Skip to main content
Erschienen in: BMC Cancer 1/2018

Open Access 01.12.2018 | Research article

High-dose thiotepa-related neurotoxicity and the role of tramadol in children

verfasst von: Christophe Maritaz, Francois Lemare, Agnes Laplanche, Sylvie Demirdjian, Dominique Valteau-Couanet, Christelle Dufour

Erschienen in: BMC Cancer | Ausgabe 1/2018

Abstract

Background

Serious neurological adverse events (NAE) have occurred during treatment with high-dose thiotepa regimens of children with high-risk solid tumours. The objective was to assess the incidence of NAE related to high-dose thiotepa and to identify potential contributing factors that could exacerbate the occurrence of this neurotoxicity.

Methods

From May 1987 to March 2011, children with solid tumours treated with high-dose thiotepa were retrospectively identified. Each NAE detected led to an independent case analysis. Potential contributing factors were pre-specified and univariate/multivariable analyses were performed.

Results

Three hundred seven courses of thiotepa (251 patients) were identified. The total dose per treatment ranged from 600 to 900 mg/m2. 81 NAE (26%) were identified. 46 NAE were related to high-dose thiotepa during the first course (18.3%) and 11 during the second course (19.6%). The symptoms appeared in a median time of 2 days after the introduction of thiotepa. Central and peripheral symptoms were headaches, tremors, confusion, seizures, cerebellar syndrome, and coma. High-dose thiotepa was reintroduced in 18 cases and symptoms reappeared in 5 children. For 3 patients who had seizures during the first course, premedication with clonazepam for the second course has prevented recurrence of NAE. As contributing factors, brain tumour and tramadol treatment increased the risk of thiotepa-related neurotoxicity by 2 to 6 times respectively.

Conclusions

The incidence of neurotoxicity was 18.3%. Brain tumours and tramadol treatment are risk factors to consider when using high-dose thiotepa. The outcome of patients was favourable without sequelae in all cases and rechallenge with thiotepa was possible.
Abkürzungen
ASCT
Autologous stem cell transplantation
CI
Confidence interval
CSF
Cerebrospinal fluid
GFR
Glomerular filtration rate
GST
Glutathione S-transferase
HDCT
High-dose chemotherapy
IQR
Interquartile range
MTD
Maximum tolerated dose
NAE
Neurological adverse events
OR
Odds Ratio

What is already known about this subject

  • Little is known about the possible relationship between exposure to high-dose thiotepa and the occurrence of neurological disorders.
  • High-dose thiotepa administered below the Maximum Tolerated Dose has never been studied in terms of neurological outcomes.

What this study adds

  • This study estimates the incidence of thiotepa-related neurological complications, unlike the neurotoxicity described when the drug was used beyond the defined limiting dose or combined with another cytotoxic drug or irradiation.
  • Our findings show that brain tumour and tramadol treatment could be independently associated with thiotepa-related neurotoxicity.

Background

High-dose chemotherapy (HDCT) with autologous stem cell transplantation (ASCT) has improved the survival of children with high-risk solid tumours. The rationale for HDCT is that escalated doses of HDCT may increase survival by capturing putative remnant malignant cells [1]. The rationale for ASCT following HDCT is a planned rescue for HDCT-related severe haematological toxicity [2].
The HDCT regimens are generally based on the use of alkylating agents [3]. Among them, thiotepa (N,N′,N″-triethylenethiophosphoramide), an organophosphorus compound with the formula SP(NC2H4)3, is one of the few drugs to have demonstrated clear activity at high doses in childhood tumours [49]. Thiotepa is extensively metabolized by cytochrome P450. Its major metabolite tepa, has similar alkylating activity [10, 11]. This hepatic biotransformation is mediated by CYP3A4 and CYP2B6, and conjugation is catalyzed by glutathione S-transferase (GST) [12, 13]. Following intravenous administration, drug exposure in cerebrospinal fluid (CSF) is almost equivalent to that of the product in plasma [14]. Thiotepa has been reported to possess an elimination half-life of 1.3–5.2 h and tepa an elimination half-life of 3–21 h [15]. Thiotepa and its metabolites are totally excreted in urine [15, 16]. Due to its metabolic profile, concomitant administration of thiotepa with CYP3A4 or CYP2B6 inhibitors may increase plasma thiotepa concentrations and potentially decrease the concentrations of the active metabolite tepa. Conversely, concomitant administration of thiotepa with CYP3A4 or CYP2B6 inducers may reduce plasma thiotepa concentrations and increase those of tepa. In addition, there is interindividual variability in product excretion since CYP3A4, CYP2B6, and GST share a genetic polymorphism with some variants which exert a significant impact on the clearance of thiotepa and tepa [17]. Relationships between the pharmacokinetics of the drug and its toxicity have been described [1821]. Toxicity to the gastrointestinal and central nervous system is dose-limiting and the maximum tolerated dose (MTD) of thiotepa is between 1005 and 1125 mg/m2 [7, 21, 22].
Little is known about the possible relationship between exposure to high-dose thiotepa and the occurrence of neurological disorders. To our knowledge, high-dose thiotepa administered below the MTD has never been studied in terms of neurological outcomes. One reason for the lack of knowledge is that thiotepa is rarely used alone. In our experience, serious neurological adverse events (NAE) have occurred during treatment with high-dose thiotepa regimens. Some of these NAE could lead to life-threatening complications. The objective of the present study was to assess the incidence of NAE related to high-dose thiotepa with ASCT in children with solid tumours and to identify potential contributing factors that could increase the occurrence of this neurotoxicity.

Methods

Patients

The institutional review board of the Gustave Roussy Cancer Campus approved this study. The parents/guardians gave their written informed consent for the retrospective analysis of clinical data according to the institutional review board of the Gustave Roussy Cancer Campus. From the institutional pediatric transplantation register, we established a retrospective cohort of all patients treated for a solid tumour with high-dose thiotepa followed by ASCT between May 1987 and March 2011 at Gustave-Roussy, center with expertise in the treatment of complex malignancies. Our analysis focused on neurotoxicity exclusively due to thiotepa, therefore patients treated with thiotepa combined with another cytotoxic drug or radiotherapy were excluded.

