liver dysfunction can occur as munosuppressant agent mostly given to transplant patients, a side effect of a number of individual AEDs, and are more and may have anti-tumour activity against malignant gliomas frequently seen in association with brain tumours than with [53]. Both temsirolimus and sirolimus constitute substrates of other co-morbidities [4]. This can be explained by overlap- iso-enzymes 3A4/5. In one study, the peak concentration of ping adverse events of CTDs, which are often more pro- temsirolimus was –73 % and of sirolimus –47 % and the AUC nounced with the concomitant use of AEDs. For example, –50 % with EIAED co-therapy. Grade-3 haematological tox- haematotoxicity can be the result of administration of PB, icity was more commonly seen (20 vs 3 %) in patients on non- PHT, CBZ, or VPA, but is also common with use of temozolo- EIAEDs [50, 53]. Also, everolimus concentrations are lower mide and other chemotherapeutic agents [80, 81]. with concomitant 3A4-inducing AEDs [71]. Also, severe systemic allergic reactions can occur with the use  Increased Toxicity by AEDs or CTDs of a number of AEDs, mainly at the time of initiation of therapy with PB, PHT, CBZ, OXC, VPA, or LTG [82]. The Increased toxicity of AEDs may occur when enzyme-inhibit- most common type of hypersensitivity is skin rash as a side ing CTDs are concurrently prescribed. Toxicity of PHT has effect of AEDs, which can be accompanied by fever, lym- been observed when combined with 5-fluorouracil, tamoxi- phadenopathy, and, occasionally, by multi-organ abnormali- fen, capecitabine, erlotinib, or doxifluridine [55, 57–59]. The ties [83]. Its frequency seems higher in patients carrying a underlying mechanism of these interactions is probably a brain tumour than in non-tumoural patients with epilepsy [84, competitive inhibition of PTH clearance by 2C9 and 2C19 85]. More severe allergic reactions are erythema multiforme iso-enzymes. The use of the alkylating agent procarbazine in or the Stevens-Johnson syndrome and toxic epidermal necro- combination with PB, PTH, CBZ, or VPA can lead to hyper- lysis with necrosis and blistering of skin and mucosal mem- sensitivity reactions of the skin. In a fotemustine-cisplatin branes, which may lead to a potential mortality of up to 40 % regimen, the use of VPA leads to a 4-fold increase of thrombo- [86]. A number of observations strongly suggests an increased penia, neutropenia, or both [60]. appearance of SJS in patients undergoing cranial irradiation and receiving anticonvulsants, particularly with PB, PHT, or  Corticosteroids CBZ, often together with co-therapy with glucocorticosteroids [87, 88]. Nevertheless, the appearance of this severe dermato- Glucocorticosteroids are frequently used in patients with logical complication is relatively rare. In one retrospective brain tumours and systemic cancer, their administration can review of 289 patients receiving radiotherapy and anticonvul-

20 EUR ASSOC NEUROONCOL MAG 2012; 2 (1) Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours sants, only 1 patient developed SJS [85]. Recently, the human  Conclusion leukocyte antigen (HLA) class 1 allele B*1502 was associ- ated with 100 % of patients with CBZ-induced SJS, and only Today, patients with brain tumours undergo intensive therapy, in 3 % of CBZ-tolerant and in 8.6 % of controls in an Asian including neurosurgery, radiation therapy, and chemotherapy, population [89]. PHT and OXZ have been associated with se- at various stages of the disease. Besides, the majority of pa- vere skin reactions in HLA-B*1502 carriers [90]. In Europe, tients needs anticonvulsants to control epileptic seizures, and one has observed that the HLA-A*3101 allele increases the often glucocorticosteroids or other ancillary therapies to con- risk of developing a CBZ-induced hypersensitivity from 5 % trol symptoms secondary to tumoural effects or to anti-tu- to 26 % [91]. By screening patients on specific HLA alleles mour therapy. For these reasons, this multitude of co-treat- before prescribing anticonvulsants these sometimes life- ments easily leads to a myriad of side effects in patients with threatening skin reactions can possibly be prevented [92]. brain tumours, drugs used simultaneously