Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 May 26.
Published in final edited form as: J Neurooncol. 2010 May 5;101(1):57–66. doi: 10.1007/s11060-010-0217-6

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

David A Reardon 1,, James J Vredenburgh 2, Annick Desjardins 3, Katherine Peters 4, Sridharan Gururangan 5, John H Sampson 6, Jennifer Marcello 7, James E Herndon II 8, Roger E McLendon 9, Dorothea Janney 10, Allan H Friedman 11, Darell D Bigner 12, Henry S Friedman 13
PMCID: PMC3102498  NIHMSID: NIHMS285417  PMID: 20443129

Abstract

Sorafenib, an oral VEGFR-2, Raf, PDGFR, c-KIT and Flt-3 inhibitor, is active against renal cell and hepatocellular carcinomas, and has recently demonstrated promising activity for lung and breast cancers. In addition, various protracted temozolomide dosing schedules have been evaluated as a strategy to further enhance its anti-tumor activity. We reasoned that sorafenib and protracted, daily temozolomide may provide complementary therapeutic benefit, and therefore performed a phase 2 trial among recurrent glioblastoma patients. Adult glioblastoma patients at any recurrence after standard temozolomide chemoradiotherapy received sorafenib (400 mg twice daily) and continuous daily temozolomide (50 mg/m2/day). Assessments were performed every eight weeks. The primary endpoint was progression-free survival at 6 months (PFS-6) and secondary end points were radiographic response, overall survival (OS), safety and sorafenib pharmacokinetics. Of 32 enrolled patients, 12 (38%) were on CYP3-A inducing anti-epileptics (EIAEDs), 17 (53%) had 2 or more prior progressions, 15 had progressed while receiving 5-day temozolomide, and 12 (38%) had failed either prior bevacizumab or VEGFR inhibitor therapy. The most common grade ≥ 3 toxicities were palmer-planter erythrodysesthesia (19%) and elevated amylase/lipase (13%). Sorafenib pharmacokinetic exposures were comparable on day 1 regardless of EIAED status, but significantly lower on day 28 for patients on EIAEDs (P = 0.0431). With a median follow-up of 93 weeks, PFS-6 was 9.4%. Only one patient (3%) achieved a partial response. In conclusion, sorafenib can be safely administered with daily temozolomide, but this regimen has limited activity for recurrent GBM. Co-administration of EIAEDs can lower sorafenib exposures in this population.

Keywords: Sorafenib, Temozolomide, Glioblastoma, Raf, VEGF

Introduction

Recurrence among glioblastoma (GBM) patients following conventional therapy with surgical debulking, external beam radiotherapy and temozolomide chemotherapy remains essentially universal. The identification of effective salvage therapies continues to be an elusive yet paramount challenge. In the pre-temozolomide era, the 6-month progression-free survival rate (PFS-6) for salvage therapies was approximately 15% [1]. Unfortunately, treatments following recurrence in the modern, temozolomide era have yielded similarly poor PFS-6 rates [2, 3].

However, therapeutics targeting vascular endothelial growth factor (VEGF) [48] or its cognate receptor (VEGFR)[9], have recently been shown to achieve durable anti-tumor benefit among some patients with recurrent malignant glioma. In fact, the FDA granted bevacizumab, a humanized monoclonal antibody against VEGF, accelerated approval for recurrent GBM patients earlier this year based on durable radiographic response [7, 8]. However, improvements in OS were only modestly improved compared to historical benchmarks suggesting that rationally designed combinatorial strategies may further enhance the anti-tumor benefit of VEGF/VEGFR-targeted therapeutics.

Sorafenib (BAY 43-9006; Bayer Pharmaceuticals, Inc. Montville, N.J.), an orally bioavailable inhibitor of VEGFR, Raf-1 and wild-type B-Raf, as well as PDGFR, c-KIT and Flt-3 [10], is FDA-approved for the treatment of renal cell carcinoma as well as hepatocellular carcinoma [11, 12].

Temozolomide, a second generation imidazotetrazine derivative, induces glioma cell apoptosis by methylating specific DNA sites, the most critical being the O6 position of guanine [13, 14]. Multiple recent clinical studies evaluating a variety of “dose-dense” temozolomide schedules as a strategy to overcome methylguanine methyltransferase (MGMT)-mediated temozolomide resistance have shown preliminary evidence of encouraging anti-glioma benefit [1525].

We hypothesized that the anti-tumor activity of sorafenib may be enhanced when administered with daily temozolomide. It is also possible, although unproven, that each of these agents may exert complementary anti-angiogenic effects which may also contribute to anti-tumor benefit. We therefore evaluated continuous daily dosing of sorafenib and temozolomide as a salvage regimen among recurrent GBM patients in the current single-arm phase 2 study.

Patients and methods

Protocol objectives

The primary objective of the study was to define the activity of sorafenib plus daily temozolomide in the treatment of adults with recurrent GBM as measured by PFS-6. Secondary objectives included: to further define the toxicity of this regimen; to evaluate other efficacy measures including radiographic response rate and overall survival (OS); and to determine the pharmacokinetics of sorafenib when combined with daily temozolomide among patients who are on and not on concurrently administered CYP3A-inducing anti-epileptics (EIAEDs).

Patient eligibility criteria

Patients were required to have histologically confirmed diagnosis of GBM that was recurrent following prior therapy as defined by the Macdonald criteria [26]. Eligibility also required: age ≥ 18 years; KPS ≥ 60%; stable corticosteroid dosing for at least 1 week prior to therapy initiation; adequate hematologic (hemoglobin ≥ 9 gm/dl; absolute neutrophil count ≥ 1500/mm3; platelet count ≥ 100,000/mm3), renal (serum creatinine ≤ 1.5 times the institutional upper limit of normal [X ULN]) and hepatic function (total bilirubin ≤ 1.5 X ULN, and AST and ALT ≤ 2.5 X ULN); PT/PTT within normal limits; at least 2 weeks between prior surgical resection (1 week for stereotactic biopsy), 4 weeks from prior chemotherapy or investigational therapy, and 12 weeks from prior radiotherapy, unless there was new radiographic enhancement outside the prior radiotherapy field or biopsy-proven recurrent tumor. All patients provided informed consent as approved by the Duke University Medical Center Institutional Review Board.