Procedures

All medical and nursing records were reviewed. Demographics data (sex, age, weight, height), race aimed at assessing the influence of the type of metabolism (slow, medium, fast), clinical data (medical and neurological history, allergies, cancer type and site of the primary tumour), and drug exposure (thiotepa and drugs administered concomitantly) from admission to hospital until the day of ASCT were collected for each patient. To assess the impact of potential organic disturbances, biological monitoring of renal and hepatic function from admission to hospital until the day of ASCT was taken into account. Biological monitoring of renal function took into account blood urea and creatinine. The glomerular filtration rate (GFR) was estimated using the Schwartz formula [23]. Monitoring of liver function took into account transaminases (SGOT, SGPT), gamma-GT, bilirubin, and serum protein. Any neurological symptom occurring during the administration of thiotepa or within seven days (5 half-lives) following the end of the last administration of thiotepa was defined as a NAE. Further examinations distinguished symptoms related to progressive disease or to other causes from NAE. NAE were classified according to the National Cancer Institute Common Terminology Criteria for Adverse Events (version 4.03). Each event detected led to a case analysis to assess the neurological outcome.
Causality assessment of adverse drug reactions (imputability) was performed using the algorithm devised by Begaud et al. which is based on a three-stage process: assessment of chronological criteria, clinical and biological findings and symptom evaluation [24]. The method separates intrinsic imputability (possible cause between drug and clinical event) from extrinsic imputability (bibliographical data) using seven criteria divided in two groups: chronology and semiology (symptoms or signs).
Potential contributing factors were pre-specified and were: (1) sex; (2) age; (3) race; (4) primary tumour site; (5) history of neurological disorders; (6) metabolic disruption; and (7) use of concomitant therapy (antiemetic, antipsychotic, analgesic, antifungals, Proton Pump Inhibitor, and histamine H2-receptor antagonist). Concomitant drugs were determined according to their potential ability to interact with thiotepa metabolism. They were captured the week before, during, and the week after thiotepa exposure.

Statistical analysis

Some patients had received more than one course of high-dose thiotepa. We decided to retain only one course per patient in order to avoid skewing the statistical analysis by including patients with individual susceptibility. In principle, the first course of high-dose chemotherapy was taken into account in the analysis. The second courses were analyzed separately to study the effect of rechallenge with thiotepa when NAE occurred during the first course. The results are expressed as percentages (qualitative data), or medians and the interquartile range (quantitative data). Concerning the analysis of factors associated with the occurrence of NAE, univariate and multivariable analyses were performed using logistic regression. Variables with a P value below 0.05 in the univariate analysis were selected for the multivariable analysis. The results are expressed as the Odds Ratio (OR) of NAE for each factor; an OR equal to 1 is associated with the reference category of each factor. All statistical analyses were two-sided, with P values of 0.05 or less deemed statistically significant. The software used was SAS, version 9.1.

Results

Between May 1987 and March 2011, 307 courses of high-dose thiotepa with ASCT were administered to 251 patients (56 patients received 2 courses of high-dose thiotepa). Baseline characteristics of these 251 children are shown in Table 1. The median age of children was 8 years (IQR 5–15) with a sex ratio of 1.4 boys per 1 girl, and the population was predominantly Caucasian (74%). The total dose of thiotepa per course was 900 mg/m2 for 46 patients (18%), 720 mg/m2 for 76 patients (30%), and 600 mg/m2 for 129 patients (51%). One hundred and sixteen patients (46%) had a primary brain tumour. Twenty children (8%) had a neurological disorder due to disease when they were admitted to hospital: moderate cerebellar syndrome (n = 9), central neuropathy (n = 1), peripheral neuropathy (n = 3), hemiparesis (n = 3), and facial paralysis (n = 4). Thirty children exhibited early-stage renal failure upon admission to hospital with a median GFR of 80.5 mL/min (IQR, 71–85 mL/min) and a median blood urea level of 4.65 mmol/L (IQR, 3–6.3 mmol/L). The 221 patients without renal insufficiency upon admission to hospital had a median GFR of 123.5 mL/min (IQR, 111–146 mL/min) and a median blood urea level of 3.9 mmol/L (IQR, 2.4–4.6 mL/min). On the day of ASCT, early-stage renal failure had persisted with a median GFR of 79 mL/min (IQR, 68–82 mL/min) and a median blood urea level of 2.6 mmol/L (IQR, 1.7–4.5 mL/min) in 12 of these 30 children. No patients had liver disruption at upon admission to hospital (SGPT median = 26 IU/L, IQR 18–38; SGOT median = 28 IU/L, IQR 22–36; total bilirubin median = 8 μmol/L, IQR 6–10; GGT median = 16 IU/L, IQR 12–24). Thirteen patients had reversible hepatic cytolysis at the end of the thiotepa regimen without cholestasis (SGPT median = 217 IU/L, IQR 177–253; SGOT median = 156 IU/L, IQR 143–179; total bilirubin median = 12 μmol/L, IQR 10–14; GGT median = 17 IU/L, IQR 8–25; serum protein median = 59 g/L, IQR 53–74). During treatment with thiotepa, 92 patients had required analgesics (42%). According to the World Health Organization analgesic ladder, 76 patients (35%) had received simple analgesics such as acetaminophen or nefopam for mild pain, 53 patients (24%) had received weak opioids such as tramadol, codeine, or dextropropoxyphen for mild to moderate pain, and 9 patients (4%) had received strong opioids such as morphine or pethidine for moderate to severe pain. Sometimes, patients had received a combination of different analgesics. Regarding anti-emetic support, every child had received at least a 5-HT3 antagonist. The anti-emetic treatment added was alizapride (43%), or aprepitant (3%). None of the patients had declared an allergy to analgesics or anti-emetic drugs upon admission to hospital.
Table 1
Patient characteristics
 
Patients (N = 251)
Gender
 Boys
145 (58%)
 Girls
106 (43%)
Age (years; median [IQR])
8 [5–15]
Race
 
 Caucasian
186 (74%)
 African
40 (16%)
 Others
25 (10%)
Brain tumour
116 (46%)
 Medulloblastoma
71 (28%)
 PNETa
26 (10%)
 Pinealoblastoma
13 (5%)
 ATRTb
5 (2%)
 Malignant GCTc
1 (0.4%)
Other tumour site
135 (54%)
 Osteosarcoma
50 (20%)
 Neuroblastoma
26 (10%)
 Ewing tumor
23 (9%)
 Rhabdomyosarcoma
20 (8%)
 Other sarcoma
4 (2%)
 Nephroblastoma
4 (2%)
 Burkitt lymphoma
4 (2%)
 Hepatoblastoma
2 (1%)
 Desmoplastic tumour
2 (1%)
Neurological disorder at baselined
20 (8%)
History of seizure
13 (5%)
Thiotepa dose
 600 mg/m2
129 (51%)
 720 mg/m2
76 (30%)
 900 mg/m2
46 (18%)
Concomitant drugse
 Alizapride
94 (43%)
 Antibiotic
49 (22%)
 Antidepressant
7 (3%)
 Antipsychotic
40 (18%)
 Aprepitant
7 (3%)
 Benzodiazepine
43 (20%)
 Histamine H2-receptor antagonist
5 (2%)
 Proton Pump Inhibitor
7 (3%)
Analgesics
92 (42%)
 Simple analgesics
76 (35%)
 i.e. acetaminophen, nefopam
 
