Abstract
Patients with cancer commonly experience seizures. Combined therapy with anticonvulsant drugs (AEDs) and chemotherapeutic drugs or tyrosine kinase inhibitors carries inherent risks on drug-drug interactions (DDIs). In this review, pharmacokinetic studies of AEDs with chemotherapeutic drugs, tyrosine kinase inhibitors, and glucocorticoids are discussed, including data on maximum tolerated dose, drug clearance, elimination half-life, and organ exposure. Enzyme-inducing AEDs (EIAEDs) cause about a 2-fold to 3-fold faster clearance of concurrent chemotherapeutic drugs metabolized along the same pathway, including cyclophosphamide, irinotecan, paclitaxel, and teniposide, and up to 4-fold faster clearance with the tyrosine kinase inhibitors crizotinib, dasatinib, imatinib, and lapatinib. The use of tyrosine kinase inhibitors, particularly imatinib and crizotinib, may lead to enzyme inhibition of concurrent therapy. Many of the newer generation AEDs do not induce or inhibit drug metabolism, but they can alter enzyme activity by other drugs including AEDs, chemotherapeutics and tyrosine kinase inhibitors. Glucocorticoids can both induce and undergo metabolic change. Quantitative data on changes in drug metabolism help to apply the appropriate dose regimens. Because the large individual variability in metabolic activity increases the risks for undertreatment and/or toxicity, we advocate routine plasma drug monitoring. There are insufficient data available on the effects of tyrosine kinase inhibitors on AED metabolism.
Keywords: anticonvulsants, cancer, chemotherapeutic agents, drug interactions, epilepsy, glioma, glucocorticoids, tyrosine kinase inhibitors
Many patients with cancer experience seizures. Two-thirds or more of patients with gliomas and one-third of patients with meningiomas have epilepsy.1,2 For patients with systemic cancer, the overall incidence of epilepsy is higher. Seizures develop in up to 60% of patients who have a brain metastasis, depending on the primary tumor. They can also be secondary to metabolic or toxic encephalopathies, or other conditions associated with cancer.3 As a rule, this necessitates anticonvulsant drugs to be given alongside anti-tumor therapy such as chemotherapy. Combining these therapies confers the risk of drug-drug interactions, with 6 times higher risk in brain tumors as opposed to systemic cancer.4
Pharmacokinetic drug-drug interactions are due to changes in absorption, distribution, metabolism, or elimination of drugs. Pharmacodynamic drug-drug interactions become manifest when drugs share characteristics related to drug-receptor binding. In daily practice, existing insights mainly relate to pharmacokinetic effects secondary to upregulation or downregulation of coenzymes belonging to the cytochrome P450 (CYP450) or UGT glucuronidation systems in the liver. Of a total of 20 CYP isoenzymes, 2C9 and 3A4 cover about 60% of all metabolic reactions.5 These reactions are mediated by ligand-dependent nuclear receptors, including PXR (pregnane-X receptor), GC (glucocorticoid) receptor, and CAR (constitutive androstane receptor), which, after exposure to the inducing agent, are translocated into the cellular nucleus and become activated.5 Phenytoin, phenobarbital, and carbamazepine represent enzyme inducers, mainly of 2C9, 2C19, and 3A4 together with a number of long-term metabolic effects.6 Enzyme induction results in faster digestion of concurrently administered drugs metabolized along the same pathway, including chemotherapeutic agents, tyrosine kinase inhibitors, and glucocorticoids. Valproic acid, eslicarbazepine acetate, oxcarbazepine, perampanel, and topiramate occasionally show enzyme inhibition depending on the CYP or UGT enzymes involved, leading to toxicity of concomitant drugs, unless dose adjustment is applied.
Therapy with chemotherapeutic agents and tyrosine kinase inhibitors may similarly affect the pharmacokinetics of concurrent therapy. Both can cause enzyme induction, and tyrosine kinase inhibitors may also increase the toxicity of concurrent drugs via enzyme inhibition. Corticosteroids, probably the most commonly used drugs in neuro-oncology, can both provoke and undergo metabolic interaction. There exists large individual variability in drug metabolism depending on CYP enzyme susceptibility, age, sex, and ethnicity, all of which contribute to the risk of drug-drug interaction. An overview of the various reciprocal interactions between AEDs, chemotherapeutic drugs, tyrosine kinase inhibitors, and corticosteroids as reported in systemic cancer and neuro-oncology is discussed here, and these interactions are presented in quantitative terms regarding maximal tolerated dose, clearance, half-life, and area under the curve (AUC).
Methods
This review on drug-drug interactinos between AEDs and chemotherapeutics, tyrosine kinase inhibitors, and glucocorticoids is based on published articles identified via searches in PubMed, last searched in January 2015, limited to the English language. Primary sources were preferred, although occasionally review articles were used. Search terms included each of the generic names of the anticonvulsant drugs registered for focal epilepsy in adults AND “chemotherapy” OR “tyrosine kinase inhibitor” OR “corticosteroid” OR “glucocorticoid” AND/OR “interaction” OR “pharmacokinetics.” Separate searches were also carried out for each of the generic names of anticonvulsant drugs registered for focal epilepsy in adults AND “glioma” OR “brain tumor” OR “cancer” AND/OR “interaction” OR ”pharmacokinetics.” The anticonvulsant drugs explored were clobazam, clonazepam, eslicarbazepine, gabapentin, lacosamide, lamotrigine, levetiracetam, midazolam, oxcarbazepine, perampanel, phenobarbital, phenytoin, pregabalin, retigabine, tiagabine, topiramate, valproic acid, vigabatrin, and zonisamide. Separate searches were also carried out for each of the generic names of chemotherapeutic drugs and tyrosine kinase inhibitors AND “drug interaction” AND/OR “anticonvulsant” AND/OR “pharmacokinetics.” All reported clinical series on drug-drug interactions between AEDs and chemotherapeutic drugs, tyrosine kinase inhibitors, and glucocorticoids are presented. Single case reports and small series (n < 5) were included if no larger series were available, or if observations were relevant. For factual data on pharmacokinetic parameters of AEDs, CTDs, and TKIs as single agents representative reviews were consulted. This review has been published in a preliminary version.7
Results
Pharmacokinetic Characteristics of AEDs
Table 1 lists the pharmacokinetic properties of anticonvulsants indicated for the focal type of epilepsy in adults, thus also representing anticonvulsants applied for seizures associated with brain tumors or with neurological complications of systemic cancer.8,9 Characteristics include dose, therapeutic plasma range, elimination half-life, protein binding, and clearance with and without enzyme induction.7,10–12 In low-grade and high-grade glioma, more than 50% of patients need more than one anticonvulsant drug for seizure control, carrying risks of drug interactions.13,14 Although newer generation AEDS have fewer enzyme-inducing effects than the classical AEDs (phenobarbital, phenytoin, carbamazepine), one does not always realize that as drug substrates they are often susceptible to the metabolic effects of other agents including AEDs. With concurrent phenytoin and carbamazepine (acting on 2C9, 2C19, 3A4), the clearance of lamotrigine, oxcarbazepine, pregabalin, tiagabine, and zonisamide becomes a factor of 1.25 to 2.0 higher, and that of clobazam 2 to 3 times higher.15,16 Weak inducing effects can occur with the use of eslicarbazepine (3A4, UGT1A1) and lamotrigine (UGT1A4) if combined with a drug metabolized by the same coenzymes. Weak inhibiting effects are seen with eslicarbazepine (2C9, 2C19), oxcarbazepine (2C19), perampanel (2C8, UGT1A9) and topiramate (2C19), often without much clinical impact.11 Valproic acid is a enzyme inhibitor (UGT1A4), causing a doubling of the AUC of lamotrigine.17 All these agents are mainly metabolized by the liver. High protein-binding drugs such as phenytoin and valproic acid, and benzodiazepines including clobazam, clonazepam, and midazolam, may cause drug-drug interactions because of competition for binding with other strongly protein-linked agents. Gabapentin, levetiracetam, lacosamide, pregabaline, and vigabatrin are mainly renally eliminated, and thus much less involved in drug interactions. For further details on reciprocal interactions between AEDs, we refer to other reviews.10,11,15,18,19 Table 2 lists for each of the anticonvulsants, the co-enzymes responsible for substrate metabolism and enzymes that become induced or inhibited in their metabolic activity.11,12,15,16,20
Table 1.
AED | Usual Oral Dose (mg/day) | Therapeutic Range (mg/L) | Oral Bioavailability (%) | Time to Steady State (days) | T1/2 (h) without Enzyme Inducers | T1/2 (h) with Enzyme Inducers | Cl (mL/kg/h) without Enzyme Inducers | Cl (mL/kg/h) with Enzyme Inducers | Protein Binding (%) |
---|---|---|---|---|---|---|---|---|---|
Carbamazepine | 400–1600 | 4–12 | 75–85 | 2–4 | 8–20 | 9–10 | 133 | 80 | 75 |
Clobazam | 5–40 | 0.1–0.3 | 80–90 | 10–30 | 10–45 | 12–60 | 40–70 | bfactor 2–3 | 85 |
Clonazepamc | 0.5–4, max.20 | 0.02–0.08 | 90 | 17–56 | 1–4 | 1–3 | 90 | factor 1.2–1.6 | 85 |
Eslicarbazepine | 400–1200 | 3–35 | 90 | 3–4 | 20–24 | 9–20 | 35–50 | 40–65 | 40 |
Gabapentin | 900–3600 | 12–20 | <60 | 1–2 | 5–9 | 5–9 | 120–230 | 120–230 | 0 |
Lacosamide | 200–400 | 10–20 | >95 | 3 | 12–16 | 12–16 | 28–40 | 28–40 | <15 |
Lamotrigine | 200–600 | 2.5–15 | >95 | 3–15 | 22–38 | 14–15 | 20–35 | 50–80 | 55 |
Levetiracetam | 1000–3000 | 8–26 | 80–90 | 1–2 | 5–11 | 5–8 | 30–50 | 45–75 | 0 |
Midazolamd | in 5–15 | 100–400 | 30–70 | 1–2 | 1–4 | 1–2 | 250–540 | 12–30 | 95–97 |
Oxcarbazepine | 900–2400 | 12–35 | 80–100 | 2–3 | 5–30 | 6–19 | 40–50 | 50–70 | 40–60 |
Perampanel | 4–12 | 0.2–1.0 | 100 | 10–19 | 66–105 | ∼25 | 8–12 | 12–20 | 95% |
Phenobarbital | 30–180 | 10–40 | 95 | 15–30 | 70–140 | 80–100 | 6–9 | 12–18 | 45–60 |
Phenytoin | 150–400 | 10–20 | 95 | 6–21 | 24–100 | <24–72 | 7–42 | factor ∼2 | 85–95 |
Pregabalin | 150–600 | 2–8 | 90 | 1–2 | 5–7 | 5–7 | 45–75 | 45–75 | 0 |
Retigabine | 600–1200 | – | 60 | 1–2 | 6–10 | 4–7 | 670 | 470 | 80 |
Topiramate | 100–500 | 5–20 | 81%–95% | 4–7 | 20–30 | 9–12 | 15–36 | 30–50 | 15 |
Valproic acid | 500–2500 | 50–100 | >95 | 2–4 | 13–18 | 2–11 | 6–15 | 4–10 | 85–95 |
Vigabatrin | 200–300 | 0.8–36 | 80–90 | 1–2 | 6–8 | 4–6 | 130–150 | 130–150 | 0 |
Zonisamide | 200–600 | 10–38 | >90 | 5–12 | 50–70 | 25–37 | 15–20 | 20–30 | 40–60 |
Abbreviations: AED, antiepileptic drug; Cl, clearance; in, intranasal.