The following patients were excluded: pregnant or nursing women; those with reproductive potential and not using an effective contraceptive method; prior sorafenib therapy; prior bevacizumab therapy within 6 weeks of study enrollment; hypersensitivity to temozolomide; significant concurrent medical illness including NYHA class > 2 congestive heart failure, new onset angina within past 3 months or myocardial infarction or stroke within 6 months, cardiac dysrhythmia requiring anti-arrhythmic therapy, uncontrolled hypertension, bleeding diathesis or coagulopathy, gastrointestinal disease that could limit absorption, or infection requiring intravenous antibiotics. Patients who received prior temozolomide were eligible unless they had either hypersensitivity, unacceptable toxicity or progressive disease following daily temozolomide therapy. Patients who received prior anti-VEGF therapy were also eligible except for those who received bevacizumab within 6 weeks of study entry.

Treatment design

Sorafenib and temozolomide were administered orally on a continuous daily dosing schedule at 400 mg twice a day and 50 mg/m2 once daily, respectively. Patients were encouraged to take their temozolomide at either the same time or within 30 min of a sorafenib dose. Patients were advised to fast for 1 h before and 2 h after taking both temozolomide and sorafenib. Pneumocystis prophylaxis was recommended for all patients. For study purposes, a cycle consisted of 4 weeks of therapy.

Dose modification and retreatment criteria

Toxicity was graded according to NCI CTC version 2 (NCI 2004). Sorafenib was reduced to 400 mg once a day and then 400 mg every other day for grade 2 or greater palmer-plantar erythrodysesthesia (PPE), grade ≥ 3 neutropenia, thrombocytopenia or non-hematologic toxicity, or any attributable toxicity requiring greater than 2 weeks to satisfy re-treatment criteria. Temozolomide was reduced to 40 mg/m2 per day and then to 50 mg/m2 every other day for grade 4 neutropenia, grade 3 thrombocytopenia or any grade ≥ 3 non-hematopoietic attributable toxicity.

Retreatment after dose interruption or following each cycle required an ANC ≥ 500 cells/mm3, platelets ≥ 500 cells/mm3, SGOT and total bilirubin ≤ 2.5 X ULN and creatinine ≤_1.5 X ULN. In addition, all toxicities were required to resolve to grade ≤ 1 for retreatment.

Patients were removed from study for evidence of PD, grade 4 non-hematologic toxicity, more than 2 sorafenib dose reductions due to toxicity, noncompliance, or voluntary withdrawal.

Supportive care

Antiemetic therapy with serotonin antagonists were prescribed as needed. Loperamide was prescribed for diarrhea according to standard guidelines. Hematopoietic growth factors and blood products were administered according to institutional practice guidelines. Patients were encouraged to use skin emollients and protect pressure-sensitive areas early if they developed signs of PPE.

Evaluations prior to and during therapy

Patients underwent physical and neurologic examinations as well as MRI scans within 2 weeks of enrollment and before every other 4-week cycle. A complete blood count with differential was performed weekly, and a serum biochemistry profile as well as urinalysis were assessed every 4 weeks. A urinalysis and coagulation profile were performed prior to the first cycle, along with a beta human chorionic gonadotropin assay in women with reproductive potential.

Response evaluation

Response determination, performed by the study investigators, was based on neurologic examination and comparison of the baseline contrast-enhanced MRI scan with those performed before each cycle according to the Macdonald criteria [26]. A complete response (CR) was defined as the disappearance of all enhancing tumor from baseline on consecutive MRI scans at least 4 weeks apart, combined with discontinuation of corticosteroids and achievement of neurologic stability or improvement. A PR was defined as ≥50% reduction from baseline in the size (measured as the product of largest perpendicular diameters) of enhancing tumor maintained for at least 6 weeks with a stable or improved neurologic exam and a stable or reduced corticosteroid requirement. Patient response was defined based on the best response achieved at any point on study relative to the pre-treatment baseline MRI. Progressive disease was defined as more than 25% increase in size of enhancing tumor or any new enhancing lesion compared to baseline or smallest post-treatment assessment. Stable disease was assigned for assessments not meeting criteria for CR, PR, or PD.

Pharmacokinetics

Blood sampling for sorafenib pharmacokinetics was performed on days 1 and 28 of cycle 1, and was obtained before and at 0.5, 1, 2, 4, 6, 8 and 24 h after the AM sorafenib dose. For each sample, plasma supernatants were separated by centrifugation and immediately frozen at −20°C. Plasma concentrations of sorafenib were analyzed using a validated liquid chromatograph tandem mass spectrometer assay with a lower limit of quantification of 0.01 μg/ml. Pharmacokinetic variables measured were area under the plasma concentration–time curve from 0 to 8 h (AUC0–8), 0 to 24 h (AUC0–24), maximum plasma concentration (Cmax) and time to maximum concentration (Tmax). Plasma concentration–time data were evaluated by noncompartmental methods using WinNolin 4.0 (Pharsight, St. Louis, MO).

Statistical considerations

The primary goal of this study was to evaluate the PFS-6 of sorafenib and daily temozolomide in the treatment of patients with recurrent GBM. Given a 21% PFS-6 with 95% confidence interval of 13 to 29% for standard, 5-day temozolomide among patients at first relapse [27], we employed a single-stage phase 2 design to differentiate between a 5 and 20% PFS-6 rate among eligible patients with one or more relapses treated on the current study. The treatment regimen would be considered worthy of further evaluation if 4 or more of the total 32 patients remained progression-free for 6 months or longer. The following characteristics were true of this study design: (1) the type I error rate (α) or probability of erroneously concluding a treatment is active (P ≥ 0.2) when it actually is ineffective (P ≤ 0.05) was 0.074; (2) the type II error rate (β) or probability of erroneously concluding that the treatment is ineffective (P ≤ 0.05) when the treatment actually is active (P ≥ 0.2) was 0.093.

Efficacy and toxicity monitoring occurred after 16 patients were accrued to the study. If 10 or more patients died or developed disease progression, or 6 or more had developed grade 4 or 5 non-hematologic toxicity, accrual was to be suspended for a careful review of all data.

The product limit estimator developed by Kaplan & Meier was used to graphically describe the distribution of OS and PFS among patients. Survival was defined as the time between treatment initiation and death. For patients remaining alive, survival time was censored at the time of last follow-up. Progression free survival was defined as the time between treatment initiation and disease progression/relapse/death. Patients remaining alive and disease-free had PFS censored at last follow-up. Within the context of these plots/models, estimates of 6-month and 12-month survival and PFS were estimated.

The PFS and OS within subgroups defined by the following patient characteristics were compared using the logrank test: age (<50 vs. ≥50 years), KPS(<90 vs. ≥90%), number of prior episodes of progression (1 vs. >1), number of prior chemotherapeutics (1 vs. >1), prior bevacizumab (yes vs. no), prior bevacizumab or VEGFR tyrosine kinase inhibitor (yes vs. no), and concurrent use of EIAEDs (yes or no).