 Weak opioids
53 (24%)
 i.e. tramadol, codeine, dextropropoxyphen
 
 Strong opioids
9 (4%)
 i.e. morphine, pethidine
 
Data are numbers (%), unless otherwise stated
aPrimitive NeuroEctodermal Tumour
bAtypical Teratoid Rhabdoid Tumour
cMalignant Germ Cell Tumour
dData missing for one patient
eData missing for 31 patients
After reviewing medical and nursing records, 81 NAE (26.4%) were unveiled during the 307 courses, 69 NAE had occurred during the 251 first courses (27.5%), and 12 NAE during the 56 s courses (21.4%). After assessment of intrinsic and extrinsic imputability, NAE were considered “very likely” related to thiotepa for 13 patients (16.0%), “likely” related to thiotepa for 35 patients (43.2%) and “possibly” related to thiotepa for 9 patients (11.1%). Twenty-four NAE (29.6%) were assessed as “dubious” or “unlikely” to be related to thiotepa, and were therefore excluded (Fig. 1). Thus, among the 307 courses of high-dose thiotepa, 57 NAE had occurred (18.6%). Forty-six NAE had occurred during the 251 first courses (18.3%), and 11 during the 56 s courses (19.6%). In this study, the incidence of high-dose thiotepa-related neurotoxicity was therefore estimated at 18.3%. Five children had experienced NAE during the two courses of high-dose thiotepa. Twenty-eight NAE (24%) had occurred in patients with a brain tumour and 18 (13%) in patients with another primary tumour site. None of the 13 patients with hepatic cytolysis had developed NAE.
The median time to the onset of neurological symptoms was 2 days (range 0–4) after the beginning of the course of thiotepa. Table 2 shows the symptoms described for the 46 NAE identified during the first courses of high-dose thiotepa. Some patients had experienced several symptoms. Most of the NAE symptoms were graded 1 or 2, corresponding to a mild or moderate disorder. Headache, dizziness, and confusion, graded 1 or 2, which had occurred in 45 cases (94%), are the neurological symptoms described in the Summary of Product Characteristics of thiotepa. Six events were graded 3: three cases of headache, dizziness, confusion; one case of tremor; one of seizure; and one of cerebellar syndrome. One case of seizure was graded 4 and resulted in life-threatening consequences. All of these events had disappeared without sequelae in a median interval of 3 days (range 1–8 days). Eighteen patients had been rechallenged with thiotepa, and NAE had reappeared in 5 cases. In 3 patients who had experienced seizure during the first course of thiotepa, prophylactic treatment with clonazepam had been administered and no further NAE had occurred. Fig. 1 presents a summary of all NAE-related data observed.
Table 2
Description of neurological adverse events (NAE)
 
Patients (N = 251)
Symptoms
n
NCI CTCAE grade
1
2
3
4
5
Headache, dizziness, confusion
48 (15.6%)
23 (47.9%)
22 (45.8%)
3 (6.3%)
  
Blurred vision
3 (1.0%)
3 (100%)
    
Tremor
8 (2.6%)
3 (37.5%)
4 (50%)
1 (12.5%)
  
Neuralgia
2 (0.7%)
1 (50%)
1 (50%)
   
Seizure
6 (2.0%)
2 (33.3%)
2 (33.3%)
1 (16.7%)
1 (16.7%)
 
Pyramidal tract syndrome
1 (0.3%)
 
1 (100%)
   
Cerebellar syndrome
3 (1.0%)
 
2 (66.7%)
1 (33.3%)
  
Opsoclonus-myoclonus syndrome
1 (0.3%)
 
1 (100%)
   
Coma
1 (0.3%)
 
1 (100%)
   
Table 3 shows details of the univariate analysis performed on the 46 NAE which had occurred during the first courses. A brain tumour (OR, 2.1; 95% CI, 1.1 to 4.0; P = 0.03), a neurological disorder at baseline (OR, 3.4; 95% CI, 1.3 to 8.8; P = 0.02), alizapride (OR, 2.0; 95% CI, 1.0 to 4·0; P = 0.04), and weak opioids (OR, 6.1; 95% CI, 3.0 to 12.4; P < 0.0001) were identified as risk factors for thiotepa-related NAE. The weak opioids used were tramadol (n = 46), codeine (n = 5), and dextropropoxyphen (n = 2). NAE had occurred in 21 cases when tramadol had been used (46%; P < 0.0001), in two cases with codeine (40%; P = 0.26), and in one case with dextropropoxyphen (50%; P = 0.29). The risk of NAE was not significantly associated with gender (P = 0.41), age (P = 0.48), race (P = 0.51), a history of seizure (P = 0.27), renal failure (P = 0.98), or other concomitant drugs. Although not significantly associated with the dose of thiotepa (P = 0.18), we observed a trend towards an increasing dose-dependent rate of neurotoxicity (600 mg/m2: 14%; 720 mg/m2: 22%; 900 mg/m2: 24%).
Table 3
Univariate and multivariate logistic regression analysis of factors contributing to NAE
 
n
NAE (%)
univariate Pa
univariate OR (95% CI)
multivariate Pb
multivariate OR (95% CI)
Gender
 Boys
145
24 (17%)
    
 Girls
106
22 (21%)
0.41
1.3 (0.7–2.5)
  
Age
 No NAE
205
9.3 (6.3)f
    
 Yes NAE
46
8.6 (5.2)f
0.48c
   
Race
 Caucasian
186
31 (17%)
    
 African
40
9 (23%)
0.51e
   
 Others
25
6 (24%)
    
Brain tumour
 No
135
18 (13%)
 
1
 
1
 Yes
116
28 (24%)
0.03
2.1 (1.1–4.0)
0.04
2.2 (1.0–4.6)
Neurological disorder at baseline
 No
230
38 (17%)
 
1
 
1
 Yes
20
8 (40%)
0.02
3.4 (1.3–8.8)
0.08
2.7 (0.9–8.4)
History of seizure
 No
238
42 (18%)
 
1
  
 Yes
13
4 (31%)
0.27
2.1 (0.6–7.1)
  
Thiotepa regimen
 600 mg/m2
129
18 (14%)
    
 720 mg/m2
76
17 (22%)
0.18e
   
 900 mg/m2
46
11 (24%)
    
Alizaprided
 No
126
19 (15%)
 
1
 
1
 Yes
94
25 (27%)
0.04
2.0 (1.0–4.0)
0.13
1.7 (0.8–3.6)
Aprepitantd
 No
213
41 (19%)
 
1
  
 Yes
7
3 (43%)
0.14
3.1 (0.7–14.6)
  
Antipsychoticd
 No
180
34 (19%)
 
1
  
 Yes
40
10 (25%)
0.39
1.4 (0.6–3.2)
  
Proton Pump Inhibitord
 No
213
43 (20%)
 