aData drawn from Johannessen SI, 200610; Bénit CP, 20127; Patsalos, 201311; De Leon, 201316; Italiano D, 2013.12
bDespite faster clearance, clinical activity of clobazam is 1.5 to 2 times higher due to increased conversion into the active metabolite N-desmethylclobazam.
cUse of clonazepam as oral or buccal (sublingual/sublabial) administration.
dMidazolam as intranasal administration on an as-needed basis.
Table 2.
AED | Metabolism | Active Metabolite | Substrate (CYP) | Inducer (CYP) | Inhibitor (CYP) |
---|---|---|---|---|---|
Carbamazepine | CYP450 | Carbamazepine epoxide | CYP3A4, 1A2, 2C8, 2C9, 2C19 | CYP 1A2, 2A6, 2B6, 2C9, CYP 2C19, 3A4, UGT1A1, 2B7, 2B15, epoxide hydrolase | – |
Clobazam | CYP450 | N-desmethylclobazam | CYP3A4, 2C19 (2B6) | CYP3A4, UGT 1A1 | CYP2C19, 2D6, UGT1A4, 1A6, 2B4 |
Clonazepam | CYP450 | 3A4 | – | – | |
Eslicarbazepine | CYP450, 1/3 by UGT | S-licarbazine | UGT 1A4, UGT 1A9 (2B4, 287, 2B17) | CYP3A4, UGT 1A1, 3A4 | 2C9, 2C19 |
Gabapentin | Not metabolized | – | – | – | |
Lacosamide | CYP 450, 40% via renal excretion | CYP2C19 (2C9, 3A4) | – | – | |
Lamotrigine | 75% via UGT | UGT 1A4 (1A1, 1A9, 2B7) | UGTs | – | |
Levetiracetam | Hydrolysis in blood, 66% via renal excretion | – | – | – | |
Midazolam | CYP450 | 3A4, 2C19, 2B6 | 3A4 | ||
Oxcarbazepine | Glucuronide conjugation, mainly via renal excretion | 10-hydroxycarbazepine | UGTs, arylketone reductase | CYP3A4, UGT1A4 | CYP2C9, 2C19 |
Perampanel | CYP450, UGT | 3A4, 3A5 | 2B6, 3A4/5 | 2C8, 1A9 | |
Phenobarbital | CYP450 | CYP2C9 (2C19, 2E1) | CYP1A2, 2B6, 2C9, 2C19, 2E1, 3A4, UGT1A1 | – | |
Phenytoin | CYP450 | CYP2C9, 2C19 | CYP1A2, 2B6, 2C8, 2C9, 2C19, CYP 3A4, epoxide hydrolase | – | |
Pregabalin | Not metabolized | – | – | – | |
Retigabine | UGTs, N-methylation, 30% via renal excretion, CYP 450 | UGT 1A4 (1A1, 1A3,1A9) | |||
Topiramate | CYP450, UGT, 40% via renal excretion | 3A4, UGT1A4, beta-oxidation | 2C19 | ||
Valproic acid | CYP450, UGT, β-oxidation | UGT 1A3, 2B7 CYP 2A6, 2C9, 2C19, 2B6 |
– | CYP2C9, UGT1A4, UGT2B7, epoxide hydrolase | |
Vigabatrine | 70% via renal excretion | – | 2C9 | 2C19 | |
Zonisamide | CYP450 | CYP3A4 (2C19, 3A5) | 2C19 |
Abbreviation: CYP, cytochrome P450 enzyme.
CYP or UGT co-enzymes within brackets in the Substrate column indicate that these are lesser involved in the metabolism of corresponding AEDs.
Influence of AEDs on Chemotherapeutic Drug Activity
Table 3 provides pharmacokinetic data on the effect of AEDs on the metabolism of chemotherapeutic drugs.21–54 We discuss here the more complicated metabolic changes of chemotherapeutic drugs with AEDs. Lomustine (CCNU) and carmustine (BCNU, applied with Gliadel wafers) are alkylating agents frequently used to treat gliomas, either as single agents, as part of the PCV (procarbazine, CCNU, vincristine) regimen, or together with bevacizumab. Although experimental data indicate enhanced metabolism of these nitrosoureas with phenobarbital, there are no pharmacokinetic data available on humans using concomitant EIAEDs.46,47 Lomustine together with valproic acid may cause hematological toxicity due to independent yet additive effects of both agents on the bone marrow.55,56
Table 3.
Group of CTD | CTD | Interfering AEDs | Mechanism of Metabolism |
No of Pts | MTD of CTD | Change in CTD Activity | Factor of Change in Metabolism | Reference | ||
---|---|---|---|---|---|---|---|---|---|---|
Substrate (CYP) | Inducer (CYP) | Inhibitor (CYP) | ||||||||
Alkylating agents | Busulfan | PHT | Glutathione conjugation |
43 | Cl ↑ T 1/2 ↓ AUC ↓ |
1.21 0.84 0.61 |
Hassan, 199321 Carreras, 201022 | |||
Cyclophosphamide Active metabolite: 4-hydroxycycloph. |
PHT CBZ PHT CBZ PB experimental |
2A6, 2B6, 2C9, 2C18 2C19; 3A4, 3A5 30%–60% by renal elimination |
2C8 | 14 1 14 1 |
Cl ↑ T 1/2 ↓ AUC ↓ AUC ↓ C max ↓ AUC ↑ AUC ↑ |
2.1 0.45 0.41 0.60 0.81 1.58 1.29–1.51 |
Slattery, 199623 De Jonge, 200524 Ekhart 200925 |
|||
Procarbazine | EIAEDs | 49 | nEI 334 mg/m2 EI 393 mg/m2 |
C max ↓ | 0.72 | Grossman, 200660 | ||||
Lomustine | PB | 3A4 | 2C11 | – | Chang, 199446 Levin, 197947 |
|||||
Temozolomide | None | – | – | – | 15 | – | – | Gilbert, 200744 Maschio, 200845 |
||
Thiotepa. Activ. metabolite:Tepa | CBZ, PHT | 2B6, 3A4 | 1 1 |
AUC ↓ AUC ↑ |
0.49–0.57 1.74–2.15 | De Jonge, 200524 Ekhart, 200925 |
||||
Vinca alkaloids (Mitotic spindle inhibitors) | Vincristine (as part of PCV) | PHT, CBZ | 3A4 | 15 | Cl ↑ T 1/2 ↓ AUC ↓ |
1.63 0.65 0.57 |
Bollini, 198326 Villikka, 199927 |
|||
Taxanes (Microtubule agents) | Docetaxel | EIAEDs | 3A4 | Cl ↑ | Chang, 199829 | |||||
Paclitaxel | EIAEDs | 2C8, 3A4 | 2C8 | 25 | nEI 140–240 mg/m2 EI 200–360 mg/m2 |
Cl ↑ Dose↑ Css = at MTD |
2.05 43.7 nM 46.2 nM |
Chang, 199829 Fetell, 199730 |
||
Anti-metabolites | MTX, HD-MTX | EIAEDs | 42 45 |
HD 1–8 gr/m2 | AUC ↓ | 0.58 | Relling, 200031 Ferreri, 200432 |
|||
Levetiracetam | Tubular excretion | 2 | HD 12 gr/m2 | Cl ↓ T 1/2 ↑ |
1.7 1.4 |
Bain, 201433 | ||||
Camptothecin derivatives (Topo-isomerase I inhibitors) | Irinotecan (CPT-11). Active metabolite: SN-38 | EIAEDs Non-EIAEDs VPA |
3A4, 1A1 | 3A4 | 32 9 53 32 46 46 |
nEI: 117–350 mg/m2 a EI: 411–750 mg/m2 nEI: 350–400 mg/m2 b EI: 750 mg/m2 nEI + TMZ: 200 mg/m2 EI + TMZ: 500 mg/m2 c SN-38 |
Cl ↑ T 1/2 = AUC ↓ Cl T 1/2 T 1/2 AUC ↓ Cl ↑ AUC ↓ |
1.99–2.34 = 0.26–0.74 1.82–2.06 = = = 0.58 1.28 0.59 |
Minami 199934 Gilbert, 200335 Prados, 200636 Jaeckle, 201037 Loghin, 200738 De Jong, 200739 |
|
Topotecan | PHT | 1 | Cl ↑ AUC ↓ |
1.45–1.47 0.40 |
Zamboni, 199840 | |||||
Podophyllotoxin derivatives (Topo-isomerase II inhibitors) | Etoposide | EIAEDs | 3A4 | 29 17 |
Cl ↑ AUC ↓ |
1.44–1.77 0.63 |
Rodman, 199441 Mross, 199442 |
|||
Teniposide | 6 42 |
Cl ↑ | 1.93–2.46 | Baker, 199243 Relling, 200031 |
Abbreviations: bid, bis in die; CBZ, carbamazepine; EIAEDs, enzyme-inducing antiepileptic drugs; PB, phenobarbital; PCV: procarbazine, CCNU, vincristine; PHT, phenytoin; VPA, valproic acid; Cl, clearance; T 1/2, Plasma drug elimination half-life; AUC, area under time-concentration curve; MTD, maximum tolerated dose; nEI, MTD without EIAEDs; EI, MTD with EIAEDs and corresponding Cl, T 1/2, or AUC.
aScheme Gilbert, 200335: 90-min i.v. infusion once weekly for 4 out of 6 weeks (as single agent).
bScheme Prados, 200636: 350 mg/m2 i.v. every 3 weeks or 750 mg/m2 with EIAEDs (as single agent).
cScheme Loghin 200738; nEI: 200 mg together with temozolomide 150 mg/m2 days 1–5 q 28 days, and i.v. irinotecan on days 1 & 15 of each cycle, vs EI: 350–550 mg/m2 with similar doses temozolomide/irinotecan.