Due to the small sample sizes, non-parametric tests of statistical significance were used to compare AUC measurements between groups. The Sign test was used to compare day 1 AUC with Day 28 AUC for each group (i.e., two separate tests were performed—one for EIAC and one for non-EIAC). Only patients with measurements at both time points were included in this analysis. The Wilcoxon Rank Sum test was used to compare AUC values between groups. Separate tests were used to compare AUC at day 1 and day 28.

Results

Patient characteristics

Thirty-two patients with recurrent GBM were enrolled at Duke University Medical Center between October 2007 and November 2008 (Table 1). All patients were assessed for the primary endpoint and safety evaluations. One patient was not evaluable for radiographic response because he underwent a gross total resection immediately prior to enrollment. Twelve patients (38%) were on EIAEDs and 10 (31%) were on dexamethasone at study enrollment. Enrolled patients were moderately pre-treated; 17 (53%) had two or more prior episodes of progressive disease and had received two or more prior chemotherapy or investigational agents. Fifteen (47%) had progressive disease after prior 5-day temozolomide dosing and 12 (38%) had failed prior bevacizumab (n = 9) or VEGFR tyrosine kinase inhibitor therapy with vandetanib (n = 3). Following progression on the current study, 23 patients (72%) received salvage therapy with a bevacizumab-based regimen.

Table 1. Patient characteristics.

n = 32
Age, median (years) 53.6
 Range 30.4–72.5
Gender
 Male 23 (72)
 Female 9 (28)
KPS
 90–100 15 (47)
 80 12 (38)
 70 5 (16)
Time from diagnosis, median (weeks) 86.3
 Range 32.0–374.4
Anti-epileptic use
 EIAED 12 (38)
 Non-EIAED 10 (31)
 None 10 (31)
Dexamethasone at entry
 Yes 10 (31)
 No 22 (69)
# Prior PDs
 1 15 (47)
 2 9 (28)
 3 8 (25)
Prior XRT 32 (100)
Surgery prior to enrollment
 GTR 1 (3)
 STR 3 (9)
 Biopsy 2 (6)
 None 26 (81)
# Prior chemotherapy or investigational agents
 1 8 (25)
 2 4 (13)
 3 2 (6)
 4 4 (13)
 ≥5 9 (28)
Failure on prior 5-day temozolomide
 Yes 15 (47)
 No 17 (53)
Time from prior 5-day temozolomide (weeks)
 ≤12 6 (40)
 12–24 3 (20)
 ≥24 6 (40)
Prior failure on bevacizumab or VEGFR2 TKI
 Yes 13 (41)
 No 19 (59)

Toxicity

Seventy-three cycles of therapy were administered to patients enrolled on this study. Twenty-three patients (72%) received less than or equal to two cycles of therapy, 8 patients (25%) received 3–6 cycles and one patient received 13 cycles of therapy. Overall the regimen was well-tolerated (Table 2). Two patients experienced grade 4 events that included symptomatic elevation of amylase and lipase. For the first patient, this event developed during cycle 1 and required hospitalization, and resolved with treatment interruption and appropriate medical management. This patient had progressive disease at the end of cycle 1 and was taken off study. In the second patient, the amylase and lipase increased within 6 weeks of initiating study therapy, but resolved following a cholecystectomy. Following a brief interruption of therapy, sorafenib was increased to full dose in this patient and was tolerated during five additional cycles of therapy prior to the development of progressive disease. Of note, grade 2 and 3 elevation of amylase or lipase occurred in 2 and 4 patients, respectively. All of these patients were able to receive additional study therapy without further difficulty although the sorafenib dose was modified to 400 mg once daily in three of these patients. Reversible grade 2 and 3 palmar-plantar erythrodysesthesia (PPE) occurred in 1(3%) and 6 (19%) patients, respectively. Three of these patients required sorafenib dose modification, while the others responded to a brief (3–5 day) interruption of sorafenib dosing and supportive care. Other common adverse events included fatigue (25%), rash (22%) and infection (19%), but were limited to grade 2 in most patients. A variety of electrolyte disturbances, which responded to replacement therapy, were noted. As expected, hematologic abnormalities were uncommon. Only one patient developed an intracranial hemorrhage which was grade 1 and occurred at the time of tumor recurrence. There were no deaths on study attributable to study therapy.

Table 2. Toxicity summary: number of patients with each adverse event.

Toxicity Grade Dose modification or discontinuation


2 3 4 Sorafenib TMZ
Amylase/lipase 2 (6) 4 (13) 2 (6) 4 (13)
Anorexia 2 (6) 1 (3)
Constipation 2 (6)
Diarrhea 2 (6)
Dyspnea 2 (6) 2 (6)
Fatigue 6 (19) 2 (6)
Hyperbilirubinemia 1 (3)
Hypertension 1 (3)
Hypersensitivity 3 (9)
Hypoalbuminemia 3 (9)
Hypocalcemia 1 (3)
Hypokalemia 2 (6)
Hypomagnesemia 1 (3)
Hypophosphatemia 2 (6) 3 (9)
Infection 5 (16) 1 (3)
Mucositis 1 (3)
Nausea 3 (9) 1 (3)
Neutropenia 4 (13) 1 (3)
Palmer-Plantar
Erythrodysesthesia 1 (3) 6 (19) 3 (9)
Rash 7 (22)
Thrombocytopenia 2 (6)
Transaminase elevation 1 (3)