1
  
 Yes
7
1 (14%)
1
0.7 (0.1–5.6)
  
Histamine H2-receptor antagonistd
 No
215
42 (20%)
 
1
  
 Yes
5
2 (40%)
0.26
2.7 (0.4–17)
  
Simple analgesicsd
      
i.e. acetaminophen, nefopam
      
 No
144
20 (13%)
 
1
  
 Yes
76
14 (18%)
0.40
1.4 (0.7–3.0)
  
Weak opioidsd
      
i.e. tramadol, codeine, dextropropoxyphen
      
 No
167
20 (12%)
 
1
 
1
 Yes
53
24 (45%)
0.0001
6.1 (3.0–12.4)
0.0001
6.3 (3.0–13.4)
Strong opioidsd
      
i.e. morphine, pethidine
      
 No
211
44 (21%)
 
1
  
 Yes
9
4 (44%)
0.09
3.0 (0.8–11.8)
  
aFisher exact
bTaking into account 4 factors: brain tumour (yes/no); neurological disorder at baseline (yes/no); alizapride intake (yes/no); weak opioids (yes/no)
cStudent t test
dData missing for 31 patients
eChi-square test (2 df)
fMean (SD)
In the multivariable analysis, a brain tumour and a weak opioid (tramadol) were identified as independent risk factors for thiotepa-related NAE (Table 3). Patients with a brain tumour were found to have a greater risk of developing NAE than patients with another primary tumour (OR, 2.2; 95% CI, 1.0 to 4.6; P = 0.04). The use of tramadol for analgesia was confirmed as a highly significant contributing factor for NAE (OR, 6.3; 95% CI, 3.0 to 13.4; P < 0.0001).

Discussion

In this imputability analysis, the incidence of neurotoxicity due to high-dose thiotepa was estimated at 18.3%. To our knowledge, this is the first study to describe the incidence of neurotoxicity related to high-dose thiotepa, which had been administered below the defined limiting dose (1005 to 1125 mg/m2) in all the courses, in accordance with the consensus guidelines [21]. Neurological complications have previously been reported after thiotepa combined with total body irradiation [2527] or with another cytotoxic agent such as busulfan [28], busulfan and melphalan [29], carboplatin and cyclophosphamide [30], etoposide [31]. It is noteworthy that most children with HDCT receive many other medicines that can cause various neurological signs: opioids and drowsiness, anti-emetics and extrapyramidal syndrome, benzodiazepine and behavioural disorders [32]. Causality studies had then failed to demonstrate unambiguous involvement of thiotepa in the development of neurological disorders. NAE observed in our study are mostly of central nervous system origin with 9.6% of symptoms described as severe (grade 3 or 4). All NAE were reversible and devoid of sequelae, and had not contraindicated rechallenge with high-dose thiotepa.
Various pathomechanisms can explain drug-induced neurological disorders. In this study, we showed that a brain tumour could be an independent risk factor, which could increase the risk for thiotepa-related neurotoxicity 2.2-fold. Drugs with the highest neurotoxicity are therefore those that readily cross the blood-brain barrier. Lipid-soluble agents with a low molecular weight, such as thiotepa and tepa enter the brain easily [32]. Damage to the blood-brain barrier could facilitate the passage of drugs into the brain. Diseases such as malignant brain tumours, which damage the blood-brain barrier, would facilitate the direct neurotoxic effect of such drugs [33, 34].
In our study, we found no influence of renal failure on the occurrence of high-dose thiotepa- related NAE. However, these results should be considered with caution. Only 12 children had renal disruption, classified as an early-stage disorder. Among them, two children had experienced a NAE. Even moderate renal disruption has been described as possibly capable of increasing exposure to thiotepa by 43% and exposure to tepa by 157% [35]. Therefore thiotepa dose adjustments regarding renal clearance remain recommended.
In this study, we showed that intake of a weak opioid, especially tramadol, could be an independent risk factor for thiotepa-related NAE. Drug-related iatrogenia is a major source of treatment failures, especially when multiple medications are used. CYP3A4 and CYP2B6 are the main cytochrome enzymes involved in the metabolism of thiotepa to tepa [12, 13]. Thiotepa was described to be a highly potent, irreversible inhibitor of CYP2B6, which can lead to a risk of a self-induced overdose via a metabolic predisposition of each patient. Caution should be exercised when using thiotepa combined with CYP2B6 substrates, such as tramadol [36, 37]. In addition, aprepitant was described as an CYP3A4 inhibitor which can lead to an overdose of thiotepa [38, 39]. In this study, comedication with aprepitant was not significantly associated with NAE, possibly because of a lack of statistical power (only 3 to 7 patients had been treated with that drug).
Otherwise, interindividual variability in alkylating agent pharmacokinetics may lead to unpredictable toxicity and efficacy. A poor metabolizer phenotype was described as responsible for an increase in exposure to thiotepa and for an increase in the severity of mucositis [40]. This variability is particularly important in cancer therapy due to the narrow therapeutic window of anticancer agents. Interindividual variability in thiotepa clearance has been described as ranging from 28 to 90%, with variability of exposure to tepa after the administration of high-dose thiotepa ranging from 15 to 50% [41]. A pharmacogenetic analysis could therefore be considered before starting high-dose chemotherapy to better predict the metabolizer phenotype [17].
In our study, we have shown that tramadol used concomitantly with high-dose thiotepa could increase the risk of NAE 6.3-fold. Tramadol is a synthetic opiate that is chemically similar to codeine. Its antinociceptive effects are mediated via a combination of the agonist effects of μ-opioid receptors and inhibition of the reuptake of serotonin and norepinephrine [42]. This second property is an important element of analgesia, but also a major element in the risk of developing NAE. Indeed, tramadol is known to lower the seizure threshold and has been reported to be responsible for seizures at therapeutic and toxic doses [4345]. Similarly, the risk of serotonin syndrome is possible at the usual doses, and seems more common when overused or combined with potent inhibitors of metabolic pathways [46]. The main metabolite of tramadol, which has a 200-fold higher agonist effect for μ-opioid receptors than tramadol, results from the metabolism of tramadol by CYP2D6. However, another metabolic pathway of tramadol, as described above, passes through CYP2B6 and CYP3A4 [47]. Thus, concomitant use of tramadol and high-dose thiotepa, a CYP2B6 inhibitor, could be responsible for an overdose of tramadol, which could promote the emergence of neurological complications via monoaminergic overactivity.
An alternate hypothesis is that there is an unmeasured confounder that leads to both increased risk of NAE and increased need for pain control, hence the observed association with exposure to the weak opiates.
In clinical research, the gold standard is the prospective randomized controlled trial. Retrospective cohort studies, as performed in our study, are in principle subject to multiple risks of bias [48]. Selection bias of patients was reduced or cancelled by the decision to analyze all children with solid tumours treated with high-dose thiotepa followed by ASCT. This bias could therefore only occur if the active file of patients under treatment in the paediatric oncology unit had already been selected simply through admission to the unit. Concerning NAE, it is likely that all qualifying events have not been recorded across the 25 year period, and in particular for low-grade events that may be missing from the records. Regarding interpretation bias, the reading grid was carried out before and the collection was standardized for all patients. Finally, we sought to prevent confounding bias by conducting an analysis of univariate and multivariable logistic regression.