may either aggra- vate or counteract each other. The use of anticonvulsant drugs illustrates each of these situations, thus – apart from seizure  Cognition control – a lot of attention is needed for avoiding or neutraliz- Besides pharmacokinetic effects, antiepileptic drugs can also ing these side effects in patients with brain tumours. Obvi- influence cognitive functioning. The conventional anticon- ously, the first issue in prescribing anticonvulsants is to take vulsants PB, PHT, CBZ, and VPA are all associated with cog- into account pharmacokinetic properties and common side nitive side effects showing impairments in memory, mental effects (Table 1). Another point of attention is the issue of rec- speed, and attention with a less favourable cognitive profile of ognition of interactions with chemotherapy due to the en- PB as compared to other anticonvulsants [93–95]. Although zyme-inducing or -inhibiting effects of conventional AEDs these side effects are mild or moderate [93], their impact can (Table 2). The substantial effect that enzyme-inducing AEDS be substantial if patients already suffer from cognitive dys- can have on the treatment of brain tumours is reflected by the function due to space-occupying effects of the tumour or by large differences of dose regimens of chemotherapeutic the sequelae of neurosurgery, radio-, and chemotherapy [96]. agents, depending on whether or not patients receive an en- As neurocognitive decline interferes with quality of life, such zyme-inducing AED. Prospective phase-I/II trials on CTDs changes may have great impact on the daily life of patients divided in strata with or without EIAEDs have produced cor- and family members [97]. responding data on drug clearance, area-under-the-curve val- ues, and median or maximum tolerated doses of new cancer In a group of 156 long-time survivors of low-grade gliomas, a agents, and illustrate the strong enzyme-inducing capabilities higher seizure frequency was associated with mental deterior- of phenobarbital, phenytoin, and carbamazepine (Table 3). ation. The use of one of the conventional anticonvulsants led to a worse performance in 6 out of 7 cognitive domains [98]. Nevertheless, these effects are not so easily detected as they Particularly, an inverse relationship was observed between are mainly manifested by a potential inefficacy of chemothe- anti-epileptic polytherapy with AEDs, ie, the number of anti- rapeutic or targeted drugs on the underlying tumour. As many convulsants taken simultaneously had an unfavourable effect types of brain tumours including low- and high-grade gliomas on executive and psychomotor functioning in patients with or systemic cancers often become resistant to anti-tumour-di- low-grade gliomas and hypothalamic hamartomas [98–100]. rected therapy, a poor response to therapy is easily accounted for by ineffective therapy rather than by co-treatment with an enzyme-inducing agent. Likewise, chemotherapeutic agents In general, the newer AEDs like GBP, LTG, OXC, PGB, and often have enzyme-inducing or -inhibiting prop- LEV confer less pronounced cognitive side effects, with the erties leading to either potential inefficacy or toxicity of exception of TPM (Table 4) [105–107]. A retrospective study antiepileptic drugs. comparing TPM (n = 429), LTG (n = 336), and LEV (n = 301) in non-brain tumour-related epilepsy showed that almost half By making use of pharmacokinetic data on drug interactions the patients using TPM discontinued its use because of mental (Tables 2, 3), one may argue that potential adverse interac- slowing in 27.8 % and dysphasia in 15 % [101, 108]. At the tions can be prevented by a timely dose adjustment according same time, almost 10 % of patients on LTG experienced posi- to the anticipated effect of these interactions. To do so reli- tive cognitive effects like improved alertness, better emo- ably, this will require therapeutic drug monitoring of the applied tional stability, or less irritability [108, 109]. In a study on 70 chemotherapy as well as anticipating and accounting for a patients with gliomas, patients on one of the conventional dose amendment at each change of the AED regimen. To what AEDs suffered from more adverse events than those