Number in parentheses is the percent of study population

Pharmacokinetics

Eight patients who were on EIAEDs had 24-h sorafenib concentration versus time profiles from both day 1 and day 28 of cycle 1. Fifteen patients who were not on EIAEDs underwent similar assessment for day 1 but only 11 of these patients had samples available for day 28 measurements (Table 3). Four patients who were on EIAEDs (50%) and five of those not on EIAEDs (33%) were also on dexamethasone during the period of their PK assessments. There were no noteworthy differences in other concomitant medications between patients on or not on EIAEDs. Of note, none of the patients on the EIAED arm who underwent PK sampling required sorafenib dose interruption or modification during cycle 1 of therapy. In contrast, five patients on the non-EIAED arm who underwent PK sampling required sorafenib dose interruption or modification during cycle 1. Sorafenib measures were comparable on day 1 among patients on and not on EIAEDs. For patients not on EIAEDs, day 28 sorafenib AUC0–24 was significantly higher compared to day 1 (P = 0.0117). This increase was expected based on established steady state kinetics, but may have also been mildly augmented by the discontinuation of dexamethasone (a mild CYP3A inducer) in 3 patients. Nonetheless, the day 28 increased exposure was noted despite one-third of the non-EIAED patients requiring decreased sorafenib dosing during cycle 1 due to toxicity compared to none of those on EIAEDs. In contrast, for patients on EIAEDs, sorafenib AUC0–24 did not differ significantly between day 1 and day 28 (P = 0.7266). Furthermore, sorafenib AUC0–24 was significantly higher on day 28 for patients not on EIAEDs compared to those who were on EIAEDs (P = 0.0431). For AUC0–8 comparisons, similar findings were observed, however, this analysis was limited by small sample size (n = 5 per group) and wide standard deviations. There was no difference in day 1exposures between patients on and not on EIAEDs, nor was there a difference between day 1 and day 28 exposures for patients on EIAEDs. For patients not on EIAEDs, day 1 and 28 AUC0–8 exposure did not achieve statistical significance when analyzed using the Sign test despite a nearly 5-fold difference in mean (day 1 = 11,082 vs. day 28 = 52061); however, the difference did achieve statistical significance using the paired t test (P = 0.0334).

Table 3. Sorafenib pharmacokinetics.

Patient Cohort T max (h) C max (μg/l) AUC(24) (μg*h/l)
EIAED
 Day 1 28 1 28 1 28
 # Patients 9 9 9 9 9 9
 Mean NA NA 3397.3 3813.9 45,309.7 47,148.2
 Median 8.2 2.1
 CV% NA NA 66.8 40.7 61.8 65.7
 Range 2.0–24.7 0–2.3
Non-EIAED
 Day 1 28 1 28 1 28
 # Patients 14 10 14 10 14 10
 Mean NA NA 3155.1 8118.8 45,238.7 128,820.8
 Median 24 4.2
 CV(%) NA NA 41.7 60.8 44.1 73.2
 Range 2.0–25.5 0–8.2

AUC(24) area under time curve of 24 h, Cmax maximum concentration, CV coefficient of variation, EIAEDCYP3A enzyme-inducing anti-epileptic drugs (phenytoin, phenobarbital, carbamazepine, oxcarbazepine and primidone), NA not applicable, Tmax median time to maximum concentration

Outcome

Confirmed partial response was observed in only 1 patient (3%). Fifteen patients achieved a best response of stable disease (47%), while 16 patients (50%) had progressive disease at first study assessment. With a median follow-up of 93 weeks (95% CI, 53.4, 106.3), the probability of remaining progression-free at 6 months was 9.4% (95% CI, 2.4, 22.3). The median progression-free and overall survivals were 6.4 weeks (95% CI, 3.9, 11.7 weeks) and 41.5 weeks (95% CI, 24.1, 55.1 weeks), respectively. The 1-year survival probability for patients with recurrent GBM was 34.4% (95% CI, 18.8, 50.6%).

Analyses provided no evidence that patient age, KPS, EIAED use, number of prior chemotherapy agents or prior bevacizumab treatment were associated with either PFS or OS; however, more than one prior PD was predictive of poorer OS (P = 0.0051).

Discussion

Despite potentially complementary direct and indirect mechanisms of anti-tumor activity, we demonstrate that sorafenib combined with daily temozolomide has minimal activity as a salvage regimen for recurrent GBM patients in this single-arm phase 2 study. Specifically, only one patient achieved a radiographic response and the PFS-6 was 9.4%. Sorafenib is an attractive therapeutic consideration in GBM because it can inhibit several biologically relevant oncogenic kinases including raf, a key mediator of the ras/MAPK kinase pathway [28]. Although mutations in ras or raf are uncommon in malignant gliomas [29], activation of the ras/MAPK pathway occurs frequently and may confer a poor prognosis [30]. Sorafenib can also block PDGFR, an oncogene associated with increased activity in malignant gliomas [3133]. as well as c-KIT, which has recently been shown to be amplified or overexpressed in a subset of GBM tumors [34, 35]. Sorafenib also may exert a potent indirect anti-tumor effect by blocking angiogenesis via inhibition of VEGFR as well as PDGFR [28].

Despite its diverse spectrum of biologically relevant targets, at the time this study was initiated, there was no data evaluating sorafenib among recurrent GBM patients. However, several studies have been completed or are ongoing to better define the anti-tumor benefit of sorafenib among recurrent as well as newly diagnosed malignant glioma patients. Of note, preliminary results of a phase I dose escalation study of sorafenib monotherapy among recurrent malignant glioma patients reported limited toxicity up to doses of 800 mg bid with further accrual ongoing to define an MTD [36]. Preliminary reports of additional studies combining sorafenib with either an mTOR inhibitor or an EGFR inhibitor reveal overall disappointing results and poor tolerance among recurrent malignant glioma patients [37, 38].

Protracted daily temozolomide was selected to partner with sorafenib in this study for two primary reasons. First, continuous daily dosing at 50 mg/m2/day provides a 1.4–1.9 fold greater temozolomide exposure compared to standard dosing at 150–200 mg/m2/day for 5 days each month. Although dose-dense schedules may enhance cytotoxicity simply due to greater tumor exposure, such schedules may also enhance anti-tumor activity by depleting MGMT. Although confirmation of this effect has not been proven in patient tumors, it has been demonstrated in peripheral blood mononuclear cells [39]. Furthermore, a wide array of “dose-dense” temozolomide schedules have shown promising anti-tumor benefit and acceptable toxicity in ongoing and completed clinical trials for malignant glioma patients [1523, 40]. In addition, recent reports demonstrate that protracted temozolomide may have anti-tumor benefit among recurrent malignant glioma patients including some who were previously treated with standard, 5-day temozolomide dosing [24, 25]. In a formal phase 2 study of daily temozolomide administered at 50 mg/m2/day, Perry and colleagues demonstrated that grade 3 malignant glioma patients and those who had progressed following completion of prior standard 5-day temozolomide achieved PFS-6 rates of 42 and 57%, respectively. In contrast, those who were treated with daily temozolomide following progression on standard 5-day temozolomide had a PFS-6 of only 17% [17]. These results led to a recently completed multicenter phase II study conducted by the NCIC. A preliminary analysis of outcome from this study of 120 patients confirmed that those with grade 3 histology as well as GBM patients who were treated 6 months after completing prior 5-day temozolomide therapy had PFS-6 rates of 38 and 28%, respectively, while those who were treated with daily temozolomide within 6 months of 5-day temozolomide dosing had a PFS-6 of only 9.5% [41].