Conclusions

This study estimates an incidence of 18.3% of high-dose thiotepa-related NAE. These neurological complications resolved without sequelae and rechallenge with thiotepa was possible. A brain tumour and tramadol treatment were found to be possible independent risk factors for thiotepa-related neurotoxicity. Identifying markers for individual susceptibilities is mandatory in order to develop customised treatment approaches for cancer patients treated with intensive chemotherapy.

Acknowledgements

We thank Lorna Saint Ange for editing this manuscript.

Funding

None

Availability of data and materials

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.
This study was approved by the local ethics committee of Gustave Roussy (n°26/007). The parents/guardians gave their written informed consent for the retrospective analysis of clinical data according to the institutional review board of the Gustave Roussy Campus.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Banna GL, Simonelli M, Santoro A. High-dose chemotherapy followed by autologous hematopoietic stem-cell transplantation for the treatment of solid tumors in adults: a critical review. Curr Stem Cell Res Ther. 2007;2:65–82.CrossRefPubMed Banna GL, Simonelli M, Santoro A. High-dose chemotherapy followed by autologous hematopoietic stem-cell transplantation for the treatment of solid tumors in adults: a critical review. Curr Stem Cell Res Ther. 2007;2:65–82.CrossRefPubMed
2.
Zurück zum Zitat Ladenstein R, Philip T, Gardner H. Autologous stem cell transplantation for solid tumors in children. Curr Opin Pediatr. 1997;9:55–69.CrossRefPubMed Ladenstein R, Philip T, Gardner H. Autologous stem cell transplantation for solid tumors in children. Curr Opin Pediatr. 1997;9:55–69.CrossRefPubMed
3.
Zurück zum Zitat Frei E 3rd, Teicher BA, Holden SA, Cathcart KN, Wang YY. Preclinical studies and clinical correlation of the effect of alkylating dose. Cancer Res. 1988;48:6417–23.PubMed Frei E 3rd, Teicher BA, Holden SA, Cathcart KN, Wang YY. Preclinical studies and clinical correlation of the effect of alkylating dose. Cancer Res. 1988;48:6417–23.PubMed
4.
Zurück zum Zitat Finlay JL, Goldman S, Wong MC, Cairo M, Garvin J, August C, Cohen BH, Stanley P, Zimmerman RA, Bostrom B, Geyer JR, Harris RE, Sanders J, Yates AJ, Boyett JM, Packer RJ. Pilot study of high-dose thiotepa and etoposide with autologous bone marrow rescue in children and young adults with recurrent CNS tumors. The Children's cancer group. J Clin Oncol. 1996;14:2495–503.CrossRefPubMed Finlay JL, Goldman S, Wong MC, Cairo M, Garvin J, August C, Cohen BH, Stanley P, Zimmerman RA, Bostrom B, Geyer JR, Harris RE, Sanders J, Yates AJ, Boyett JM, Packer RJ. Pilot study of high-dose thiotepa and etoposide with autologous bone marrow rescue in children and young adults with recurrent CNS tumors. The Children's cancer group. J Clin Oncol. 1996;14:2495–503.CrossRefPubMed
5.
Zurück zum Zitat Ridola V, Grill J, Doz F, Gentet JC, Frappaz D, Raquin MA, Habrand JL, Sainte-Rose C, Valteau-Couanet D, Kalifa C. High-dose chemotherapy with autologous stem cell rescue followed by posterior fossa irradiation for local medulloblastoma recurrence or progression after conventional chemotherapy. Cancer. 2007;110:156–63.CrossRefPubMed Ridola V, Grill J, Doz F, Gentet JC, Frappaz D, Raquin MA, Habrand JL, Sainte-Rose C, Valteau-Couanet D, Kalifa C. High-dose chemotherapy with autologous stem cell rescue followed by posterior fossa irradiation for local medulloblastoma recurrence or progression after conventional chemotherapy. Cancer. 2007;110:156–63.CrossRefPubMed
6.
Zurück zum Zitat Saarinen-Pihkala UM, Hovi L, Koivusalo A, Jahnukainen K, Karikoski R, Sariola H, Wikström S. Thiotepa and melphalan based single, tandem, and triple high dose therapy and autologous stem cell transplantation for high risk neuroblastoma. Pediatr Blood Cancer. 2012;59:1190–7.CrossRefPubMed Saarinen-Pihkala UM, Hovi L, Koivusalo A, Jahnukainen K, Karikoski R, Sariola H, Wikström S. Thiotepa and melphalan based single, tandem, and triple high dose therapy and autologous stem cell transplantation for high risk neuroblastoma. Pediatr Blood Cancer. 2012;59:1190–7.CrossRefPubMed
7.
Zurück zum Zitat Saarinen UM, Pihko H, Makipernaa A. High-dose thiotepa with autologous bone marrow rescue in recurrent malignant oligodendroglioma: a case report. J Neuro-Oncol. 1990;9:57–61.CrossRef Saarinen UM, Pihko H, Makipernaa A. High-dose thiotepa with autologous bone marrow rescue in recurrent malignant oligodendroglioma: a case report. J Neuro-Oncol. 1990;9:57–61.CrossRef
8.
Zurück zum Zitat Saarinen UM, Hovi L, Makipernaa A, Riikonen P. High-dose thiotepa with autologous bone marrow rescue in pediatric solid tumors. Bone Marrow Transplant. 1991;8:369–76.PubMed Saarinen UM, Hovi L, Makipernaa A, Riikonen P. High-dose thiotepa with autologous bone marrow rescue in pediatric solid tumors. Bone Marrow Transplant. 1991;8:369–76.PubMed
9.
Zurück zum Zitat Tabone MD, Kalifa C, Rodary C, Raquin M, Valteau-Couanet D, Lemerle J. Osteosarcoma recurrences in pediatric patients previously treated with intensive chemotherapy. J Clin Oncol. 1994;12:2614–20.CrossRefPubMed Tabone MD, Kalifa C, Rodary C, Raquin M, Valteau-Couanet D, Lemerle J. Osteosarcoma recurrences in pediatric patients previously treated with intensive chemotherapy. J Clin Oncol. 1994;12:2614–20.CrossRefPubMed