Cyclophosphamide is applied in malignant lymphoma, leukemia, and in carcinoma of ovary, breast, endometrium, and lung, and often coadministered with thiotepa. When cyclophosphamide is metabolized, it is converted into the active metabolite 4-hydroxycyclophosphamide.23,57 Concurrent therapy with the enzyme inducers carbamazepine or phenytoin yields smaller peak concentrations, increased clearance, and diminished AUC of cyclophosphamide, alongside higher peak concentrations and larger AUC of 4-hydroxycyclophosphamide.23,25 These observations illustrate that in case of coenzyme-dependent conversion of a parent drug into the active metabolite, a concurrently administered enzyme inducer produces enhanced effects of the parent drug despite acceleration of its own metabolism.
Thiotepa is an alkylating agent applied in bladder cancer and malignant lymphoma, and metabolized into its active metabolite tepa. Tepa shows a longer elimination half-life than thiotepa and similar pharmacological properties.24 Concurrent thiotepa and carbamazepine or phenytoin result in accelerated clearance of the primary drug, and organ exposure to tepa is increased by a factor of 2.24,25 The use of vincristine with carbamazepine or phenytoin results in a substantially shorter elimination half-life and smaller AUC.27
Methotrexate, particularly high-dose methotrexate, is an essential part of chemotherapy for leukemia and non-Hodgkin's lymphoma, including CNS lymphoma. In children with acute leukemia, combining methotrexate with EIAEDs was associated with worse survival (HR=2.7) and faster clearance of methotrexate and teniposide.31 Pharmacokinetic studies in primary CNS lymphoma show that methotrexate and concurrent EIAEDs result in half the AUC, possibly depending on altered aldehyde oxidase activity.32 Alternatively, EIAEDs may lead to reduced cellular uptake of methotrexate secondary to diminished intracellular folate carrier activity.58 Based on these observations, one might be inclined to prescribe a noninteracting AED like levetiracetam. However, a potential source of interaction between high-dose methotrexate and levetiracetam is competition for tubular excretion. This leads to a 1.7 lower clearance of methotrexate and patients show signs of hypertension and renal failure.33,59
Camptothecin derivatives are topoisomerase-I inhibitors, including irinotecan (CPT-11), 9-aminocamptothecin (9-AC), and topotecan. Irinotecan is applied in colorectal cancer and malignant glioma, and is transformed by 2C8 and 3A4 into the active metabolite SN-38 (7-ethyl-10-hydroxy camptothecin) via a carboxylesterase. Subsequently, SN-38 is glucuronidated by UGT1A1.38 With concurrent EIAEDs, the clearance of irinotecan rises and its maximum tolerated dose becomes 3.5 times higher.35–37 Combination therapy of valproic acid with irinotecan results unexpectedly in a 41% lower systemic exposure to SN-38, possibly caused by altered protein binding.39 The clearance of 9-AC doubles if combined with EIAEDs.34 Topotecan with phenytoin results in a faster clearance (factor 1.45) and smaller systemic exposure (factor 0.45).40 Also, leucopenia, neutropenia, and thrombopenia occur in 71% with concomitant EIAEDs as opposed to 59% with non-EIAEDs, which is difficult to explain.34 The topoisomerase II inhibitors etoposide and teniposide are mainly applied in lung and ovarian cancer, and are susceptible to the effects of concurrent enzyme inducers. Use of phenobarbital or phenytoin leads to a 3-fold increase of the clearance of tenoposide.41,43
Influence of Chemotherapeutic Drugs on AED Activity
Table 4 provides pharmacokinetic data on the effect of chemotherapeutic drugs on the metabolism of AEDs, leading to either diminished antiseizure activity or increased toxicity of AEDs.48–63 Procarbazine (PCN) is one of the few chemotherapeutic agents that inhibits the 2C and 3A coenzymes, particularly on a prolonged dosing schedule when signs of hepatotoxicity may occur, partly due to autoinhibition of its own metabolism. These effects are only seen with PCN as single agent at doses approaching the maximum tolerated dose (350–400 mg/m2), and not at a conventional single daily dose of 150 mg/m2 or at doses as low as 60 mg/m2 as part of the PCV regimen.60 The inhibiting effects of PCN on the pharmacokinetics of 3A4 substrate drugs possibly explain frequent skin hypersensitivity related to AED plasma levels.61 There are no signs that EIAEDs affect the pharmacokinetics of procarbazine.60
Table 4.
AED | CTD | CTD/dose | Involved Mechanism | No. of Pateints | Change in AED Activity | Factor of Change | Reference |
---|---|---|---|---|---|---|---|
Phenytoin Carbamazepine |
Cisplatin | 20 mg/m2 | Protein displacement/ diminished absorption |
1 | Cl ↑ | 2.29 | Neef, 198848 Grossman, 198949 Dofferhof, 199050 |
Phenytoin | CCNU Cisplatin |
40 mg/m2 40 mg/m2 |
17 | Dose ↑ | 1,39 | ||
Phenytoin | Carboplatin | 400 mg/m2 | 1 | Dose ↑ | 1.21 | ||
Phenytoin | 5-Fluorouracil Capecitabine Doxifluridine |
370–425 mg/m2 800 mg/d |
2C9 inhibition | 2 1 1 |
Dose ↓ Dose ↓ Plasma level ↑ |
0.53–0.72 0.46 4 |
Brickell, 200351 Konishi, 200262 Privitera, 201152 |
Phenytoin | Tamoxifen | 3A, 2C19, 2D6 | 1 | Dose ↑ | 1.16 | Rabinowicz 199553 | |
Valproic acid | Cisplatin | 30 mg/day (with BEP regimen) | Protein displacement/ diminished absorption |
Plasma level ↓ | 0.5 | Neef, 198848 Ikeda, 200528 |
|
Valproic acid | Methotrexate | 1 g/m2 | 1 | Plasma level ↓ | 0.25 | Schroder, 199454 | |
Phenytoine | Vinblastine | 10 mg | Diminished Absorption |
1 | Plasma level ↓ | 0.59 | Bollini, 198326 |
Cisplatin leads to 50% lower plasma levels of phenytoin, carbamazepine and valproic acid, probably based on a combination of lesser intestinal absorption and protein displacement.28,48,49 Impairment of absorption by vinblastine also results in lower plasma levels of these AEDs.26,53
High-dose methotrexate may lower plasma concentrations and induces faster clearance of concomitant phenytoin by both diminished gastrointestinal absorption and folic acid rescue.26 Rapid decline of serum valproate concentrations during high-dose methotrexate treatment can be explained by competition for albumin binding as larger proportions of unbound valproic acid become available for liver breakdown. The routine alkalization during methotrexate infusion increases its renal elimination together with enhanced excretion of valproic acid.54 The pyrimidine antagonists 5-fluorouracil, doxifluridine, and the prodrug capecitabine applied in colorectal cancer are 2C9 inhibitors, leading to 2-fold to 4-fold higher plasma levels of phenytoin and phenobarbital.51,52,62
Interactions With Tyrosine Kinase Inhibitors and Other Targeted Agents
Table 5 presents pharmacokinetic data on drug-drug interactions between anticonvulsants and tyrosine kinase inhibitors or other targeted agents.36,63–91
Table 5.