In general, our study results affirm that daily temozolomide in combination with sorafenib has an acceptable toxicity profile. This finding is particularly relevant in that several ongoing studies are combing sorafenib with temozolomide for newly diagnosed GBM patients. Other studies have confirmed that sorafenib can be safely administered with a variety of chemotherapy agents for other malignancies [4247]. Seven patients (22%) in the current study required dose modification or discontinuation of sorafenib due to adverse events and none of the patients required either dose modification or discontinuation of daily temozolomide. Elevated amylase and/or lipase has been reported with sorafenib and other VEGFR inhibitors. In our series, only two of the four patients who developed this complication were symptomatic and in one of these two patients, underlying gallstones were the likely etiology. Palmar-plantar erythrodysesthesia (PPE) was noted in seven patients (22%) but was manageable with brief sorafenib dose interruptions and supportive care in most patients.

There are several possible factors that may have contributed to the disappointing outcome observed on this study. First, in general patients on this study were heavily pre-treated. Thirteen patients (41%) had received at least 4 prior chemotherapy or investigational agents and 15 patients (47%) had progressed on prior 5-day temozolomide. Such patients have been shown to have a low response to daily temozolomide [17, 41]. Although the combination of temozolomide plus sorafenib showed encouraging activity among advanced melanoma patients without prior temozolomide treatment, the regimen showed minimal activity previously treated with temozolomide in a recently reported phase II study [48]. Second, 13 patients (41%) had previously failed either bevacizumab or an alternative VEGFR-2-targeting TKI. Recent data demonstrates that progressive disease on bevacizumab confers a particularly poor prognosis with a low likelihood of response to subsequent salvage therapy [49, 50]. Another possible contributing factor to the poor outcome of our study was that patients were unselected for sorafenib target expression. Data from at least one study suggests that there may be subsets of patients who are more likely to respond to sorafenib therapy. Specifically, melanoma patients with high VEGF-R2 expression were more likely to respond to sorafenib plus chemotherapy, whereas those with elevated ERK1/2 levels did not respond [47].

Finally, our PK data suggest that EIAEDs, which were concurrently administered to 12 (38%) of our study patients, may have significantly induced sorafenib metabolism by day 28, leading to overall lower exposures and potentially poorer anti-tumor efficacy. Prior pharmacokinetic studies reveal that sorafenib metabolism exhibits significant interpatient variability [51], and that exposures increase by 3–4 fold over seven days, but reach steady-state at that point [52]. In vitro microsomal data indicate that sorafenib metabolism is mediated by phase I oxidation through cytochrome P450 (CYP) 3A4 and phase II conjugation via UGT1A9 [53]. Although co-administration of ketoconazole, a known CYP3A inhibitor, with low-dose sorafenib (50 mg/day) did not result in unexpected adverse events or evidence of altered sorafenib pharmacokinetics, presumably due to increased glucuronide elimination [53], the effect of CYP3A inducers on sorafenib metabolism are less clear. Recently reported data shows that co-administration of rifampin, a CYP3A4 and UGT 1A9 inducer, led to 37% reduction in sorafenib exposure [54]. In a recent phase I dose escalation study among malignant glioma patients, sorafenib exposures were comparable between patients on and not on EIAEDs on day one of cycle one [36]. Our study results support this finding. However, the phase I study did not evaluate later time point measures. Our study therefore provides the only source of data of steady-state pharmacokinetic measures of sorafenib among brain tumor patients. We demonstrate that sorafenib exposures differ significantly by day 28 between patients on and not on EIAEDs. Specifically, exposures significantly increased by 3–4 fold among patients not on EIAEDs by day 28 as expected based on time required to achieve steady state exposures as previously documented [52]. However, day 28 exposure did not increase compared to day 1 for patients on EIAEDs, presumably due to CYP3A induction. Alternatively, it is possible that continued sorafenib dosing may have led to auto-induction of metabolism. However, this effect has not been observed in other studies [52, 55], and should have been equally apparent regardless of EIAED status in the current study. Diminished exposures of several anti-tumor agents due to EIAEDs have been observed among brain tumor patients including irinotecan [5658], imatinib [5961] and erlotinib [62]. However, such an effect has not been previously recognized with sorafenib in this patient population, and is particularly relevant as several ongoing studies of sorafenib in both newly diagnosed and recurrent malignant glioma patients are employing uniform dosing regardless of EIAED status. Our study data suggests that patients on EIAEDs may be significantly underdosed relative to patients not on EIAEDs and that additional studies should be performed to determine optimal sorafenib dosing among patients on EIAEDs. In conclusion, our study demonstrates that the combination of sorafenib plus daily temozolomide is not active among unselected, recurrent GBM patients. Ongoing and future studies among malignant glioma patients should further define the effect of EIAEDs on sorafenib pharmacokinetics and also attempt to determine whether response to sorafenib-based therapy may be linked to tumor markers.

Acknowledgments

This work was supported by NIH Grants NS20023 and CA11898; NIH Grant MO1 RR 30, GCRC Program, NCRR; and NCI SPORE 1 P20 CA096890.

Abbreviations

CR

Complete response

EIAEDs

Enzyme-inducing antieptileptic drugs

GBM

Glioblastoma multiforme

ITT

Intent-to treat

KPS

Karnofsky performance status

MAPK

Mitogen-activated protein kinase

OS

Overall survival

PD

Progressive disease

PFS

Progression-free survival

PR

Partial response

SD

Stable disease

VEGF

Vascular endothelial growth factor

VEGFR

Vascular endothelial growth factor receptor

Contributor Information

David A. Reardon, Email: reard003@mc.duke.edu, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA; The Brain Tumor Center at Duke, Duke University Medical Center, Box 3624, Durham, NC 27710, USA.

James J. Vredenburgh, Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA

Annick Desjardins, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA.

Katherine Peters, Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA.

Sridharan Gururangan, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA.

John H. Sampson, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA

Jennifer Marcello, Department of Cancer Center Biostatistics, Duke University Medical Center, Durham, NC 27710, USA.

James E. Herndon, II, Department of Cancer Center Biostatistics, Duke University Medical Center, Durham, NC 27710, USA.

Roger E. McLendon, Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA

Dorothea Janney, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.