10.
Zurück zum Zitat Huitema AD, Mathot RA, Tibben MM, Schellens JH, Rodenhuis S, Beijnen JH. Population pharmacokinetics of thioTEPA and its active metabolite TEPA in patients undergoing high-dose chemotherapy. Br J Clin Pharmacol. 2001;51:61–70.CrossRefPubMedCentralPubMed Huitema AD, Mathot RA, Tibben MM, Schellens JH, Rodenhuis S, Beijnen JH. Population pharmacokinetics of thioTEPA and its active metabolite TEPA in patients undergoing high-dose chemotherapy. Br J Clin Pharmacol. 2001;51:61–70.CrossRefPubMedCentralPubMed
11.
Zurück zum Zitat Miller B, Tenenholz T, Egorin MJ, Sosnovsky G, Rao NU, Gutierrez PL. Cellular pharmacology of N,N',N”-triethylene thiophosphoramide. Cancer Lett 1988;41:157–168. Miller B, Tenenholz T, Egorin MJ, Sosnovsky G, Rao NU, Gutierrez PL. Cellular pharmacology of N,N',N”-triethylene thiophosphoramide. Cancer Lett 1988;41:157–168.
12.
Zurück zum Zitat Jacobson PA, Green K, Birnbaum A, Remmel RP. Cytochrome P450 isozymes 3A4 and 2B6 are involved in the in vitro human metabolism of thiotepa to TEPA. Cancer Chemother Pharmacol. 2002;49:461–7.CrossRefPubMed Jacobson PA, Green K, Birnbaum A, Remmel RP. Cytochrome P450 isozymes 3A4 and 2B6 are involved in the in vitro human metabolism of thiotepa to TEPA. Cancer Chemother Pharmacol. 2002;49:461–7.CrossRefPubMed
13.
Zurück zum Zitat Dirven HA, Dictus EL, Broeders NL, van Ommen B, van Bladeren PJ. The role of human glutathione S-transferase isoenzymes in the formation of glutathione conjugates of the alkylating cytostatic drug thiotepa. Cancer Res. 1995;55:1701–6.PubMed Dirven HA, Dictus EL, Broeders NL, van Ommen B, van Bladeren PJ. The role of human glutathione S-transferase isoenzymes in the formation of glutathione conjugates of the alkylating cytostatic drug thiotepa. Cancer Res. 1995;55:1701–6.PubMed
14.
Zurück zum Zitat Heideman RL, Packer RJ, Reaman GH, Allen JC, Lange B, Horowitz ME, Steinberg SM, Gillespie A, Kovnar EH, Balis FM. A phase II evaluation of thiotepa in pediatric central nervous system malignancies. Cancer. 1993;72:271–5.CrossRefPubMed Heideman RL, Packer RJ, Reaman GH, Allen JC, Lange B, Horowitz ME, Steinberg SM, Gillespie A, Kovnar EH, Balis FM. A phase II evaluation of thiotepa in pediatric central nervous system malignancies. Cancer. 1993;72:271–5.CrossRefPubMed
15.
Zurück zum Zitat Maanen MJ, Smeets CJ, Beijnen JH. Chemistry, pharmacology and pharmacokinetics of N,N',N" -triethylenethiophosphoramide (ThioTEPA). Cancer Treat Rev 2000;26:257–268. Maanen MJ, Smeets CJ, Beijnen JH. Chemistry, pharmacology and pharmacokinetics of N,N',N" -triethylenethiophosphoramide (ThioTEPA). Cancer Treat Rev 2000;26:257–268.
16.
Zurück zum Zitat Cohen BE, Egorin MJ, Kohlhepp EA, Aisner J, Gutierrez PL. Human plasma pharmacokinetics and urinary excretion of thiotepa and its metabolites. Cancer Treat Rep. 1986;70:859–64.PubMed Cohen BE, Egorin MJ, Kohlhepp EA, Aisner J, Gutierrez PL. Human plasma pharmacokinetics and urinary excretion of thiotepa and its metabolites. Cancer Treat Rep. 1986;70:859–64.PubMed
17.
Zurück zum Zitat Ekhart C, Doodeman VD, Rodenhuis S, Smits PH, Beijnen JH, Huitema AD. Polymorphisms of drug-metabolizing enzymes (GST, CYP2B6 and CYP3A) affect the pharmacokinetics of thiotepa and tepa. Br J Clin Pharmacol. 2009;67:50–60.CrossRefPubMedCentralPubMed Ekhart C, Doodeman VD, Rodenhuis S, Smits PH, Beijnen JH, Huitema AD. Polymorphisms of drug-metabolizing enzymes (GST, CYP2B6 and CYP3A) affect the pharmacokinetics of thiotepa and tepa. Br J Clin Pharmacol. 2009;67:50–60.CrossRefPubMedCentralPubMed
18.
Zurück zum Zitat Herzig R, Brown R, Fay J. Phase I and II studies of high dose N,N′,N″-triethylene thiophosphoramide and autologous marrow transplantation in patients with refractory malignancies. Cancer Res Ther Control 1990;1:141–153. Herzig R, Brown R, Fay J. Phase I and II studies of high dose N,N′,N″-triethylene thiophosphoramide and autologous marrow transplantation in patients with refractory malignancies. Cancer Res Ther Control 1990;1:141–153.
19.
Zurück zum Zitat Lucidarme N, Valteau-Couanet D, Oberlin O, Couanet D, Kalifa C, Beaujean F, Lapierre V, Hartmann O. Phase II study of high-dose thiotepa and hematopoietic stem cell transplantation in children with solid tumors. Bone Marrow Transplant. 1998;22:535–40.CrossRefPubMed Lucidarme N, Valteau-Couanet D, Oberlin O, Couanet D, Kalifa C, Beaujean F, Lapierre V, Hartmann O. Phase II study of high-dose thiotepa and hematopoietic stem cell transplantation in children with solid tumors. Bone Marrow Transplant. 1998;22:535–40.CrossRefPubMed
20.
Zurück zum Zitat Przepiorka D, Madden T, Ippoliti C, Estrov Z, Dimopoulos M. Dosing of thioTEPA for myeloablative therapy. Cancer Chemother Pharmacol. 1995;37:155–60.CrossRefPubMed Przepiorka D, Madden T, Ippoliti C, Estrov Z, Dimopoulos M. Dosing of thioTEPA for myeloablative therapy. Cancer Chemother Pharmacol. 1995;37:155–60.CrossRefPubMed
21.
Zurück zum Zitat Wolff SN, Herzig RH, Fay JW, LeMaistre CF, Brown RA, Frei-Lahr D, Stranjord S, Giannone L, Coccia P, Weick JL. High-dose N,N',N"-triethylenethiophosphoramide (thiotepa) with autologous bone marrow transplantation: phase I studies. Semin Oncol 1990;17:2–6. Wolff SN, Herzig RH, Fay JW, LeMaistre CF, Brown RA, Frei-Lahr D, Stranjord S, Giannone L, Coccia P, Weick JL. High-dose N,N',N"-triethylenethiophosphoramide (thiotepa) with autologous bone marrow transplantation: phase I studies. Semin Oncol 1990;17:2–6.