Tyrosine Kinase Inhibitor | Mechanism (Target) | AED and Dose | Involved CYPs/UGTs |
No of Patients | Dose at MTD | Change in CTD Activity | Factor of Change of Metab | Reference | ||
---|---|---|---|---|---|---|---|---|---|---|
Substrate | Inducer | Inhibitor | ||||||||
Bortezomib | Proteasome inhibitor | EIAEDS | CYP3A (2C19) | 3 | nEI + TMZ 1.3 mg/m2 EI + TMZ ? |
Cl ↑ AUC ↓ |
2.75 0.54 |
Portnow, 201263 | ||
Crizotinib | ALK/MET/ROS1 inhibitor | Midazolam | CYP3A4 (1A9, 2D6, 2C19) | CYP2B6, 2C8, 2C9, UGT | CYP3A4 | AUC ↑ | 3.7 | Mao, 201364 | ||
Dasatinb | SCR, Bcr-Abl, | EIAEDs | CYP3A (2C8, UGT) | CYP3A4, 2C8, 1A1 | AUC ↓ | 0.45 | Reardon, 201265 | |||
Enzastaurin | PI3 K/AKT | EIAEDs | CYP3A (2C19) | 15 | nEI 525 mg | Cl ↑ AUC ↓ |
6.96 0.21 |
Kreisl, 201066 | ||
Erlotinib | EGFR | EIAEDs Midazolam |
CYP3A (1A1, 1A2, 2C8, 2D6) | CYP3A4 | 90 110 |
nEI + TMZ 150 mg/d nEI 200 mg/d EI + TMZ 450 mg/d EI 500 mg/d |
AUC ↓ AUC ↓ |
0.45–0.63 0.76 |
Prados, 200692 Vd Bent, 200967 Shao, 201468 |
|
Gefitinib | EGFR | EIAEDs | CYP3A, 2D6 (1A1, 2C19) | 2C19, 2D6 | 68 30 |
nEI + TMZ 250 mg/d EI + TMZ 1000 mg/d |
Cl ↑ T 1/2 ↓ AUC ↓ |
2.27–3.42 0.84 0.31–0.71 |
Reardon, 200669 Prados, 200870 |
|
Imatinib | Bcr-Abl, c-kit, PDGFR | EIAEDs Levetiracetam Valproic acid Lamotrigine Phenytoin Carbamazepine Oxcarbazepine Topiramate |
CYP3A (2C9, 2D6) | 3A4, 2C8 | 3A4/5, 2C9, 2D6 |
33 | nEI 1200 mg/d EI 1200 mg/d nEI + TMZ 1000 mg/d EI + TMZ 1000 mg/d |
Cl ↑ T 1/2 ↓ AUC ↓ Cl ↑ T 1/2 ↓ AUC ↓ Through Plasma levels |
3.42–4.13 0.30–0.60 0.26–0.79 1.30 0.39 0.76 0.97 1.02 1.01 0.31 0.29 0.45 0.58 |
Reardon, 200871 Wen, 200672 Pursche, 200873 |
Lapatinib | EGFR, HER2 | CBZ Midazolam |
CYP3A (1A2, 2C8, 2C9, 2C19, 2D6) | 3A4, 2C8 | 16 24 |
EI 1000 mg | Cl ↑ T 1/2 ↓ AUC ↓ AUC ↑ |
8.83 0.28 0.27–0.28 1.4 |
Smith, 200975 Thiessen, 201074 Shao, 201468 |
|
Pazopanib | c-kit, FGFR, PDGFR, VEGFR1–3 | EIAEDs Midazolam |
CYP3A (1A2, 2C8) | 3A4 | CYP3A4, 2D6 2B6, 2C9, 2C19 |
75 | AUC ↓ AUC ↑ |
∼0.5 1.35 |
Reardon, 201376 Shao, 201468 |
|
Evorolimus | mTOR | EIAEDs | CYP3A (2C19) | 32 | AUC ↓ | 0.48 | Mason, 201277 | |||
Sirolimus | mTOR | EIAEDs | CYP3A (2C19) | 23 25 36 |
T 1/2 ↓ AUC ↓ |
0.46–0.75 0.54–0.62 |
Boni, 200778 Kuhn, 200779 Galanis, 201180 |
|||
Temsirolimus | mTOR | EIAEDs Valproic acid |
CYP3A (2C19) | 25 36 8 |
nEI 170 mg/d EI 250 mg/d |
Cl ↑ T 1/2 = AUC ↓ C max |
1.20–1.47 0.66–0.85 ∼ 2 |
Boni, 200778 Kuhn, 200779 Galanis, 200580 Coulter, 201381 |
||
Sorafenib | c-kit, PDGFR, RAF, VEGFR1-3 |
EIAEDs Midazolam |
CYP3A, UGT1A9 | CYP3A4, 2B6, 2C8, 2C9, 2D6, UGT1A1, 1A9 | 32 | T1/2 ↓ AUC ↓ AUC ↓ |
0.39–0.56 0.36–0.49 0.85 |
Reardon, 201182 Flaherty, 201183 |
||
Sunitinib | PDGFR, VEGFR, c-KIT | EIAEDs | CYP3A (2C19) | Widmer, 201486 Bilbao, 201584 |
||||||
Tamoxifen | Estrogene receptor | Phenytoin | CYP3A (2C19) | 1 | Dose ↓ | 0.46 | Gryn, 201485 | |||
Tipifarnib | Ras, Farnesyl-TF inhibitor | EIAEDs | CYP3A (2C19) | nEI 300 mg bid EI 600 mg bid |
Cl ↑ T 1/2 = AUC ↓ |
2.90 = 0.52 |
Cloughesy, 200587 | |||
Vandetanib | EGFR, RET, VEGFR2 | EIAEDS | CYP3A | CYP2D6 | 79 | Cl = T 1/2 = AUC = |
Kreisl, 201288 | |||
Vatalanib | C-kit, PDFGR VEGFR1-3 |
EIAEDs | CYP3A (2C19) | 10 | nEI 1000 mg bid EI 1000 mg bid |
Cl ↑ T 1/2 = AUC ↓ |
2.68–3.09 0.42–0.82 |
Reardon, 200989 Gerstner, 201190 |
||
Vemurafenib | V600E BRAF kinase | CYP3A4 | CYP3A4 | CYP1A2, 2D6 | Da Rocha Dias, 201391 |
Abbreviations: bid, bis in die; CBZ, carbamazepine; EIAEDs, enzyme-inducing anti-epileptic drugs; PB, phenobarbital; PCV: procarbazine, CCNU, vincristine; PHT, phenytoin; VPA, valproic acid; Cl, clearance; T 1/2, plasma drug elimination half-life; AUC, area under time-concentration curve; MTD, maximum tolerated dose; nEI, MTD without EIAEDs; EI, MTD with EIAEDs and corresponding Cl, T 1/2, or AUC.
A number of tyrosine kinase inhibitors have been tested in phase 1 and 2 trials for gliomas, and many of these trials also examined pharmacokinetics with concurrent EIAEDs and non-EIAEDs. When combined with 3A4-inducing AEDs, tyrosine kinase inhibitors usually have a 2-fold higher clearance rate and corresponding reduction of AUC. Crizotinib, dasatinib, imatinib, and lapatinib particularly show substantially faster metabolism with concurrent EIAEDs.64,65,72–78,92 For imatinib and lapatinib, organ exposure is about 4 times lower without dose adjustment, representing a moderate drug interaction. Drug interactions are defined as strong if they produce a larger than 5-fold change in metabolism, moderate as 2-fold to 5-fold, and mild if between 1.25-fold and 2-fold.68 Some tyrosine kinase inhibitors are 3A4 inhibitors with inherent risks of toxicity when combined with other 3A4 substrate drugs. Examples of a higher organ exposure of combined drugs are that of crizotinib with midazolam by a factor 3.7, of imatinib with simvastatin by a factor 3.5, and sorafenib with docetaxel by a factor 1.5 to 1.8.64,68 One may expect to see similar changes in metabolism with other 3A4 substrate drugs including both older and newer generation AEDs. Until now, however, hardly any data on the effect of TKIs on AED metabolism are available. For pharmacokinetic characteristics of individual tyrosine kinase inhibitors, we refer to other reviews.68,93–95
Temsirolimus (CCI-779) is an ester of sirolimus (rapamycin) inhibiting the mTOR protein, which regulates key molecules of the PI3K and AKT pathway. mTOR blockers are applied in renal cell carcinoma, tuberous sclerosis, and subependymal giant cell astrocytomas, and has been tested as phase II drug in glioblastoma.78–80 Combined use with EIAEDS produces a diminished systemic exposure to temsirolimus by a factor of 0.66 to 0.85, to everolimus by a factor of 0.48, and to sirolimus by a factor of 0.54 to 0.62.77,79 Valproic acid reduces the maximum tolerated dose of temsirolimus to 35 mg/m2 in adults and to 150 mg/m2 in children, possibly due to inhibition of CYP3A4.81 A dose as low as 25 mg/m2 is sufficient to reduce mTOR activity.81
Interactions With Glucocorticoids
Glucocorticoids are frequently applied in cancer for multiple reasons, and in neuro-oncology mainly to control tumor-induced brain edema. Glucocorticoids can influence the activity of a number of CYP coenzymes including 3A4, 3A5, and to a smaller extent 2C8, 2C9, and C19 by activation of the nuclear GC receptor. GCs control transcription of a wide spectrum of genes, including 3A5 and 2C9. Corticosteroids are mainly 3A4 enzyme inducers. In this way, they influence the pharmacokinetics of concurrent drugs, although clinical studies on interactions between steroids and AEDs are relatively scarce. (Table 6)96–101 A clinically relevant dose of 16 mg/day of dexamethasone increases 3A4 activity by 25%, but there is substantial individual variability ranging from no increase to a 49% to 70% increase in one-third of patients.100 This explains observations of faster clearance and subtherapeutic levels of phenytoin with concurrent dexamethasone.99 Increasing phenytoin dosing by a factor of 1.5 to 2.0 is necessary to maintain therapeutic plasma levels. After upward dose adjustment of phenytoin with concurrent steroids, and once arriving at the stage of steroid tapering, phenytoin concentrations can easily rise to toxic levels if it is not also tapered.101 However, increased phenytoin levels occur occasionally in combination with dexamethasone, which has been explained by competition for enzyme-binding. These observations underscore the possibility of unexpected drug-drug interactions.20,102
Table 6.
AED | Steroid | No. of Patients | Change in Steroid Activity | Factor of Change | Reference |
---|---|---|---|---|---|
Carbamazepine | Prednisolone | 6 | Cl ↑ T 1/2 ↓ |
1.41 0.64 |
Bartoszek, 198796 |
Phenobarbital | 6 | Cl ↑ T 1/2 ↓ |
1.79 0.44 |
Bartoszek, 198796 | |
Phenytoin | 2 | Cl ↑ T 1/2 ↓ |
1.77 0.71 |
Bartoszek, 198796 | |
Carbamazepine | Methylprednisolone | 5 | Cl ↑ T 1/2 ↓ |
3.09 0.46 |
Bartoszek, 198796 |
Phenobarbital | 5 | Cl ↑ T 1/2 ↓ |
4.42 0.46 |
Bartoszek, 198796 | |
Phenytoin | 2 | Cl ↑ T 1/2 ↓ |
5.79 0.29 |
Bartoszek, 198796 | |
Phenytoin | Dexamethasone | 15 | Cl ↑ T 1/2 ↓ |
2.93 0.54 |
Chalk, 198497 |
Phenytoin | 6 | Plasma Conc ↓ | 0.5 | Wong, 198598 |
Abbreviations: bid, bis in die; CBZ, carbamazepine; EIAEDs, enzyme-inducing anti-epileptic drugs; PB, phenobarbital; PCV: procarbazine, CCNU, vincristine; PHT, phenytoin; VPA, valproic acid; Cl, clearance; T ½, plasma drug elimination half-life; AUC, area under time-concentration curve; MTD, maximum tolerated dose; nEI, MTD without EIAEDs; EI, MTD with EIAEDs and corresponding Cl, T ½, or AUC.
Concurrent carbamazepine, phenytoin, and phenobarbital lead to faster metabolism of methylprednisolone, prednisolone, and dexamethasone.96,97 The inducing effects of phenytoin on the clearance of dexamethasone vary between a factor of 3 up to 12 with a correspondingly low AUC of 0.13.96,103 Phenytoin together with prednisone or prednisolone results in a faster clearance of steroids by a factor of 1.5. Overall, the plasma half-life of steroids shortens to about half its original value with concurrent EIAEDs. The biological half-life as being more decisive for the duration of clinical activity of glucocorticoids is probably not or much less affected. During cancer treatment, corticosteroids are usually prescribed at supratherapeutic doses, which may explain why signs of insufficient steroid dosing often remain undetected.