Allan H. Friedman, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA

Darell D. Bigner, Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA

Henry S. Friedman, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA

References

  • 1.Wong ET, Hess KR, Gleason MJ, Jaeckle KA, Kyritsis AP, Prados MD, Levin VA, Yung WK. Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol. 1999;17:2572–2578. doi: 10.1200/JCO.1999.17.8.2572. [DOI] [PubMed] [Google Scholar]
  • 2.Lamborn KR, Yung WK, Chang SM, Wen PY, Cloughesy TF, Deangelis LM, Robins HI, Lieberman FS, Fine HA, Fink KL, Junck L, Abrey L, Gilbert MR, Mehta M, Kuhn JG, Aldape KD, Hibberts J, Peterson PM, Prados MD. Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro-oncology. 2008;10:162–170. doi: 10.1215/15228517-2007-062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Ballman KV, Buckner JC, Brown PD, Giannini C, Flynn PJ, LaPlant BR, Jaeckle KA. The relationship between six-month progression-free survival and 12-month overall survival end points for phase II trials in patients with glioblastoma multiforme. Neuro-oncology. 2007;9:29–38. doi: 10.1215/15228517-2006-025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Vredenburgh JJ, Desjardins A, Herndon JE, II, Dowell JM, Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan S, Wagner M, Bigner DD, Friedman AH, Friedman HS. Phase II trial of bevacizumab and irinotecan in recurrent malignant glioma. Clin Cancer Res. 2007;13:1253–1259. doi: 10.1158/1078-0432.CCR-06-2309. [DOI] [PubMed] [Google Scholar]
  • 5.Vredenburgh JJ, Desjardins A, Herndon JE, II, Marcello J, Reardon DA, Quinn JA, Rich JN, Sathornsumetee S, Gururangan S, Sampson J, Wagner M, Bailey L, Bigner DD, Friedman AH, Friedman HS. Bevacizumab plus irinotecan in recurrent glioblastoma multiforme. J Clin Oncol. 2007;25:4722–4729. doi: 10.1200/JCO.2007.12.2440. [DOI] [PubMed] [Google Scholar]
  • 6.Desjardins A, Reardon DA, Herndon JE, II, Marcello J, Quinn JA, Rich JN, Sathornsumetee S, Gururangan S, Sampson J, Bailey L, Bigner DD, Friedman AH, Friedman HS, Vredenburgh JJ. Bevacizumab plus irinotecan in recurrent WHO grade 3 malignant gliomas. Clin Cancer Res. 2008;14:7068–7073. doi: 10.1158/1078-0432.CCR-08-0260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Friedman HS, Prados MD, Wen PY, Mikkelsen T, Schiff D, Abrey LE, Yung WK, Paleologos N, Nicholas MK, Jensen R, Vredenburgh J, Huang J, Zheng M, Cloughesy T. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27:4733–4740. doi: 10.1200/JCO.2008.19.8721. [DOI] [PubMed] [Google Scholar]
  • 8.Kreisl TN, Kim L, Moore K, Duic P, Royce C, Stroud I, Garren N, Mackey M, Butman JA, Camphausen K, Park J, Albert PS, Fine HA. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol. 2009;27:740–745. doi: 10.1200/JCO.2008.16.3055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Batchelor TT, Sorensen AG, di Tomaso E, Zhang WT, Duda DG, Cohen KS, Kozak KR, Cahill DP, Chen PJ, Zhu M, Ancukiewicz M, Mrugala MM, Plotkin S, Drappatz J, Louis DN, Ivy P, Scadden DT, Benner T, Loeffler JS, Wen PY, Jain RK. AZD2171, a Pan-VEGF receptor tyrosine kinase inhibitor, normalizes tumor vasculature and alleviates edema in glioblastoma patients. Cancer Cell. 2007;11:83–95. doi: 10.1016/j.ccr.2006.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, Schwartz B, Simantov R, Kelley S. Discovery and development of sorafenib: a multikinase inhibitor for treating cancer. Nat Rev Drug Discov. 2006;5:835–844. doi: 10.1038/nrd2130. [DOI] [PubMed] [Google Scholar]
  • 11.Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E, Desai AA, Rolland F, Demkow T, Hutson TE, Gore M, Freeman S, Schwartz B, Shan M, Simantov R, Bukowski RM. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125–134. doi: 10.1056/NEJMoa060655. [DOI] [PubMed] [Google Scholar]
  • 12.Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, de Oliveira AC, Santoro A, Raoul JL, Forner A, Schwartz M, Porta C, Zeuzem S, Bolondi L, Greten TF, Galle PR, Seitz JF, Borbath I, Haussinger D, Giannaris T, Shan M, Moscovici M, Voliotis D, Bruix J. Sorafenib in advanced hepatocellular carcinoma. N Engl J Med. 2008;359:378–390. doi: 10.1056/NEJMoa0708857. [DOI] [PubMed] [Google Scholar]
  • 13.Denny BJ, Wheelhouse RT, Stevens MF, Tsang LL, Slack JA. NMR and molecular modeling investigation of the mechanism of activation of the antitumor drug temozolomide and its interaction with DNA. Biochemistry. 1994;33:9045–9051. doi: 10.1021/bi00197a003. [DOI] [PubMed] [Google Scholar]
  • 14.Newlands ES, Stevens MF, Wedge SR, Wheelhouse RT, Brock C. Temozolomide: a review of its discovery, chemical properties, pre-clinical development and clinical trials. Cancer Treat Rev. 1997;23:35–61. doi: 10.1016/s0305-7372(97)90019-0. [DOI] [PubMed] [Google Scholar]
  • 15.Brock CS, Newlands ES, Wedge SR, Bower M, Evans H, Colquhoun I, Roddie M, Glaser M, Brampton MH, Rustin GJ. Phase I trial of temozolomide using an extended continuous oral schedule. Cancer Res. 1998;58:4363–4367. [PubMed] [Google Scholar]
  • 16.Wick A, Felsberg J, Steinbach JP, Herrlinger U, Platten M, Blaschke B, Meyermann R, Reifenberger G, Weller M, Wick W. Efficacy and tolerability of temozolomide in an alternating weekly regimen in patients with recurrent glioma. J Clin Oncol. 2007;25:3357–3361. doi: 10.1200/JCO.2007.10.7722. [DOI] [PubMed] [Google Scholar]