22.
Zurück zum Zitat Lazarus HM, Reed MD, Spitzer TR, Rabaa MS, Blumer JL. High-dose i.v. thiotepa and cryopreserved autologous bone marrow transplantation for therapy of refractory cancer. Cancer Treat Rep. 1987;71:689–95.PubMed Lazarus HM, Reed MD, Spitzer TR, Rabaa MS, Blumer JL. High-dose i.v. thiotepa and cryopreserved autologous bone marrow transplantation for therapy of refractory cancer. Cancer Treat Rep. 1987;71:689–95.PubMed
23.
Zurück zum Zitat Schwartz GJ, Haycock GB, Edelmann CM Jr, Spitzer A. A Simple estimate of glomerular filtration rate in children derived from body length and plasma creatinine. Pediatrics. 1976;58:259–63.PubMed Schwartz GJ, Haycock GB, Edelmann CM Jr, Spitzer A. A Simple estimate of glomerular filtration rate in children derived from body length and plasma creatinine. Pediatrics. 1976;58:259–63.PubMed
24.
Zurück zum Zitat Begaud B. Criteria of imputability in accidents of drug-induced origin. Rev Prat. 2000;50:1803–6.PubMed Begaud B. Criteria of imputability in accidents of drug-induced origin. Rev Prat. 2000;50:1803–6.PubMed
25.
Zurück zum Zitat Freilich RJ, Delattre JY, Monjour A, DeAngelis LM. Chemotherapy without radiation therapy as initial treatment for primary CNS lymphoma in older patients. Neurology. 1996;46:435–9.CrossRefPubMed Freilich RJ, Delattre JY, Monjour A, DeAngelis LM. Chemotherapy without radiation therapy as initial treatment for primary CNS lymphoma in older patients. Neurology. 1996;46:435–9.CrossRefPubMed
26.
Zurück zum Zitat Weiss HD, Walker MD, Wiernik PH. Neurotoxicity of commonly used antineoplastic agents (first of two parts). N Engl J Med. 1974;291:75–81.CrossRefPubMed Weiss HD, Walker MD, Wiernik PH. Neurotoxicity of commonly used antineoplastic agents (first of two parts). N Engl J Med. 1974;291:75–81.CrossRefPubMed
27.
Zurück zum Zitat Kaplan RS, Wiernik PH. Neurotoxicity of antineoplastic drugs. Semin Oncol. 1982;9:103–30.PubMed Kaplan RS, Wiernik PH. Neurotoxicity of antineoplastic drugs. Semin Oncol. 1982;9:103–30.PubMed
28.
Zurück zum Zitat Kalifa C, Hartmann O, Demeocq F, Vassal G, Couanet D, Terrier-Lacombe MJ, Valteau D, Brugieres L, Lemerle J. High-dose Busulfan and thiotepa with autologous bone marrow transplantation in childhood malignant brain tumors: a phase II study. Bone Marrow Transplant. 1992;9:227–33.PubMed Kalifa C, Hartmann O, Demeocq F, Vassal G, Couanet D, Terrier-Lacombe MJ, Valteau D, Brugieres L, Lemerle J. High-dose Busulfan and thiotepa with autologous bone marrow transplantation in childhood malignant brain tumors: a phase II study. Bone Marrow Transplant. 1992;9:227–33.PubMed
29.
Zurück zum Zitat Hara J, Osugi Y, Ohta H, Matsuda Y, Nakanishi K, Takai K, Fujisaki H, Tokimasa S, Fukuzawa M, Okada A, Okada S. Double-conditioning regimens consisting of thiotepa, melphalan and busulfan with stem cell rescue for the treatment of pediatric solid tumors. Bone Marrow Transplant. 1998;22:7–12.CrossRefPubMed Hara J, Osugi Y, Ohta H, Matsuda Y, Nakanishi K, Takai K, Fujisaki H, Tokimasa S, Fukuzawa M, Okada A, Okada S. Double-conditioning regimens consisting of thiotepa, melphalan and busulfan with stem cell rescue for the treatment of pediatric solid tumors. Bone Marrow Transplant. 1998;22:7–12.CrossRefPubMed
30.
Zurück zum Zitat Antman K, Eder JP, Elias A, Ayash L, Shea TC, Weissman L, Critchlow J, Schryber SM, Begg C, Teicher BA. High-dose thiotepa alone and in combination regimens with bone marrow support. Semin Oncol. 1990;17:33–8.PubMed Antman K, Eder JP, Elias A, Ayash L, Shea TC, Weissman L, Critchlow J, Schryber SM, Begg C, Teicher BA. High-dose thiotepa alone and in combination regimens with bone marrow support. Semin Oncol. 1990;17:33–8.PubMed
31.
Zurück zum Zitat Kramer ED, Packer RJ, Ginsberg J, Goldman S, Thompson S, Bayer LA, Shen V, Harris R, Khan S, Finlay JL. Acute neurologic dysfunction associated with high-dose chemotherapy and autologous bone marrow rescue for primary malignant brain tumors. Pediatr Neurosurg. 1997;27:23–237.CrossRef Kramer ED, Packer RJ, Ginsberg J, Goldman S, Thompson S, Bayer LA, Shen V, Harris R, Khan S, Finlay JL. Acute neurologic dysfunction associated with high-dose chemotherapy and autologous bone marrow rescue for primary malignant brain tumors. Pediatr Neurosurg. 1997;27:23–237.CrossRef
32.
Zurück zum Zitat Orbach D, Brisse H, Doz F. Central neurological manifestations during chemotherapy in children. Arch Pediatr. 2003;10:533–9.CrossRefPubMed Orbach D, Brisse H, Doz F. Central neurological manifestations during chemotherapy in children. Arch Pediatr. 2003;10:533–9.CrossRefPubMed
33.
Zurück zum Zitat Provenzale JM, Mukundan S, Dewhirst M. The role of blood-brain barrier permeability in brain tumor imaging and therapeutics. AJR Am J Roentgenol. 2005;185:763–7.CrossRefPubMed Provenzale JM, Mukundan S, Dewhirst M. The role of blood-brain barrier permeability in brain tumor imaging and therapeutics. AJR Am J Roentgenol. 2005;185:763–7.CrossRefPubMed
34.
Zurück zum Zitat Deeken JF, Loscher W. The blood-brain barrier and cancer: transporters, treatment, and Trojan horses. Clin Cancer Res. 2007;13:1663–74.CrossRefPubMed Deeken JF, Loscher W. The blood-brain barrier and cancer: transporters, treatment, and Trojan horses. Clin Cancer Res. 2007;13:1663–74.CrossRefPubMed