Discussion
Epilepsy occurs frequently in cancer, particularly with gliomas, meningiomas, and brain metastasis.1–3 In general, patients undergo intensive therapy, including surgery, radiation therapy, and one or more lines of chemotherapy. In parallel, patients will receive antiepileptic drugs and almost all need steroids at some stage of their disease. Not surprisingly, this setting carries a high risk for drug-drug interactions.4 As antineoplastic drugs often have a narrow therapeutic window close to the maximum tolerated dose, these interactions can easily result in insufficient antitumor therapy or in drug toxicity. This may have a major clinical impact, as illustrated by observations of shorter survival in children with acute lymphoblastic leukemia receiving concurrent EIAEDs.31 Temozolomide and bevacizumab are probably the most frequently applied antitumor agents in neuro-oncology, although neither are subject to known drug-drug interactions.44,45 However, hematological toxicity associated with chemotherapeutics, including temozolomide, may be aggravated by direct toxic effects of valproic acid on the bone marrow.55,104 In cases of combined temozolomide and valproic acid, multifactorial analysis indicates that the former is decisive for developing thrombopenia.105,106 Recently, valproic acid is under study as an anti-tumor agent based on ability to block the histone deacetylase enzyme. Clinical application of valproic acid as a histone deacetylase I and II blocker, particularly combined with DNA-targeting chemotherapy, has shown promising results in glioblastoma and other cancers.14,104,107
The large majority of chemotherapies and tyrosine kinase inhibitors are susceptible to or may induce drug-drug interactions. In order to apply proper drug regimens, phase 1/2 trials testing new agents against glioma often include separate arms with and without EIAEDs, providing data on corresponding maximum tolerated dose, clearance, half-life, and AUC (see Tables 3–5). Despite this body of information, we hardly know to what extent standardized dose adjustment is applied in the daily practice of cancer treatment. Likewise, in cases of tumor progression, it may be unclear if the progression is caused by insufficient drug delivery or is a consequence of drug-drug interactions. One may argue that since we now have a large number of second and third generation AEDs with only a mild tendency to or no drug interactions, the issue of drug-drug interactions has become outdated.9 A number of circumstances, however, make this less likely. First, it is unclear to what extent the classic AEDs are being prescribed in daily neuro-oncological practice as compared to second or third generation AEDs. Studies on physician compliance show that EIAEDs are routinely given to patients with brain tumors as seizure prophylaxis, despite current guidelines advising against this practice.108–110 In a recent survey of 28 Australian cancer centers, the enzyme-inducer phenytoin was given as first-line anticonvulsant, followed by levetiracetam and carbamazepine.111 Still, in many underdeveloped nations the first generation anticonvulsants are the preferred choice, and we have no good insight to what extent in the developed world financial hurdles restrain the prescribing of newer AEDs. Even if one prescribes an AED that does not kindle drug-drug interactions, it is important to realize that many recently approved AEDs are themselves susceptible to the inducing or inhibiting effects of concurrent chemotherapeutic drugs or tyrosine kinase inhibitors, implying risks of insufficient seizure control or central nervous system toxicity.68,112 In addition, some of the most recently approved AEDs are not exempt from drug interactions, particularly eslicarbazepine and perampanel.11
For clinical practice, it is important to realize that in case of coenzyme-dependent conversion of a parent drug into its active metabolite, a concurrently given enzyme inducer not only causes accelerated metabolism of the parent drug, but also enhanced formation of the active metabolite. In this way, the net effect will be enhanced drug activity. Examples are combined use of an EIAED with cyclophosphamide, ifosfamide, or thiotepa, and a number of tyrosine kinase inhibitors, including erlotinib, imatinib, gefitinib, lapatinib, and sorafenib.113
As many tyrosine kinase inhibitors are 3A4 inhibitors, particularly crizotinib and imatinib, one may expect toxicity when combined with 3A4-substrate drugs including some AEDs and chemotherapeutic agents. Although trials of new tyrosine kinase inhibitors in gliomas often include an analysis of the pharmacokinetic effects of concurrent AED use, little is known about how tyrosine kinase inhibitors may affect the metabolism of AEDs. The size of these interactions may well be comparable to the effects on other 3A4 substrate drugs.68 As tyrosine kinase inhibitors often cause autoinhibition of their own metabolism, the effects of enzyme inhibition on concurrent therapy are often limited. A big obstacle for obtaining factual data on the influence of tyrosine kinase inhibitors on a concurrent therapy is that once an agent has been approved and introduced to the market, it is difficult to find the means for additional drug trials. An exception might be studies on new drug indications for other cancer types than those already approved. It would be a great advantage if such additional investigations include pharmacokinetics on common drug associations in systemic cancer and neuro-oncology, particularly how tyrosine kinase inhibitors affect the metabolism of concurrent therapies such as AEDs and glucocorticoids.
A separate problem is the large variability among individuals with respect to the metabolism of drugs. The activity of CYP enzymes shows high individual variability, including their susceptibility to the effects of drug inducers or inhibitors. The efficacy of PXR and GC receptor as transcription factors involved in regulating these enzymes is also variable, contributing to differences in enzyme activity among individuals.5,103 CYP activity is also dependent on age, sex, and ethnicity, as well as dietary and organ factors like hepatic dysfunction.12,114
The observations on variability in drug metabolism underscore the need of therapeutic drug monitoring by measuring drug concentrations in plasma to detect drug-drug interactions.11,12,94,95,114 In this way, underdosing or overdosing of AEDs can be recognized, allowing drug adjustment in case of insufficient effectiveness or toxicity.10,115 To what extent therapeutic drug monitoring is applied in daily neuro-oncologypractice is uncertain and harmful interactions may take place at a larger scale than we assume.4 Unfortunately, there are few studies on the value of drug monitoring for the proper management of epilepsy, although low therapeutic AED levels carry a higher risk of seizures.116 A recent guideline of the American Academy of Neurology and International League of Epilepsy (ILEA) on combined AED and retroviral therapy is fully based on therapeutic-drug-monitoring data.117 A position paper of the ILAE has defined when to apply therapeutic drug monitoring in the daily practice of seizure management.115 The ILAE recommends performing plasma drug measurements once a desired clinical response has been achieved based on the variable therapeutic range of an AED, the persistence of seizures, and factors as age, comorbidity or concomitant therapy. Similar calls for therapeutic drug monitoring of chemotherapeutics and tyrosine kinase inhibitors have been made in the field of systemic cancer treatment.86,94,95,115,118 Given the prevalence of multidrug regimens for patients with seizures and cancer, routine monitoring of plasma levels of AEDs and anticancer agents is probably indispensable.
Electronic databases can be consulted to provide easy access and information on drug interactions with AEDs.119–121 However, these may contain large discrepancies, particularly for newer generation AEDs.122 In this review, these shortcomings have been avoided by reporting on all studies examining interactions between AEDs and chemotherapeutic drugs, tyrosine kinase inhibitors, or glucocorticoids and expressing these in quantitative rather than qualitative terms.
In conclusion, the risks of drug-drug interactions causing ineffective cancer treatment, organ dysfunction, or neurotoxicity illustrate the importance of effective and well-tolerated AEDs that do not interfere with cancer treatment. The strongest effects of the EIAEDs carbamazepine, phenytoin, and phenobarbital are seen with cyclophosphamide, camptothecin derivatives, taxanes, and topoisomerase II inhibitors, showing about a 2-fold to 3-fold higher clearance and a doubling of maximum tolerated dose. The inhibiting activity of valproic acid is mainly limited to temsirolimus, and it may aggravate thrombopenia caused by chemotherapeutic drugs via a direct effect on the bone marrow. Cisplatin and high-dose methotrexate lead to lower plasma levels of phenytoin, valproic acid, tiagabine, and clobazam or other benzodiazepines by competition for protein binding. The enzyme-inhibiting effect of 5-fluorouracil causes a 2-fold to 4-fold higher organ exposure to phenytoin and phenobarbital. Many tyrosine kinase inhibitors are 3A4 inhibitors, particularly imatinib and crizotinib, requiring lower dosing of concurrent therapy.
As more than 50% of patients with gliomas need AED polytherapy, the risk of drug-drug interaction is not easily avoidable. If possible, the EIAEDs carbamazepine, phenytoin, and phenobarbital should be avoided. Fortunately, there are a number of effective and well-tolerated AEDs that cause little to no drug interactions; these include levetiracetam, lamotrigine, lacosamide, and zonisamide.9 Although many of the tyrosine kinase inhibitors are 3A4 inhibitors, it not known how strongly they affect concurrent therapy. The routine of treating of low-grade and high-grade gliomas or brain metastasis with multidrug regimens consisting of AEDs, chemotherapeutics, tyrosine kinase inhibitors, and glucocorticoids, combined with the individual variability in drug metabolism, underlines the importance of plasma drug monitoring. Similar calls for therapeutic drug monitoring of chemotherapeutic drugs and tyrosine kinase inhibitors have been made for systemic cancer. Future studies on the pharmacokinetics of AEDs with concurrent antitumor agents, particularly tyrosine kinase inhibitors and glucocorticoids, will hopefully provide more insight into the size of these interactions, allowing the proper dosing of combined therapies.
Websites to be consulted on Drug Interactions:
Conflict of interest statement. Charles Vecht received a speaker's fee from UCB.