  • 17.Perry JR, Rizek P, Cashman R, Morrison M, Morrison T. Temozolomide rechallenge in recurrent malignant glioma by using a continuous temozolomide schedule: the “rescue” approach. Cancer. 2008;113:2152–2157. doi: 10.1002/cncr.23813. [DOI] [PubMed] [Google Scholar]
  • 18.Neyns B, Chaskis C, Joosens E, Menten J, D'Hondt L, Branle F, Sadones J, Michotte A. A multicenter cohort study of dose-dense temozolomide (21 of 28 days) for the treatment of recurrent anaplastic astrocytoma or oligoastrocytoma. Cancer Invest. 2008;26:269–277. doi: 10.1080/07357900701708393. [DOI] [PubMed] [Google Scholar]
  • 19.Brandes AA, Tosoni A, Cavallo G, Bertorelle R, Gioia V, Franceschi E, Biscuola M, Blatt V, Crino L, Ermani M. Temozolomide 3 weeks on and 1 week off as first-line therapy for recurrent glioblastoma: phase II study from gruppo italiano cooperativo di neuro-oncologia (GICNO) Br J Cancer. 2006;95:1155–1160. doi: 10.1038/sj.bjc.6603376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Balmaceda C, Peereboom D, Pannullo S, Cheung YK, Fisher PG, Alavi J, Sisti M, Chen J, Fine RL. Multi-institutional phase II study of temozolomide administered twice daily in the treatment of recurrent high-grade gliomas. Cancer. 2008;112:1139–1146. doi: 10.1002/cncr.23167. [DOI] [PubMed] [Google Scholar]
  • 21.Wick W, Platten M, Weller M. New (alternative) temozolomide regimens for the treatment of glioma. Neuro-oncology. 2009;11:69–79. doi: 10.1215/15228517-2008-078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Spence AM, Peterson RA, Scharnhorst JD, Silbergeld DL, Rostomily RC. Phase II study of concurrent continuous Temozolomide (TMZ) and Tamoxifen (TMX) for recurrent malignant astrocytic gliomas. J Neurooncol. 2004;70:91–95. doi: 10.1023/b:neon.0000040837.68411.97. [DOI] [PubMed] [Google Scholar]
  • 23.Khan RB, Raizer JJ, Malkin MG, Bazylewicz KA, Abrey LE. A phase II study of extended low-dose temozolomide in recurrent malignant gliomas. Neuro-oncology. 2002;4:39–43. doi: 10.1215/15228517-4-1-39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kong DS, Lee JI, Kim WS, Son MJ, Lim do H, Kim ST, Park K, Kim JH, Eoh W, Nam DH. A pilot study of metronomic temozolomide treatment in patients with recurrent temozolomide-refractory glioblastoma. Oncol Rep. 2006;16:1117–1121. [PubMed] [Google Scholar]
  • 25.Berrocal A, Perez Segura P, Gil M, Balana C, Garcia Lopez J, Yaya R, Rodriguez J, Reynes G, Gallego O, Iglesias L. Extended-schedule dose-dense temozolomide in refractory gliomas. J Neurooncol. 2010;96(3):417–422. doi: 10.1007/s11060-009-9980-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Macdonald DR, Cascino TL, Schold SC, Jr, Cairncross JG. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8:1277–1280. doi: 10.1200/JCO.1990.8.7.1277. [DOI] [PubMed] [Google Scholar]
  • 27.Yung WK, Prados MD, Yaya-Tur R, Rosenfeld SS, Brada M, Friedman HS, Albright R, Olson J, Chang SM, O'Neill AM, Friedman AH, Bruner J, Yue N, Dugan M, Zaknoen S, Levin VA. Multicenter phase II trial of temozolomide in patients with anaplastic astrocytoma or anaplastic oligoastrocytoma at first relapse. Temodal Brain Tumor Group. J Clin Oncol. 1999;17:2762–2771. doi: 10.1200/JCO.1999.17.9.2762. [DOI] [PubMed] [Google Scholar]
  • 28.Wilhelm SM, Adnane L, Newell P, Villanueva A, Llovet JM, Lynch M. Preclinical overview of sorafenib, a multikinase inhibitor that targets both Raf and VEGF and PDGF receptor tyrosine kinase signaling. Mol Cancer Ther. 2008;7:3129–3140. doi: 10.1158/1535-7163.MCT-08-0013. [DOI] [PubMed] [Google Scholar]
  • 29.Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA. Mutations of the BRAF gene in human cancer. Nature. 2002;417:949–954. doi: 10.1038/nature00766. [DOI] [PubMed] [Google Scholar]
  • 30.Pelloski CE, Lin E, Zhang L, Yung WK, Colman H, Liu JL, Woo SY, Heimberger AB, Suki D, Prados M, Chang S, Barker FG, III, Fuller GN, Aldape KD. Prognostic associations of activated mitogen-activated protein kinase and Akt pathways in glioblastoma. Clin Cancer Res. 2006;12:3935–3941. doi: 10.1158/1078-0432.CCR-05-2202. [DOI] [PubMed] [Google Scholar]
  • 31.Martinho O, Longatto-Filho A, Lambros MB, Martins A, Pinheiro C, Silva A, Pardal F, Amorim J, Mackay A, Milanezi F, Tamber N, Fenwick K, Ashworth A, Reis-Filho JS, Lopes JM, Reis RM. Expression, mutation and copy number analysis of platelet-derived growth factor receptor A (PDGFRA) and its ligand PDGFA in gliomas. Br J Cancer. 2009;101:973–982. doi: 10.1038/sj.bjc.6605225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hermanson M, Funa K, Hartman M, Claesson-Welsh L, Heldin CH, Westermark B, Nister M. Platelet-derived growth factor and its receptors in human glioma tissue: expression of messenger RNA and protein suggests the presence of autocrine and paracrine loops. Cancer Res. 1992;52:3213–3219. [PubMed] [Google Scholar]
  • 33.Shih AH, Dai C, Hu X, Rosenblum MK, Koutcher JA, Holland EC. Dose-dependent effects of platelet-derived growth factor-B on glial tumorigenesis. Cancer Res. 2004;64:4783–4789. doi: 10.1158/0008-5472.CAN-03-3831. [DOI] [PubMed] [Google Scholar]
  • 34.Joensuu H, Puputti M, Sihto H, Tynninen O, Nupponen NN. Amplification of genes encoding KIT, PDGFRalpha and VEGFR2 receptor tyrosine kinases is frequent in glioblastoma multiforme. J Pathol. 2005;207:224–231. doi: 10.1002/path.1823. [DOI] [PubMed] [Google Scholar]