35.
Zurück zum Zitat Ekhart C, Kerst JM, Rodenhuis S, Beijnen JH, Huitema AD. Altered cyclophosphamide and thiotepa pharmacokinetics in a patient with moderate renal insufficiency. Cancer Chemother Pharmacol. 2009;63:375–9.CrossRefPubMed Ekhart C, Kerst JM, Rodenhuis S, Beijnen JH, Huitema AD. Altered cyclophosphamide and thiotepa pharmacokinetics in a patient with moderate renal insufficiency. Cancer Chemother Pharmacol. 2009;63:375–9.CrossRefPubMed
36.
Zurück zum Zitat Richter T, Schwab M, Eichelbaum M, Zanger UM. Inhibition of human CYP2B6 by N,N',N”-triethylenethiophosphoramide is irreversible and mechanism-based. Biochem Pharmacol 2005;69:517–524. Richter T, Schwab M, Eichelbaum M, Zanger UM. Inhibition of human CYP2B6 by N,N',N”-triethylenethiophosphoramide is irreversible and mechanism-based. Biochem Pharmacol 2005;69:517–524.
37.
Zurück zum Zitat Nelson EM, Philbrick AM. Avoiding serotonin syndrome: the nature of the interaction between tramadol and selective serotonin reuptake inhibitors. Ann Pharmacother. 2012;46:1712–6.CrossRefPubMed Nelson EM, Philbrick AM. Avoiding serotonin syndrome: the nature of the interaction between tramadol and selective serotonin reuptake inhibitors. Ann Pharmacother. 2012;46:1712–6.CrossRefPubMed
38.
Zurück zum Zitat de Jonge ME, Huitema AD, Holtkamp MJ, van Dam SM, Beijnen JH, Rodenhuis S. Aprepitant inhibits cyclophosphamide bioactivation and thiotepa metabolism. Cancer Chemother Pharmacol. 2005;56:370–8.CrossRefPubMed de Jonge ME, Huitema AD, Holtkamp MJ, van Dam SM, Beijnen JH, Rodenhuis S. Aprepitant inhibits cyclophosphamide bioactivation and thiotepa metabolism. Cancer Chemother Pharmacol. 2005;56:370–8.CrossRefPubMed
39.
Zurück zum Zitat Howell JE, Szabatura AH, Hatfield Seung A, Nesbit SA. Characterization of the occurrence of ifosfamide-induced neurotoxicity with concomitant aprepitant. J Oncol Pharm Pract. 2008;14:157–62.CrossRefPubMed Howell JE, Szabatura AH, Hatfield Seung A, Nesbit SA. Characterization of the occurrence of ifosfamide-induced neurotoxicity with concomitant aprepitant. J Oncol Pharm Pract. 2008;14:157–62.CrossRefPubMed
40.
Zurück zum Zitat Huitema AD, Spaander M, Mathjt RA, Tibben MM, Holtkamp MJ, Beijnen JH, Rodenhuis S. Relationship between exposure and toxicity in high-dose chemotherapy with cyclophosphamide, thiotepa and carboplatin. Ann Oncol. 2002;13:374–84.CrossRefPubMed Huitema AD, Spaander M, Mathjt RA, Tibben MM, Holtkamp MJ, Beijnen JH, Rodenhuis S. Relationship between exposure and toxicity in high-dose chemotherapy with cyclophosphamide, thiotepa and carboplatin. Ann Oncol. 2002;13:374–84.CrossRefPubMed
41.
Zurück zum Zitat Huitema AD, Smits KD, Mathot RA, Schellens JH, Rodenhuis S, Beijnen JH. The clinical pharmacology of alkylating agents in high-dose chemotherapy. Anti-Cancer Drugs. 2000;11:515–33.CrossRefPubMed Huitema AD, Smits KD, Mathot RA, Schellens JH, Rodenhuis S, Beijnen JH. The clinical pharmacology of alkylating agents in high-dose chemotherapy. Anti-Cancer Drugs. 2000;11:515–33.CrossRefPubMed
42.
43.
Zurück zum Zitat Jovanovic-Cupic V, Martinovic Z, Nesic N. Seizures associated with intoxication and abuse of tramadol. Clin Toxicol (Phila). 2006;44:143–6.CrossRef Jovanovic-Cupic V, Martinovic Z, Nesic N. Seizures associated with intoxication and abuse of tramadol. Clin Toxicol (Phila). 2006;44:143–6.CrossRef
44.
Zurück zum Zitat Ripple MG, Pestaner JP, Levine BS, Smialek JE. Lethal combination of tramadol and multiple drugs affecting serotonin. Am J Forensic Med Pathol. 2000;21(4):370.CrossRefPubMed Ripple MG, Pestaner JP, Levine BS, Smialek JE. Lethal combination of tramadol and multiple drugs affecting serotonin. Am J Forensic Med Pathol. 2000;21(4):370.CrossRefPubMed
45.
Zurück zum Zitat Daubin C, Quentin C, Goulle JP, Guillotin D, Lehoux P, Lepage O, Charbonneau P. Refractory shock and asystole related to tramadol overdose. Clin Toxicol (Phila). 2007;45:961–4.CrossRef Daubin C, Quentin C, Goulle JP, Guillotin D, Lehoux P, Lepage O, Charbonneau P. Refractory shock and asystole related to tramadol overdose. Clin Toxicol (Phila). 2007;45:961–4.CrossRef
46.
Zurück zum Zitat Sansone RA, Sansone LA. Tramadol: seizures, serotonin syndrome, and coadministered antidepressants. Psychiatry (Edgmont). 2009;6:17–21. Sansone RA, Sansone LA. Tramadol: seizures, serotonin syndrome, and coadministered antidepressants. Psychiatry (Edgmont). 2009;6:17–21.
47.
Zurück zum Zitat Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin Pharmacokinet. 2004;43:879–923.CrossRefPubMed Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin Pharmacokinet. 2004;43:879–923.CrossRefPubMed
48.
Zurück zum Zitat Tripepi G, Jager KJ, Dekker FW, Zoccali C. Selection bias and information bias in clinical research. Nephron Clin Pract. 2010;115:94–9.CrossRef Tripepi G, Jager KJ, Dekker FW, Zoccali C. Selection bias and information bias in clinical research. Nephron Clin Pract. 2010;115:94–9.CrossRef
Metadaten
Titel
High-dose thiotepa-related neurotoxicity and the role of tramadol in children
verfasst von
Christophe Maritaz
Francois Lemare
Agnes Laplanche
Sylvie Demirdjian
Dominique Valteau-Couanet
Christelle Dufour
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2018
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4090-6

Weitere Artikel der Ausgabe 1/2018

BMC Cancer 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.