References
- 1. van Breemen MSM, Wilms EB, Vecht CJ. Epilepsy in patients with brain tumours: epidemiology, mechanisms, and management. Lancet Neurol. 2007;6(5):421–430. [DOI] [PubMed] [Google Scholar]
- 2. Pallud J, Audureau E, Blonski M et al. . Epileptic seizures in diffuse low-grade gliomas in adults. Brain. 2014;137:449–462. [DOI] [PubMed] [Google Scholar]
- 3. Soffietti R, Cornu P, Delattre JY et al. . EFNS Guidelines on diagnosis and treatment of brain metastases: report of an EFNS Task Force. Eur J Neurol. 2006;13(7):674–681. [DOI] [PubMed] [Google Scholar]
- 4. Riechelmann RP, Tannock IF, Wang L, Saad ED, Taback NA, Krzyzanowska MK. Potential drug interactions and duplicate prescriptions among cancer patients. J Natl Cancer Inst. 2007;99(8):592–600. [DOI] [PubMed] [Google Scholar]
- 5. Zanger UM, Klein K, Thomas M et al. . Genetics, epigenetics, and regulation of drug-metabolizing cytochrome P450 enzymes. Clin Pharmacol Ther. 2014;95(3):258–261. [DOI] [PubMed] [Google Scholar]
- 6. Brodie MJ, Mintzer S, Pack AM, Gidal BE, Vecht CJ, Schmidt D. Enzyme induction with antiepileptic drugs: Cause for concern? Epilepsia. 2013;54(1):11–27. [DOI] [PubMed] [Google Scholar]
- 7. Benit CP, Vecht CJ. Spectrum of side-effects of anticonvulsants in patients with brain tumours. Eur Assoc Neuro-Oncol Mag. 2012;2(1):15–24. [Google Scholar]
- 8. Glauser T, Ben-Menachem E, Bourgeois B et al. . Updated ILAE evidence review of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes. Epilepsia. 2013;54(3):551–563. [DOI] [PubMed] [Google Scholar]
- 9. Vecht CJ, Kerkhof M, Duran-Pena A. Seizure prognosis in brain tumors: new insights and evidence-based management. Oncologist. 2014;19(7):751–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Johannessen SI, Tomson T. Pharmacokinetic variability of newer antiepileptic drugs - When is monitoring needed? Clin Pharmacokinet. 2006;45(11):1061–1075. [DOI] [PubMed] [Google Scholar]
- 11. Patsalos PN. Drug Interactions with the Newer Antiepileptic Drugs (AEDs)-Part 1: Pharmacokinetic and Pharmacodynamic Interactions Between AEDs. Clin Pharmacokinet. 2013;52(11):927–966. [DOI] [PubMed] [Google Scholar]
- 12. Italiano D, Perucca E. Clinical Pharmacokinetics of New-Generation Antiepileptic Drugs at the Extremes of Age: An Update. Clin Pharmacokinet. 2013;52(8):627–645. [DOI] [PubMed] [Google Scholar]
- 13. van Breemen MSM, Rijsman RM, Taphoorn MJB, Walchenbach R, Zwinkels H, Vecht CJ. Efficacy of anti-epileptic drugs in patients with gliomas and seizures. J Neurol. 2009;256(9):1519–1526. [DOI] [PubMed] [Google Scholar]
- 14. Kerkhof M, Dielemans JCM, van Breemen MS et al. . Effect of valproic acid on seizure control and on survival in patients with glioblastoma multiforme. Neuro Oncol. 2013;15(7):961–967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Johannessen Landmark C, Johannessen SI, Tomson T. Host factors affecting antiepileptic drug delivery-pharmacokinetic variability. Adv Drug Deliv Rev. 2012;64(10):896–910. [DOI] [PubMed] [Google Scholar]
- 16. de Leon J, Spina E, Diaz FJ. Clobazam Therapeutic Drug Monitoring: A Comprehensive Review of the Literature With Proposals to Improve Future Studies. Ther Drug Monit. 2013;35(1):30–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Gidal BE, Sheth R, Parnell J, Maloney K, Sale M. Evaluation of VPA dose and concentration effects on lamotrigine pharmacokinetics: implications for conversion to lamotrigine monotherapy. Epilepsy Res. 2003;57(2–3):85–93. [DOI] [PubMed] [Google Scholar]
- 18. Patsalos PN, Perucca E. Clinically important drug interactions in epilepsy: general features and interactions between antiepileptic drugs. Lancet Neurol. 2003;2(6):347–356. [DOI] [PubMed] [Google Scholar]
- 19. Patsalos PN. Drug interactions with the newer antiepileptic drugs (AEDs)-Part 2: Pharmacokinetic and pharmacodynamic interactions between AEDs and drugs used to treat non-epilepsy disorders. Clin Pharmacokinet. 2013;52(12):1045–1061. [DOI] [PubMed] [Google Scholar]
- 20. Vecht CJ, Wagner GL, Wilms EB. Interactions between antiepileptic and chemotherapeutic drugs. Lancet Neurol. 2003;2(7):404–409. [DOI] [PubMed] [Google Scholar]
- 21. Hassan M, Oberg G, Bjorkholm M, Wallin I, Lindgren M. Influence of prophylactic anticonvulsant therapy on high-dose busulfan kinetics. Cancer Chemother Pharmacol. 1993;33(3):181–186. [DOI] [PubMed] [Google Scholar]
- 22. Carreras E, Cahn JY, Puozzo C et al. . Influence on Busilvex (R) pharmacokinetics of clonazepam compared to previous phenytoin historical data. Anticancer Res. 2010;30(7):2977–2984. [PubMed] [Google Scholar]
- 23. Slattery JT, Kalhorn TF, McDonald GB et al. . Conditioning regimen-dependent disposition of cyclophosphamide and hydroxycyclophosphamide in human marrow transplantation patients. J Clin Oncol. 1996;14(5):1484–1494. [DOI] [PubMed] [Google Scholar]
- 24. de Jonge ME, Huitema ADR, van Dam SM, Beijnen JH, Rodenhuis S. Significant induction of cyclophosphamide and thiotepa metabolism by phenytoin. Cancer Chemother Pharmacol. 2005;55(5):507–510. [DOI] [PubMed] [Google Scholar]
- 25. Ekhart C, Rodenhuis S, Beijnen JH, Huitema AD. Carbamazepine induces bioactivation of cyclophosphamide and thiotepa. Cancer Chemother Pharmacol. 2009;63(3):543–547. [DOI] [PubMed] [Google Scholar]
- 26. Bollini P, Riva R, Albani F et al. . Decreased phenytoin level during antineoplastic therapy: a case report. Epilepsia. 1983;24(1):75–78. [DOI] [PubMed] [Google Scholar]
- 27. Villikka K, Kivisto KT, Maenpaa H, Joensuu H, Neuvonen PJ. Cytochrome P450-inducing antiepileptics increase the clearance of vincristine in patients with brain tumors. Clin Pharmacol Ther. 1999;66(6):589–593. [DOI] [PubMed] [Google Scholar]
- 28. Ikeda H, Murakami T, Takano M, Usui T, Kihira K. Pharmacokinetic interaction on valproic acid and recurrence of epileptic seizures during chemotherapy in an epileptic patient. Br J Clin Pharmacol. 2005;59(5):593–597. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Chang SM, Kuhn JG, Rizzo J et al. . Phase I study of paclitaxel in patients with recurrent malignant glioma: A North American brain tumor consortium report. J Clin Oncol. 1998;16(6):2188–2194. [DOI] [PubMed] [Google Scholar]
- 30. Fetell MR, Grossman SA, Fisher JD et al. . Preirradiation paclitaxel in glioblastoma multiforme: Efficacy, pharmacology, and drug interactions. J Clin Oncol. 1997;15(9):3121–3128. [DOI] [PubMed] [Google Scholar]
- 31. Relling MV, Pui CH, Sandlund JT et al. . Adverse effect of anticonvulsants on efficacy of chemotherapy for acute lymphoblastic leukaemia. Lancet. 2000;356(9226):285–290. [DOI] [PubMed] [Google Scholar]
- 32. Ferreri AJM, Guerra E, Regazzi M et al. . Area under the curve of methotrexate and creatinine clearance are outcome-determining factors in primary CNS lymphomas. Br J Cancer. 2004;90(2):353–358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Bain E, Birhiray RE, Reeves DJ. Drug-drug interaction between methotrexate and levetiracetam resulting in delayed methotrexate elimination. Ann Pharmacother. 2014;48(2):292–296. [DOI] [PubMed] [Google Scholar]
- 34. Minami H, Lad TE, Nicholas MK, Vokes EE, Ratain MJ. Pharmacokinetics and pharmacodynamics of 9-aminocamptothecin infused over 72 hours in phase II studies. Clin Cancer Res. 1999;5(6):1325–1330. [PubMed] [Google Scholar]
- 35. Gilbert MR, Supko JG, Batchelor T et al. . Phase I clinical and pharmacokinetic study of irinotecan in adults with recurrent malignant glioma. Clin Cancer Res. 2003;9(8):2940–2949. [PubMed] [Google Scholar]
- 36. Prados MD, Lamborn K, Yung WKA et al. . A phase 2 trial of irinotecan (CPT-11) in patients with recurrent malignant glioma: A North American Brain Tumor Consortium study. Neuro Oncol. 2006;8(2):189–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Jaeckle KA, Ballman KV, Giannini C et al. . Phase II NCCTG trial of RT plus irinotecan and adjuvant BCNU plus irinotecan for newly diagnosed GBM. J Neurooncol. 2010;99(1):73–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Loghin ME, Prados MD, Wen P et al. . Phase I study of temozolomide and irinotecan for recurrent malignant gliomas in patients receiving enzyme-inducing antiepileptic drugs: a north american brain tumor consortium study. Clin Cancer Res. 2007;13(23):7133–7138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. de Jong FA, van der Bol JM, Mathijssen RH et al. . Irinotecan chemotherapy during valproic acid treatment: pharmacokinetic interaction and hepatotoxicity. Cancer Biol Ther. 2007;6(9):1368–1374. [DOI] [PubMed] [Google Scholar]
- 40. Zamboni WC, Gajjar AJ, Heideman RL et al. . Phenytoin alters the disposition of topotecan and N-desmethyl topotecan in a patient with medulloblastoma. Clin Cancer Res. 1998;4(3):783–789. [PubMed] [Google Scholar]
- 41. Rodman JH, Murry DJ, Madden T, Santana VM. Altered etoposide pharmacokinetics and time to engraftment in pediatric-patients undergoing autologous bone-marrow transplantation. J Clin Oncol. 1994;12(11):2390–2397. [DOI] [PubMed] [Google Scholar]
- 42. Mross K, Bewermeier P, Kruger W, Stockschlader M, Zander A, Hossfeld DK. Pharmacokinetics of undiluted or diluted high-dose etoposide with or without busulfan administered to patients with hematologic malignancies. J Clin Oncol. 1994;12(7):1468–1474. [DOI] [PubMed] [Google Scholar]
- 43. Baker DK, Relling MV, Pui CH, Christensen ML, Evans WE, Rodman JH. Increased teniposide clearance with concomitant anticonvulsant therapy. J Clin Oncol. 1992;10(2):311–315. [DOI] [PubMed] [Google Scholar]