on OXC extent this approach is indeed feasible in daily practise is un- (42.9 % vs 11.4 %) and psychomotor slowness was only seen certain, as medical oncologists taking care of cancer treat- with the traditional AEDs (21.7 %) [102]. In another study on ment not seldom work in other hospitals than neurologists 40 patients with seizures and gliomas, switching to leveti- prescribing and monitoring anticonvulsants. In practice, this, racetam monotherapy showed no negative effects on 6 do- makes proper adjustment of medication difficult to pursue or mains of cognition. LEV might even have positive effects as even elusive. Probably, the risks of harmful interactions take there were no signs of cognitive decline despite the presence place on a much larger scale than we assume, as physicians of a brain tumour or administration of ancillary anti-tumour probably often do not realise or are unaware of the possibility therapy [104]. Two smaller observational studies (including of interactions. 11 and 29 patients, respectively) showed mild cognitive im- pairment, it is unclear whether this is caused by the disease Obviously, the use of anticonvulsants may lead to interactions (ie, tumour progression) or by the use of LEV [14, 103]. not only with chemotherapeutic agents, but also with many

EUR ASSOC NEUROONCOL MAG 2012; 2 (1) 21 Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours

other kinds of drugs, for many other conditions or co-morbidi-

ive ties, which may accompany pa- tients with brain tumours. One

cognitive noteworthy side effect is the oc- or cognitive func- or cognitive Main drawbacks cognitive functioning follow-up Retrospective tioning, short of study of currence of severe dermatological complications, which is the result of an intricate interaction between anticonvulsants and the adminis- tration of cranial radiotherapy, possibly related to co-medication with steroids and expression of certain HLA alleles. otor slowness of 47 treated with Retrospect

een baseline and sub- as an abstract Apart from interactions, many differences in neuro-differences only So far, anticonvulsants have side effects psychological psychological assessment published sequent follow-up measure- sequent follow-up entering the study betw with traditional AEDs, no with traditional enced cognitive side effectsenced cognitive in comparison to 2/33 patientsAEDs prior to treated with other baseline cognitive functioning ments cognitive side effects with OXC side effects cognitive Cognitive Cognitive side effects and 1 on VPA) was seen only was VPA) and 1 on that can be extra damaging in pa- tients already at risk for organ dysfunction like bone marrow MEN, %, 4 patients on PB (21.7 assessment of suppression or liver dysfunction. LGA, AA,LGA, Psychom AA, GBM,out Two Compared to the second- and third-generation anticonvulsants, stoma multiforme; HGG: high-grade glioma; LGA: low-grade astrocytoma; low-grade HGG: high-grade glioma; LGA: stoma multiforme; LGO, LGO, hemangiopericy-giopericytoma, into functioning MBT diagnosis lymphoma AO, GBM, AO, LGG, HGG No the use of conventional anticon- vulsants PB, PHT, and CBZ more often leads to hepatic dysfunction and, to a lesser extent, also to bone marrow suppression. (months) Lastly, also the risk on neurotox- prospective: prospective: 16.5MBT MEN, experi- TMP monotherapy assessment of icity is of major importance in pa- tients with brain tumours and sei- zures, as both the need for anti- convulsants and the number of anticonvulsants used are each stronger contributing factors to opsychological 12 the severity of cognitive dysfunc- itive domains at base-itive line, 3, 6, and 12 months telephone phoma f Method of cognitive Mean follow- Tumour function assessment up tion than having gone through earlier neurosurgical excision or radiation therapy (Table 4). Al- though reviewing satisfactory sei- zure control is beyond the scope monotherapy Clinical notes Retrospective: 4.5; LGG, of this paper, the risk of ineffec- C monotherapy Clinical notes and 13.7 tive cancer treatment, organ dys- Methods collected retro- spectively their own controlstheir own n or as add-on side effects was effects side function, and neurotoxicity illus- trates the importance of effective and well-tolerated anticonvulsants

ational, TPM that do not interfere with cancer treatment. Fortunately, there is a Prospective LEV monotherapy, Neur follow-up patients served as assessment of 6 cog- retrospectiveAEDs vs traditional hospital charts number of anticonvulsants that possess these properties. Oxcar- T, bazepine, lamotrigine, and topira- mate have only weak enzyme-in- ducing or -inhibiting properties. Although valproic acid can have LTG, VPA, LEV, VPA, LTG, VPA 14 TPM 14 asadd-on (also treated with CBZ, PH PB, and prospective Information on PB, CBZ, PHT, PB, patients study OXC) enzyme-inhibiting activity and can lead to hepatic dysfunction

AEDof Number of Type and thrombopenia, its histone-

Cognitive side effects of AEDs in patients with brain tumours of side effects Cognitive deacetylase-inhibiting activity may help to control tumour progres- sion. Lastly, gabapentin, levetir- et al, 2011 al, et [104] et al, 2008[101] monotherapy, retrospective, Table 4. 4. Table Status. ephone Interview Cognitive for Tel TICS: metastatic MMSE: Mini Mental State brain tumour; Examination; oligodendroglioma; MEN: meningioma; MBT: LGO: low-grade De Groot LEV 40 Maschioet al, 2010 LEV[14] 29 Prospective follow-up LEV monotherapy, MMSE patients served as 8.1 controls their own AO, LGA, LGO, on MMSE Significant worsening MMSE provides AA, GBM, MEN, only partial insight et al. 2009[102] 35 controls on Maschio OXC vs 35 on OXC Observational, OX Maschio TMP 33 TPM Observ Useryet al, 2010[103] LEV 11 Prospective LEV monotherapy, weekly were Patients 1 patients served as interviewed by controls their own AA, GBM, LGO, %) were patients (18.2 Two Unknown mea- lym- MEN, MBT, cognitively impaired surement tools AED: antiepileptic drug (abbreviations see Table 1); AA: anaplastic astrocytoma; AO: anaplastic oligodendroglioma; GBM: gliobla AO: AA: anaplastic astrocytoma; 1); Table see (abbreviations AED: antiepileptic drug acetam, pregabalin, and lacosami-

22 EUR ASSOC NEUROONCOL MAG 2012; 2 (1) Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours de do all show no drug interactions. Future studies need to drug interactions. New Approaches to Brain Central Cancer Treatment Group Study. J Tumor Therapy Central Nervous System Con- Clin Oncol 2005; 23: 5294–304. show which of these agents or combinations would be the best sortium. J Clin Oncol 1997; 15: 3121–8. 51. Kuhn JG, Chang SM, Wen PY, et al. match to achieve effective seizure control together with good 35. Chang SM, Kuhn JG, Rizzo J, et al. Pharmacokinetic and tumor distribution tolerability. Phase I study of paclitaxel in patients with characteristics of temsirolimus in patients recurrent malignant glioma: a North Ameri- with recurrent malignant glioma. Clin Can- can Brain Tumor Consortium report. J Clin cer Res 2007; 13: 7401–6. Oncol 1998; 16: 2188–94. 52. Reardon DA, Quinn JA, Vredenburgh JJ, 36. Chang JW, Chang JH, Park SC, et al. et al. Phase 1 trial of gefitinib plus sirolimus Radiologically confirmed de novo glioblas- in adults with recurrent malignant glioma. 18. Patsalos PN, Perucca E. Clinically impor- References: toma multiforme and hippocampal sclerosis Clin Cancer Res 2006; 12: 860–8. tant drug interactions in epilepsy: general associated with the first onset of noncon- 1. Van Breemen MS, Wilms EB, Vecht CJ. features and interactions between antiepi- vulsive simple partial status epilepticus. 53. Reardon DA, Desjardins A, Vredenburgh Epilepsy in patients with brain tumours: epi- leptic drugs. Lancet Neurol 2003; 2: 347–56. Acta Neurochir (Wien) 2001; 143: 297–300. JJ, et al. Phase 2 trial of erlotinib plus demiology, mechanisms, and management. 19. Patsalos PN, Perucca E. Clinically impor- sirolimus in adults with recurrent glioblas- Lancet Neurol 2007; 6: 421–30. 37. Baker DK, Relling MV, Pui CH, et al. In- toma. J Neurooncol 2010; 96: 219–30. tant drug interactions in epilepsy: interac- creased teniposide clearance with concomi- 2. Singh G, Rees JH, Sander JW. Seizures tions between antiepileptic drugs and other tant anticonvulsant therapy. J Clin Oncol 54. Gilbar PJ, Brodribb TR. Phenytoin and and epilepsy in oncological practice: causes, drugs. Lancet Neurol 2003; 2: 473–81. 