- 44. Gilbert MR, Armstrong TS. Management of patients with newly diagnosed malignant primary brain tumors with a focus on the evolving role of temozolomide. Ther Clin Risk Manag. 2007;3(6):1027–1033. [PMC free article] [PubMed] [Google Scholar]
- 45. Maschio M, Albani F, Jandolo B et al. . Temozolomide treatment does not affect topiramate and oxcarbazepine plasma concentrations in chronically treated patients with brain tumor-related epilepsy. J Neurooncol. 2008;90(2):217–221. [DOI] [PubMed] [Google Scholar]
- 46. Chang TKH, Chen HY, Waxman DJ. 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU) modulates rat-liver microsomal cyclophosphamide and ifosphamide activation by suppressing cytochrome-P450 2C11 messenger-RNA levels. Drug Metab Dispos. 1994;22(5):673–679. [PubMed] [Google Scholar]
- 47. Levin VA, Stearns J, Byrd A, Finn A, Weinkam RJ. Effect of phenobarbital pretreatment on the anti-tumor activity of 1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU), 1-(2-chloroethyl)-3-cyclohexyl-1-nitrosourea (CCNU) and 1-(2-chloroethyl)-3-(2,6-dioxo-3-piperidyl-1-nitrosourea (PCNU), and on the plasma pharmacokinetics and biotransformation of BCNU. J Pharmacol Exp Ther. 1979;208(1):1–6. [PubMed] [Google Scholar]
- 48. Neef C, de Voogd-van der Straaten I. An interaction between cytostatic and anticonvulsant drugs. Clin Pharmacol Ther. 1988;43(4):372–375. [DOI] [PubMed] [Google Scholar]
- 49. Grossman SA, Sheidler VR, Gilbert MR. Decreased phenytoin levels in patients receiving chemotherapy. Am J Med. 1989;87(5):505–510. [DOI] [PubMed] [Google Scholar]
- 50. Dofferhoff ASM, Berendsen HH, Vandernaalt J, Haaxmareiche H, Smit EF, Postmus PE. Decreased phenytoin level after carboplatin treatment. Am J Med. 1990;89(2):247–248. [DOI] [PubMed] [Google Scholar]
- 51. Brickell K, Porter D, Thompson P. Phenytoin toxicity due to fluoropyrimidines (5FU/capecitabine): three case reports. Br J Cancer. 2003;89(4):615–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Privitera M, de Los Ríos la Rosa F. Capecitabine-phenytoin interaction is dose dependent with an unexpected time course. Anticancer Drugs. 2011;22(10):1027–1029. [DOI] [PubMed] [Google Scholar]
- 53. Rabinowicz AL, Hinton DR, Dyck P, Couldwell WT. High-dose tamoxifen in treatment of brain tumors: interaction with antiepileptic drugs. Epilepsia. 1995;36(5):513–515. [DOI] [PubMed] [Google Scholar]
- 54. Schroder H, Ostergaard JR. Interference of high-dose methotrexate in the metabolism of valproate. Pediatr Hematol Oncol. 1994;11(4):445–449. [DOI] [PubMed] [Google Scholar]
- 55. Bourg V, Lebrun C, Chichmanian RM, Thomas P, Frenay M. Nitroso-urea-cisplatin-based chemotherapy associated with valproate: Increase of haematologic toxicity. Ann Oncol. 2001;12(2):217–219. [DOI] [PubMed] [Google Scholar]
- 56. Verrotti A, Scaparrotta A, Grosso S, Chiarelli F, Coppola G. Anticonvulsant drugs and hematological disease. Neurol Sci. 2014;35(7):983–993. [DOI] [PubMed] [Google Scholar]
- 57. de Jonge ME, Huitema ADR, Rodenhuis S, Beijnen JH. Clinical pharmacokinetics of cyclophosphamide. Clin Pharmacokinet. 2005;44(11):1135–1164. [DOI] [PubMed] [Google Scholar]
- 58. Halwachs S, Lakoma C, Schaefer I, Seibel P, Honscha W. The antiepileptic drugs phenobarbital and carbamazepine reduce transport of methotrexate in rat choroid plexus by down-regulation of the reduced folate carrier. Mol Pharmacol. 2011;80(4):621–629. [DOI] [PubMed] [Google Scholar]
- 59. Parentelli AS, Phulpin-Weibel A, Mansuy L, Contet A, Trechot P, Chastagner P. Drug-drug interaction between methotrexate and levetiracetam in a child treated for acute lymphoblastic leukemia. Pediatr Blood Cancer. 2013;60(2):340–341. [DOI] [PubMed] [Google Scholar]
- 60. Grossman SA, Carson KA, Batchelor TT et al. . The effect of enzyme-inducing antiseizure drugs on the pharmacokinetics and tolerability of procarbazine hydrochloride. Clin Cancer Res. 2006;12(17):5174–5181. [DOI] [PubMed] [Google Scholar]
- 61. Lehmann DF, Hurteau TE, Newman N, Coyle TE. Anticonvulsant usage is associated with an increased risk of procarbazine hypersensitivity reactions in patients with brain tumors. Clin Pharmacol Ther. 1997;62(2):225–229. [DOI] [PubMed] [Google Scholar]
- 62. Konishi H, Morita K, Minouchi T, Nakajima M, Matsuda M, Yamaji A. Probable metabolic interaction of doxifluridine with phenytoin. Ann Pharmacother. 2002;36(5):831–834. [DOI] [PubMed] [Google Scholar]
- 63. Portnow J, Frankel P, Koehler S et al. . A phase I study of bortezomib and temozolomide in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2012;69(2):505–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Mao J, Johnson TR, Shen Z, Yamazaki S. Prediction of Crizotinib-Midazolam interaction using the simcyp population-based simulator: Comparison of CYP3A time-dependent inhibition between human liver microsomes versus hepatocytes. Drug Metab Dispos. 2013;41(2):343–352. [DOI] [PubMed] [Google Scholar]
- 65. Reardon DA, Vredenburgh JJ, Desjardins A et al. . Phase 1 trial of dasatinib plus erlotinib in adults with recurrent malignant glioma. J Neurooncol. 2012;108(3):499–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Kreisl TN, Kotliarova S, Butman JA et al. . A phase I/II trial of enzastaurin in patients with recurrent high-grade gliomas. Neuro Oncol. 2010;12(2):181–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. van den Bent MJ, Brandes AA, Rampling R et al. . Randomized Phase II Trial of Erlotinib versus Temozolomide or Carmustine in Recurrent Glioblastoma: EORTC Brain Tumor Group Study 26034. J Clin Oncol. 2009;27(8):1268–1274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Shao J, Markowitz JS, Bei D, An G. Enzyme-and transporter-mediated drug interactions with small molecule tyrosine kinase inhibitors. J Pharm Sci. 2014;103(12):3810–3833. [DOI] [PubMed] [Google Scholar]
- 69. Reardon DA, Quinn JA, Vredenburgh JJ et al. . Phase 1 trial of gefitinib plus sirolimus in adults with recurrent malignant glioma. Clin Cancer Res. 2006;12(3):860–868. [DOI] [PubMed] [Google Scholar]
- 70. Prados MD, Yung WKA, Wen PY et al. . Phase-1 trial of gefitinib and temozolomide in patients with malignant glioma: a North American brain tumor consortium study. Cancer Chemother Pharmacol. 2008;61(6):1059–1067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Reardon DA, Desjardins A, Vredenburgh JJ et al. . Safety and pharmacokinetics of dose-intensive imatinib mesylate plus temozolomide: Phase 1 trial in adults with malignant glioma. Neuro Oncol. 2008;10(3):330–340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Wen PY, Yung WKA, Lamborn KR et al. . Phase I/II study of imatinib mesylate for recurrent malignant gliomas: North American Brain Tumor Consortium Study 99–08. Clin Cancer Res. 2006;12(16):4899–4907. [DOI] [PubMed] [Google Scholar]
- 73. Pursche S, Schleyer E, von Bonin M et al. . Influence of enzyme-inducing antiepileptic drugs on trough level of imatinib in glioblastoma patients. Curr Clin Pharmacol. 2008;3(3):198–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Thiessen B, Stewart C, Tsao M et al. . A phase I/II trial of GW572016 (lapatinib) in recurrent glioblastoma multiforme: clinical outcomes, pharmacokinetics and molecular correlation. Cancer Chemother Pharmacol. 2010;65(2):353–361. [DOI] [PubMed] [Google Scholar]
- 75. Smith DA, Koch KM, Arya N, Bowen CJ, Herendeen JM, Beelen A. Effects of ketoconazole and carbamazepine on lapatinib pharmacokinetics in healthy subjects. Br J Clin Pharmacol. 2009;67(4):421–426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Reardon DA, Groves MD, Wen PY et al. . A Phase I/II Trial of Pazopanib in Combination with Lapatinib in Adult Patients with Relapsed Malignant Glioma. Clin Cancer Res. 2013;19(4):900–908. [DOI] [PubMed] [Google Scholar]
- 77. Mason WP, MacNeil M, Kavan P et al. . A phase I study of temozolomide and everolimus (RAD001) in patients with newly diagnosed and progressive glioblastoma either receiving or not receiving enzyme-inducing anticonvulsants: an NCIC CTG study. Invest New Drugs. 2012;30(6):2344–2351. [DOI] [PubMed] [Google Scholar]
- 78. Boni J, Leister C, Burns J, Cincotta M, Hug B. Pharmacokinetic profile of temsirolimus with concomitant administration of cytochrome p450-inducing medications. J Clin Pharmacol. 2007;47(11):1430–1439. [DOI] [PubMed] [Google Scholar]
- 79. Kuhn JG, Chang SM, Wen PY et al. . Pharmacokinetic and tumor distribution characteristics of temsirolimus in patients with recurrent malignant glioma. Clin Cancer Res. 2007;13(24):7401–7406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Galanis E, Buckner JC, Maurer MJ et al. . Phase II trial of temsirolimus (CCI-779) in recurrent glioblastoma multiforme: A north central cancer treatment group study. J Clin Oncol. 2005;23(23):5294–5304. [DOI] [PubMed] [Google Scholar]
- 81. Coulter DW, Walko C, Patel J et al. . Valproic acid reduces the tolerability of temsirolimus in children and adolescents with solid tumors. Anticancer Drugs. 2013;24(4):415–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Reardon DA, Vredenburgh JJ, Desjardins A et al. . Effect of CYP3A-inducing anti-epileptics on sorafenib exposure: results of a phase II study of sorafenib plus daily temozolomide in adults with recurrent glioblastoma. J Neurooncol. 2011;101(1):57–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Flaherty KT, Lathia C, Frye RF et al. . Interaction of sorafenib and cytochrome P450 isoenzymes in patients with advanced melanoma: a phase I/II pharmacokinetic interaction study. Cancer Chemother Pharmacol. 2011;68(5):1111–1118. [DOI] [PubMed] [Google Scholar]