1992; 10: 311–5. fluorouracil interaction. Ann Pharmacother course, mechanisms and treatment. J Neurol 20. Gidal BE. Drug absorption in the elderly: 2001; 35: 1367–70. Neurosurg Psychiatry 2007; 78: 342–9. 38. Rodman JH, Murry DJ, Madden T, et al. biopharmaceutical considerations for the Altered etoposide pharmacokinetics and 55. Brickell K, Porter D, Thompson P. Pheny- 3. Van Breemen MS, Rijsman RM, Taphoorn antiepileptic drugs. Epilepsy Res 2006; 68 time to engraftment in pediatric patients un- toin toxicity due to fluoropyrimidines (5FU/ MJ, et al. Efficacy of anti-epileptic drugs in (Suppl 1): S65–S69. dergoing autologous bone marrow trans- capecitabine): three case reports. Br J Can- patients with gliomas and seizures. J Neurol 21. Hassan M, Oberg G, Bjorkholm M, et al. plantation. J Clin Oncol 1994; 12: 2390–7. cer 2003; 89: 615–6. 2009; 256: 1519–26. Influence of prophylactic anticonvulsant 39. Villikka K, Kivisto KT, Maenpaa H, et al. 56. Privitera M, de Los Rios la Rosa F. 4. Glantz MJ, Cole BF, Forsyth PA, et al. therapy on high-dose busulphan kinetics. Cytochrome P450-inducing antiepileptics in- Capecitabine-phenytoin interaction is dose Practice parameter: anticonvulsant prophy- Cancer Chemother Pharmacol 1993; 33: crease the clearance of vincristine in pa- dependent with an unexpected time course. laxis in patients with newly diagnosed brain 181–6. tients with brain tumors. Clin Pharmacol Anticancer Drugs 2011; 22: 1027–9. tumors. Report of the Quality Standards 22. De Jonge ME, Huitema AD, Rodenhuis Ther 1999; 66: 589–93. 57. Konishi H, Morita K, Minouchi T, et al. Subcommittee of the American Academy of S, et al. Clinical pharmacokinetics of cyclo- 40. Phuphanich S, Baker SD, Grossman SA, Probable metabolic interaction of doxifluri- Neurology. Neurology 2000; 54: 1886–93. phosphamide. Clin Pharmacokinet 2005; 44: et al. Oral sodium phenylbutyrate in patients dine with phenytoin. Ann Pharmacother 5. Patsalos PN. Properties of antiepileptic 1135–64. with recurrent malignant gliomas: a dose 2002; 36: 831–4. drugs in the treatment of idiopathic general- 23. Jao JY, Jusko WJ, Cohen JL. Phenobar- escalation and pharmacologic study. Neuro ized epilepsies. Epilepsia 2005; 46 (Suppl 9): bital effects on cyclophosphamide pharma- Oncol 2005; 7: 177–82. 58. Grenader TF, Gipps MF, Shavit LF, et al. 140–8. Significant drug interaction: phenytoin toxic- cokinetics in man. Cancer Res 1972; 32: 41. Kreisl TN, Kotliarova S, Butman JA, et 6. Johannessen SI, Battino D, Berry DJ, 2761–4. ity due to erlotinib. Lung Cancer 2007; 57: al. A phase I/II trial of enzastaurin in pa- 404–6. et al. Therapeutic drug monitoring of the 24. Ducharme MP, Bernstein ML, Granvil CP, tients with recurrent high-grade gliomas. newer antiepileptic drugs. Ther Drug Monit et al. Phenytoin-induced alteration in the N- Neuro Oncol 2010; 12: 181–9. 59. Rabinowicz AL, Hinton DR, Dyck P, et al. 2003; 25: 347–63. High-dose tamoxifen in treatment of brain dechloroethylation of ifosfamide stereoiso- 42. Prados MD, Lamborn KR, Chang S, et al. tumors: interaction with antiepileptic drugs. 7. Sills GJ, Brodie MJ. Update on the mecha- mers. Cancer Chemother Pharmacol 1997; Phase 1 study of erlotinib HCl alone and Epilepsia 1995; 36: 513–5. nisms of action of antiepileptic drugs. Epilep- 40: 531–3. combined with temozolomide in patients tic Disord 2001; 3: 165–72. 25. De Jonge ME, Huitema AD, van Dam with stable or recurrent malignant glioma. 60. Bourg V, Lebrun C, Chichmanian RM, et 8. Luszczki JJ. Third-generation antiepileptic SM, et al. Significant induction of cyclo- Neuro Oncol 2006; 8: 67–78. al. Nitroso-urea-cisplatin-based chemo- drugs: mechanisms of action, pharmacoki- phosphamide and thiotepa metabolism by 43. Raizer JJ, Abrey LE, Lassman AB, et al. therapy associated with valproate: increase netics and interactions. Pharmacol Rep 2009; phenytoin. Cancer Chemother Pharmacol A phase I trial of erlotinib in patients with of haematologic toxicity. Ann Oncol 2001; 61: 197–216. 2005; 55: 507–10. nonprogressive glioblastoma multiforme 12: 217–9. 9. Oberndorfer S, Piribauer M, Marosi C, et 26. Relling MV, Pui CH, Sandlund JT, et al. postradiation therapy, and recurrent malig- 61. Lehmann DF, Hurteau TE, Newman N, al. P450 enzyme inducing and non-enzyme Adverse effect of anticonvulsants on effi- nant gliomas and meningiomas. Neuro et al. Anticonvulsant usage is associated inducing antiepileptics in glioblastoma pa- cacy of chemotherapy for acute lymphoblas- Oncol 2010; 12: 87–94. with an increased risk of procarbazine hy- tients treated with standard chemotherapy. tic leukaemia. Lancet 2000; 356: 285–90. 44. Prados MD, Yung WK, Wen PY, et al. persensitivity reactions in patients with J Neurooncol 2005; 72: 255–60. 27. Riva M. Brain tumoral epilepsy: a re- Phase-1 trial of gefitinib and temozolomide brain tumors. Clin Pharmacol Ther 1997; 62: 225–9. 10. Weller M, Gorlia T, Cairncross JG, et al. view. Neurol Sci 2005; 26 (Suppl 1): S40– in patients with malignant glioma: a North Prolonged survival with valproic acid use in S42. American Brain Tumor Consortium Study. 62. Chen J, Ohnmacht C, Hage DS. Studies the EORTC/NCIC temozolomide trial for glio- 28. Grossman SA, Hochberg F, Fisher J, et Cancer Chemother Pharmacol 2008; 61: of phenytoin binding to human serum albu- blastoma. Neurology 2011; 77: 1156–64. al. Increased 9-aminocamptothecin dose re- 1059–67. min by high-performance affinity chromato- quirements in patients on anticonvulsants. 11. Li XN, Shu Q, Su JM, et al. Valproic acid 45. Wen PY, Yung WK, Lamborn KR, et al. graphy. J Chromatogr B Analyt Technol NABTT CNS Consortium. The New Ap- induces growth arrest, apoptosis, and se- Phase I/II study of imatinib mesylate for re- Biomed Life Sci 2004; 809: 137–45. proaches to Brain Tumor Therapy. Cancer current malignant gliomas: North American nescence in medulloblastomas by increas- 63. Hassan M, Oberg G, Bjorkholm M, et al. Chemother Pharmacol 1998; 42: 118–26. Brain Tumor Consortium Study 99-08. Clin ing histone hyperacetylation and regulating Influence of prophylactic anticonvulsant Cancer Res 2006; 12: 4899–907. expression of p21Cip1, CDK4, and CMYC. 29. Minami H, Lad TE, Nicholas MK, et al. therapy on high-dose busulphan kinetics. Mol Cancer Ther 2005; 4: 1912–22. Pharmacokinetics and pharmacodynamics of 46. Pursche S, Schleyer E, von Bonin M, et al. Cancer Chemother Pharmacol 1993; 33: 9-aminocamptothecin infused over 72 hours Influence of enzyme-inducing antiepileptic 12. Wagner GL, Wilms EB, Van Donselaar 181–6. in phase II studies. Clin Cancer Res 1999; 5: drugs on trough level of imatinib in glioblas- CA, et al. Levetiracetam: preliminary experi- 1325–30. toma patients. Curr Clin Pharmacol 2008; 3: 64. Vecht CJ, Wagner GL, Wilms EB. Inter- ence in patients with primary brain tumours. actions between antiepileptic and chemo- 30. Gilbert MR, Supko JG, Batchelor T, et al. 198–203. Seizure 2003; 12: 585–6. therapeutic drugs. Lancet Neurol 2003; 2: Phase I clinical and pharmacokinetic study 47. Reardon DA, Vredenburgh JJ, Desjardins 13. Maschio M, Albani F, Baruzzi A, et al. 404–9. of irinotecan in adults with recurrent malig- A, et al. Effect of CYP3A-inducing anti-epi- Levetiracetam therapy in patients with brain nant glioma. Clin Cancer Res 2003; 9: 2940– leptics on sorafenib exposure: results of 65. Leveque D, Jehl F. Molecular pharma- tumour and epilepsy. J Neurooncol 2006; 80: 9. a phase II study of sorafenib plus daily temo- cokinetics of catharanthus (vinca) alkaloids. 97–100. 31. Prados MD, Lamborn K, Yung WK, et al. zolomide in adults with recurrent glio- J Clin Pharmacol 2007; 47: 579–88. 14. Maschio M, Dinapoli L, Sperati F, et al. A phase 2 trial of irinotecan (CPT-11) in pa- blastoma. J Neurooncol 2011; 101: 57–66. 66. Spratlin J, Sawyer MB. Pharmacogenet- Levetiracetam monotherapy in patients with tients with recurrent malignant glioma: a 48. Cloughesy TF, Wen PY, Robins HI, et al. ics of paclitaxel metabolism. Crit Rev Oncol brain tumor-related epilepsy: seizure con- North American Brain Tumor Consortium Phase II trial of tipifarnib in patients with re- Hematol 2007; 61: 222–9. trol, safety, and quality of life. J Neurooncol study. Neuro Oncol 2006; 8: 189–93. current malignant glioma either receiving or 2011; 104: 205–14. 