- 84. Bilbao-Meseguer I, Jose BS, Lopez-Gimenez LR et al. . Drug interactions with sunitinib. J Oncol Pharm Pract. 2015;21(1):52–66. [DOI] [PubMed] [Google Scholar]
- 85. Gryn SE, Teft WA, Kim RB. Profound reduction in the tamoxifen active metabolite endoxifen in a patient on phenytoin for epilepsy compared with a CYP2D6 genotype matched cohort. Pharmacogenet Genomics. 2014;24(7):367–369. [DOI] [PubMed] [Google Scholar]
- 86. Widmer N, Bardin C, Chatelut E et al. . Review of therapeutic drug monitoring of anticancer drugs part two - Targeted therapies. Eur J Cancer. 2014;50(12):2020–2036. [DOI] [PubMed] [Google Scholar]
- 87. Cloughesy TF, Kuhn J, Robins HI et al. . Phase I trial of tipifarnib in patients with recurrent malignant glioma taking enzyme-inducing antiepileptic drugs: A North American brain tumor consortium study. J Clin Oncol. 2005;23(27):6647–6656. [DOI] [PubMed] [Google Scholar]
- 88. Kreisl TN, McNeill KA, Sul J, Iwamoto FM, Shih J, Fine HA. A phase I/II trial of vandetanib for patients with recurrent malignant glioma. Neuro Oncol. 2012;14(12):1519–1526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Reardon DA, Egorin MJ, Desjardins A et al. . Phase I pharmacokinetic study of the vascular endothelial growth factor receptor tyrosine kinase inhibitor Vatalanib (PTK787) plus imatinib and Hydroxyurea for malignant glioma. Cancer. 2009;115(10):2188–2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Gerstner ER, Eichler AF, Plotkin SR et al. . Phase I trial with biomarker studies of vatalanib (PTK787) in patients with newly diagnosed glioblastoma treated with enzyme inducing anti-epileptic drugs and standard radiation and temozolomide. J Neurooncol. 2011;103(2):325–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. da Rocha Dias S, Salmonson T, van Zwieten-Boot B et al. . The European Medicines Agency review of vemurafenib (Zelboraf) for the treatment of adult patients with BRAF V600 mutation-positive unresectable or metastatic melanoma: summary of the scientific assessment of the Committee for Medicinal Products for Human Use. Eur J Cancer. 2013;49(7):1654–1661. [DOI] [PubMed] [Google Scholar]
- 92. Prados MD, Lamborn KR, Chang S et al. . Phase 1 study of erlotinib HCl alone and combined with temozolomide in patients with stable or recurrent malignant glioma. Neuro Oncol. 2006;8(1):67–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Klumpen H-J, Samer CF, Mathijssen RHJ, Schellens JHM, Gurney H. Moving towards dose individualization of tyrosine kinase inhibitors. Cancer Treat Rev. 2011;37(4):251–260. [DOI] [PubMed] [Google Scholar]
- 94. Yu H, Steeghs N, Nijenhuis CM, Schellens JHM, Beijnen JH, Huitema ADR. Practical guidelines for therapeutic drug monitoring of anticancer tyrosine kinase inhibitors: Focus on the pharmacokinetic targets. Clin Pharmacokinet. 2014;53(4):305–325. [DOI] [PubMed] [Google Scholar]
- 95. de Wit D, Guchelaar H-J, den Hartigh J, Gelderblom H, van Erp NP. Individualized dosing of tyrosine kinase inhibitors: are we there yet? Drug Discov Today. 2015;20(1):18–36. [DOI] [PubMed] [Google Scholar]
- 96. Bartoszek M, Szefler SJ. Corticosteroid-therapy in adolescent patients. J Adolesc Health. 1987;8(1):84–91. [DOI] [PubMed] [Google Scholar]
- 97. Chalk JB, Ridgeway K, Brophy TRO, Yelland JDN, Eadie MJ. Phenytoin impairs the bioavailability of dexamethasone in neurological and neurosurgical patients. J Neurol Neurosurg Psychiatry. 1984;47(10):1087–1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Wong DD, Longenecker RG, Liepman M, Baker S, Lavergne M. Phenytoin-dexamethasone - a possible drug-drug interaction. JAMA. 1985;254(15):2062–2063. [DOI] [PubMed] [Google Scholar]
- 99. Gattis WA, May DB. Possible interaction involving phenytoin, dexamethasone, and antineoplastic agents: a case report and review. Ann Pharmacother. 1996;30(5):520–526. [DOI] [PubMed] [Google Scholar]
- 100. McCune JS, Hawke RL, LeCluyse EL et al. . In vivo and in vitro induction of human cytochrome P4503A4 by dexamethasone. Clin Pharmacol Ther. 2000;68(4):356–366. [DOI] [PubMed] [Google Scholar]
- 101. Lackner TE. Interaction of dexamethasone with phenytoin. Pharmacotherapy. 1991;11(4):344–347. [PubMed] [Google Scholar]
- 102. Lawson GJ, Chakraborty J, Dumasia MC, Baylis EM. Methylprednisolone hemisuccinate and metabolites in urine from patients receiving high-dose corticosteroid-therapy. Ther Drug Monit. 1992;14(1):20–26. [DOI] [PubMed] [Google Scholar]
- 103. Matoulkova P, Pavek P, Maly J, Vlcek J. Cytochrome P450 enzyme regulation by glucocorticoids and consequences in terms of drug interaction. Expert Opin Drug Metab Toxicol. 2014;10(3):425–435. [DOI] [PubMed] [Google Scholar]
- 104. Weller M, Gorlia T, Cairncross JG et al. . Prolonged survival with valproic acid use in the EORTC/NCIC temozolomide trial for glioblastoma. Neurology. 2011;77(12):1156–1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Simo M, Velasco R, Graus F et al. . Impact of antiepileptic drugs on thrombocytopenia in glioblastoma patients treated with standard chemoradiotherapy. J Neurooncol. 2012;108(3):451–458. [DOI] [PubMed] [Google Scholar]
- 106. Tinchon A, Oberndorfer S, Marosi C et al. . Haematological toxicity of Valproic acid compared to Levetiracetam in patients with glioblastoma multiforme undergoing concomitant radio-chemotherapy: a retrospective cohort study. J Neurol. 2015;262(1):179–186. [DOI] [PubMed] [Google Scholar]
- 107. Krauze AV, Myrehaug SD, Chang MG et al. . A phase II study of concurrent radiation therapy, temozolomide and the histone deacetylase inhibitor valproic acid for patients with glioblastoma. Int J Radiat Oncol Biol Phys. 2015;92(5):986–992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. de Oliveira JA, Santana IA, Caires IQS et al. . Antiepileptic drug prophylaxis in primary brain tumor patients: is current practice in agreement to the consensus? J Neurooncol. 2014;120(2):399–403. [DOI] [PubMed] [Google Scholar]
- 109. Glantz MJ, Forsyth PA, Recht LD et al. . Practice parameter: Anticonvulsant prophylaxis in patients with newly diagnosed brain tumors - Report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology. 2000;54(10):1886–1893. [DOI] [PubMed] [Google Scholar]
- 110. Soffietti R, Baumert BG, Bello L et al. . Guidelines on management of low-grade gliomas: report of an EFNS-EANO* Task Force. Eur J Neurol. 2010;17(9):1124–1133. [DOI] [PubMed] [Google Scholar]
- 111. Chen JY, Hovey E, Rosenthal M, Livingstone A, Simes J. Neuro-oncology practices in Australia: A Cooperative Group for Neuro-Oncology patterns of care study. Asia Pac J Clin Oncol. 2014;10(2):162–167. [DOI] [PubMed] [Google Scholar]
- 112. Teo YL, Ho HK, Chan A. Metabolism-related pharmacokinetic drug-drug interactions with tyrosine kinase inhibitors: current understanding, challenges and recommendations. Br J Clin Pharmacol. 2015;79(2):241–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. van Erp NP, Gelderblom H, Guchelaar H-J. Clinical pharmacokinetics of tyrosine kinase inhibitors. Cancer Treat Rev. 2009;35(8):692–706. [DOI] [PubMed] [Google Scholar]
- 114. Johannessen Landmark C, Baftiu A, Tysse I et al. . Pharmacokinetic variability of four newer antiepileptic drugs, lamotrigine, levetiracetam, oxcarbazepine, and topiramate: a comparison of the impact of age and comedication. Ther Drug Monit. 2012;34(4):440–445. [DOI] [PubMed] [Google Scholar]
- 115. Patsalos PN, Berry DJ, Bourgeois BFD et al. . Antiepileptic drugs - best practice guidelines for therapeutic drug monitoring: A position paper by the subcommission on therapeutic drug monitoring, ILAE Commission on Therapeutic Strategies. Epilepsia. 2008;49(7):1239–1276. [DOI] [PubMed] [Google Scholar]
- 116. Handoko KB, Rijkom JEFZ-v, Visee HF, Hermens WAJJ, Hekster YA, Egberts TCG. Drug treatment-related factors of inadequate seizure control. Epilepsy Behav. 2008;13(3):545–548. [DOI] [PubMed] [Google Scholar]
- 117. Birbeck GL, French JA, Perucca E et al. . Evidence-based guideline: Antiepileptic drug selection for people with HIV/AIDS Report of the Quality Standards Subcommittee of the American Academy of Neurology and the Ad Hoc Task Force of the Commission on Therapeutic Strategies of the International League Against Epilepsy. Neurology. 2012;78(2):139–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Paci A, Veal G, Bardin C et al. . Review of therapeutic drug monitoring of anticancer drugs part 1-Cytotoxics. Eur J Cancer. 2014;50(12):2010–2019. [DOI] [PubMed] [Google Scholar]
- 119. Truven Health Analytics Inc. http://micromedex.com/mobile. Micromedex Drug Interactions. Greenwood Village, CO 80111, USA; 2015.
- 120. Wolterskluwerhealth.com. http://webstore.lexi.com/Store/PDA-Software-for-Pharmacists/Lexi-Drugs-Interact. New York, NY 10001, USA; 2015.
- 121. Drug Interactions. Elsevier's Gold Standard. http://www.goldstandard.com/product/drug-reference-patient-education/clinical-pharmacology/. Oxford, OX5 1GB, UK; 2015.
- 122. Ekstein D, Tirosh M, Eyal Y, Eyal S. Drug interactions involving antiepileptic drugs: Assessment of the consistency among three drug compendia and FDA-approved labels. Epilepsy Behav. 2015;44:218–224. [DOI] [PubMed] [Google Scholar]