67. Motl S, Zhuang Y, Waters CM, et al. 32. Reardon DA, Egorin MJ, Quinn JA, et al. not receiving enzyme-inducing antiepileptic Pharmacokinetic considerations in the treat- 15. Kerrigan S, Grant R. Antiepileptic drugs Phase II study of imatinib mesylate plus hy- drugs: a North American Brain Tumor Con- ment of CNS tumours. Clin Pharmacokinet for treating seizures in adults with brain tu- droxyurea in adults with recurrent glioblas- sortium Study. J Clin Oncol 2006; 24: 3651– 2006; 45: 871–903. mours. Cochrane Database Syst Rev 2011; toma multiforme. J Clin Oncol 2005; 23: 6. 68. Balis FM. Pharmacokinetic drug interac- (8): CD008586. 9359–68. 49. Reardon DA, Egorin MJ, Desjardins A, tions of commonly used anticancer drugs. 16. Patsalos PN, Froscher W, Pisani F, et al. et al. Phase I pharmacokinetic study of the 33. Zamboni WC, Gajjar AJ, Heideman RL, Clin Pharmacokinet 1986; 11: 223–35. The importance of drug interactions in epi- et al. Phenytoin alters the disposition of vascular endothelial growth factor receptor lepsy therapy. Epilepsia 2002; 43: 365–85. topotecan and N-desmethyl topotecan in a tyrosine kinase inhibitor vatalanib (PTK787) 69. Burch PA, Bernath AM, Cascino TL, et al. 17. Vecht CJ, Wagner GL, Wilms EB. Treat- patient with medulloblastoma. Clin Cancer plus imatinib and hydroxyurea for malignant A North Central Cancer Treatment Group ing seizures in patients with brain tumors: Res 1998; 4: 783–9. glioma. Cancer 2009; 115: 2188–98. phase II trial of topotecan in relapsed glio- mas. Invest New Drugs 2000; 18: 275–80. Drug interactions between antiepileptic and 34. Fetell MR, Grossman SA, Fisher JD, et 50. Galanis E, Buckner JC, Maurer MJ, et al. chemotherapeutic agents. Semin Oncol al. Preirradiation paclitaxel in glioblastoma Phase II trial of temsirolimus (CCI-779) in re- 70. Peereboom DM, Supko JG, Carson KA, 2003; 30 (Suppl 19): 49–52. multiforme: efficacy, pharmacology, and current glioblastoma multiforme: a North et al. A phase I/II trial and pharmacokinetic

EUR ASSOC NEUROONCOL MAG 2012; 2 (1) 23 Spectrum of Side Effects of Anticonvulsants in Patients with Brain Tumours

study of ixabepilone in adult patients with 81. Su YW, Chang MC, Chiang MF, et al. 90. Hung SI, Chung WH, Liu ZS, et al. Com- 101. Maschio M, Dinapoli L, Zarabla A, et al. recurrent high-grade gliomas. J Neurooncol Treatment-related myelodysplastic syn- mon risk allele in aromatic antiepileptic- Outcome and tolerability of topiramate in 2010; 100: 261–8. drome after temozolomide for recurrent drug induced Stevens-Johnson syndrome brain tumor associated epilepsy. J Neuro- high-grade glioma. J Neurooncol 2005; 71: and toxic epidermal necrolysis in Han Chi- oncol 2008; 86: 61–70. 71. Krueger DA, Care MM, Holland K, et al. 315–8. nese. Pharmacogenomics 2010; 11: 349–56. Everolimus for subependymal giant-cell as- 102. Maschio M, Dinapoli L, Vidiri A, et al. trocytomas in tuberous sclerosis. N Engl J 82. Mockenhaupt M, Messenheimer J, 91. McCormack M, Alfirevic A, Bourgeois S, The role side effects play in the choice of Med 2010; 363: 1801–11. Tennis P, et al. Risk of Stevens-Johnson et al. HLA-A*3101 and carbamazepine-in- antiepileptic therapy in brain tumor-related syndrome and toxic epidermal necrolysis duced hypersensitivity reactions in Europe- 72. Li AP, Kaminski DL, Rasmussen A. epilepsy: a comparative study on traditional in new users of antiepileptics. Neurology ans. N Engl J Med 2011; 364: 1134–43. antiepileptic drugs versus oxcarbazepine. Substrates of human hepatic cytochrome 2005; 64: 1134–8. P450 3A4. Toxicology 1995; 104: 1–8. 92. Man CB, Kwan P, Baum L, et al. Associa- J Exp Clin Cancer Res 2009; 28: 60. 83. Schlienger RG, Shear NH. Antiepileptic tion between HLA-B*1502 allele and anti- 73. Brophy TR, McCafferty J, Tyrer JH, et al. 103. Usery JB, Michael LM, Sills AK, et al. drug hypersensitivity syndrome. Epilepsia epileptic drug-induced cutaneous reactions Bioavailability of oral dexamethasone dur- A prospective evaluation and literature re- 1998; 39 (Suppl 7): S3–S7. in Han Chinese. Epilepsia 2007; 48: 1015–8. ing high dose steroid therapy in neurological view of levetiracetam use in patients with 84. Rzany B, Correia O, Kelly JP, et al. Risk patients. Eur J Clin Pharmacol 1983; 24: 93. Aldenkamp AP. Effects of antiepileptic brain tumors and seizures. J Neurooncol of Stevens-Johnson syndrome and toxic 103–8. drugs on cognition. Epilepsia 2001; 42 2010; 99: 251–60. epidermal necrolysis during first weeks of (Suppl 1): 46–9. 74. Chalk JB, Ridgeway K, Brophy T, et al. antiepileptic therapy: a case-control study. 104. De Groot M, Toering S, Douw L, et al. Phenytoin impairs the bioavailability of dexa- Study Group of the International Case 94. Meador KJ. Cognitive and memory ef- Efficacy and tolerability of levetiracetam methasone in neurological and neurosurgi- Control Study on Severe Cutaneous Ad- fects of the new antiepileptic drugs. Epi- monotherapy in patients with primary brain cal patients. J Neurol Neurosurg Psychiatry verse Reactions. Lancet 1999; 353: 2190– lepsy Res 2006; 68: 63–7. tumors and epilepsy. Epilepsia 2009; 50 (Suppl 10): 101. 1984; 47: 1087–90. 4. 95. Park SP, Kwon SH. Cognitive effects of 75. Bartoszek M, Brenner AM, Szefler SJ. 85. Mamon HJ, Wen PY, Burns AC, et al. antiepileptic drugs. J Clin Neurol 2008; 4: 105. Bootsma HP, Ricker L, Diepman L, et al. Prednisolone and methylprednisolone kinet- Allergic skin reactions to anticonvulsant 99–106. Levetiracetam in clinical practice: long-term medications in patients receiving cranial experience in patients with refractory epi- ics in children receiving anticonvulsant the- 96. Bosma I, Reijneveld JC, Douw L, et al. radiation therapy. Epilepsia 1999; 40: lepsy referred to a tertiary epilepsy center. rapy. Clin Pharmacol Ther 1987; 42: 424–32. Health-related quality of life of long-term 341–4. Epilepsy Behav 2007; 10: 296–303. 76. Wong DD, Longenecker RG, Liepman M, high-grade glioma survivors. Neuro Oncol et al. Phenytoin-dexamethasone: a possible 86. Roujeau JC, Kelly JP, Naldi L, et al. 2009; 11: 51–8. 106. Blum D, Meador K, Biton V, et al. Cog- Medication use and the risk of Stevens- nitive effects of lamotrigine compared with drug-drug interaction. JAMA 1985; 254: 97. Bosma I, Vos MJ, Heimans JJ, et al. The Johnson syndrome or toxic epidermal topiramate in patients with epilepsy. Neu- 2062–3. course of neurocognitive functioning in necrolysis. N Engl J Med 1995; 333: rology 2006; 67: 400–6. 77. Gattis WA, May DB. Possible interaction 1600–7. high-grade glioma patients. Neuro Oncol involving phenytoin, dexamethasone, and 2007; 9: 53–62. 107. Aldenkamp AP, Baker G, Mulder OG, et 87. Delattre JY, Safai B, Posner JB. Ery- antineoplastic agents: a case report and re- al. A multicenter, randomized clinical study thema multiforme and Stevens-Johnson 98. Klein M, Engelberts NH, van der Ploeg view. Ann Pharmacother 1996; 30: 520–6. to evaluate the effect on cognitive function syndrome in patients receiving cranial ir- HM, et al. Epilepsy in low-grade gliomas: of topiramate compared with valproate as 78. Lawson LA, Blouin RA, Smith RB, et al. radiation and phenytoin. Neurology 1988; the impact on cognitive function and quality add-on therapy to carbamazepine in patients Phenytoin-dexamethasone interaction: a 38: 194–8. of life. Ann Neurol 2003; 54: 514–20. previously unreported observation. Surg with partial-onset seizures. Epilepsia 2000; 99. Correa DD, DeAngelis LM, Shi W, et al. Neurol 1981; 16: 23–4. 88. Micali G, Linthicum K, Han N, et al. In- 41: 1167–78. creased risk of erythema multiforme major Cognitive functions in low-grade gliomas: 79. Lackner TE. Interaction of dexametha- with combination anticonvulsant and ra- disease and treatment effects. J Neurooncol 108. Bootsma HP, Ricker L, Hekster YA, et al. sone with phenytoin. Pharmacotherapy diation therapies. Pharmacotherapy 1999; 2007; 81: 175–84. The impact of side effects on long-term re- 1991; 11: 344–7. 19: 223–7. tention in three new antiepileptic drugs. 100. Prigatano GP, Wethe JV, Gray JA, et al. Seizure 2009; 18: 327–31. 80. Hildebrand JF, Lecaille CF, Perennes JF, 89. Chung WH, Hung SI, Hong HS, et al. Intellectual functioning in presurgical pa- et al. Epileptic seizures during follow-up of Medical genetics: a marker for Stevens- tients with hypothalamic hamartoma and re- 109. Aldenkamp AP, de Krom M, Reijs R. patients treated for primary brain tumors. Johnson syndrome. Nature 2004; 428: fractory epilepsy. Epilepsy Behav 2008; 13: Newer antiepileptic drugs and cognitive Neurology 2005; 65: 212–5. 486. 149–55. issues. Epilepsia 2003; 44 (Suppl 4): 21–9.

24 EUR ASSOC NEUROONCOL MAG 2012; 2 (1)