  • 35.Puputti M, Tynninen O, Sihto H, Blom T, Maenpaa H, Isola J, Paetau A, Joensuu H, Nupponen NN. Amplification of KIT, PDGFRA, VEGFR2, and EGFR in gliomas. Mol Cancer Res. 2006;4:927–934. doi: 10.1158/1541-7786.MCR-06-0085. [DOI] [PubMed] [Google Scholar]
  • 36.Nabors LB, Rosenfeld MG, Chamberlain SC, Phuphanich S, Batchelor T, Supko JG, Desideri S, Xiaobu Y, Wright J, Grossman S. A Phase I trial of sorafenib (BAY 743-9006) for patients with recurrent or progressive malignant glioma (NABTT 0401). 2007 ASCO Annual Meeting Proceedings; Chicago, IL. 2007. p. 89. [Google Scholar]
  • 37.Prados M, Gilbert M, Kuhn K, Lamborn K, Cloughesy T, Lieberman F, Puduvalli VK, Robins HI, Lassman A, Wen PY. Phase I/II study of sorefenib and erlotinib for patients with recurrent glioblastoma (GBM)(NABTC 05-02) American Society of Clinical Oncology; Orlando, FL: 2009. p. 88. [Google Scholar]
  • 38.Wen PY, Cloughesy T, Kuhn J, Lamborn K, Abrey LE, Lieberman F, Robins HI, Wright J, Prados MD, Gilbert M. Phase I/II study of sorafenib and temsirolimus for patients with recurrent glioblastoma (GBM)(NABTC 05-02) American Society of Clinical Oncology; Orlando, FL: 2009. p. 88. [Google Scholar]
  • 39.Tolcher AW, Gerson SL, Denis L, Geyer C, Hammond LA, Patnaik A, Goetz AD, Schwartz G, Edwards T, Reyderman L, Statkevich P, Cutler DL, Rowinsky EK. Marked inactivation of O6-alkylguanine-DNA alkyltransferase activity with protracted temozolomide schedules. Br J Cancer. 2003;88:1004–1011. doi: 10.1038/sj.bjc.6600827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Tosoni A, Cavallo G, Ermani M, Scopece L, Franceschi E, Ghimenton C, Gardiman M, Pasetto L, Blatt V, Brandes AA. Is protracted low-dose temozolomide feasible in glioma patients? Neurology. 2006;66:427–429. doi: 10.1212/01.wnl.0000196465.83423.ec. [DOI] [PubMed] [Google Scholar]
  • 41.Perry JR, Belanger K, Mason WP, Fulton D, Kavan P, Easaw J, Shields C, Kirby S, Macdonald DR, Eisenstat DD, Thiessen B, Forsyth P, Pouliot JF. A phase II trial of continuous dose-intense temozolomide in recurrent malignant glioma. RESCUE study. J Clin Oncol. 2010;28(12):2051–2057. doi: 10.1200/JCO.2009.26.5520. [DOI] [PubMed] [Google Scholar]
  • 42.Siu LL, Awada A, Takimoto CH, Piccart M, Schwartz B, Giannaris T, Lathia C, Petrenciuc O, Moore MJ. Phase I trial of sorafenib and gemcitabine in advanced solid tumors with an expanded cohort in advanced pancreatic cancer. Clin Cancer Res. 2006;12:144–151. doi: 10.1158/1078-0432.CCR-05-1571. [DOI] [PubMed] [Google Scholar]
  • 43.Flaherty KT, Brose M, Schuchter L, et al. Phase I/II trial of BAY 43-9006, carboplatin and paclitaxel demonstrated preliminary antitumor activity in the expansion cohort of patients with metastatic melanoma. J Clin Oncol. 2004:711. [Google Scholar]
  • 44.Richly H, Kupsch P, Passage K, Grubert M, Hilger RA, Voigtmann R, Schwartz B, Brendel E, Christensen O, Haase CG, Strumberg D. Results of a phase I trial of BAY 43-9006 in combination with doxorubicin in patients with primary hepatic cancer. Int J Clin Pharmacol Ther. 2004;42:650–651. doi: 10.5414/cpp42650. [DOI] [PubMed] [Google Scholar]
  • 45.Kupsch P, Henning BF, Passarge K, Richly H, Wiesemann K, Hilger RA, Scheulen ME, Christensen O, Brendel E, Schwartz B, Hofstra E, Voigtmann R, Seeber S, Strumberg D. Results of a phase I trial of sorafenib (BAY 43-9006) in combination with oxaliplatin in patients with refractory solid tumors, including colorectal cancer. Clin Colorectal Cancer. 2005;5:188–196. doi: 10.3816/ccc.2005.n.030. [DOI] [PubMed] [Google Scholar]
  • 46.Hauschild A, Agarwala SS, Trefzer U, Hogg D, Robert C, Hersey P, Eggermont A, Grabbe S, Gonzalez R, Gille J, Peschel C, Schadendorf D, Garbe C, O'Day S, Daud A, White JM, Xia C, Patel K, Kirkwood JM, Keilholz U. Results of a phase III, randomized, placebo-controlled study of sorafenib in combination with carboplatin and paclitaxel as second-line treatment in patients with unresectable stage III or stage IV melanoma. J Clin Oncol. 2009;27:2823–2830. doi: 10.1200/JCO.2007.15.7636. [DOI] [PubMed] [Google Scholar]
  • 47.Jilaveanu L, Zito C, Lee SJ, Nathanson KL, Camp RL, Rimm DL, Flaherty KT, Kluger HM. Expression of sorafenib targets in melanoma patients treated with carboplatin, paclitaxel and sorafenib. Clin Cancer Res. 2009;15:1076–1085. doi: 10.1158/1078-0432.CCR-08-2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Amaravadi RK, Schuchter LM, McDermott DF, Kramer A, Giles L, Gramlich K, Carberry M, Troxel AB, Letrero R, Nathanson KL, Atkins MB, O'Dwyer PJ, Flaherty KT. Phase II trial of temozolomide and sorafenib in advanced melanoma patients with or without brain metastases. Clin Cancer Res. 2009;15:7711–7718. doi: 10.1158/1078-0432.CCR-09-2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Norden AD, Young GS, Setayesh K, Muzikansky A, Klufas R, Ross GL, Ciampa AS, Ebbeling LG, Levy B, Drappatz J, Kesari S, Wen PY. Bevacizumab for recurrent malignant gliomas: efficacy, toxicity, and patterns of recurrence. Neurology. 2008;70:779–787. doi: 10.1212/01.wnl.0000304121.57857.38. [DOI] [PubMed] [Google Scholar]
  • 50.Quant EC, Norden AD, Drappatz J, Muzikansky A, Doherty L, Lafrankie D, Ciampa A, Kesari S, Wen PY. Role of a second chemotherapy in recurrent malignant glioma patients who progress on bevacizumab. Neuro-oncology. 2009;11:550–555. doi: 10.1215/15228517-2009-006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Clark JW, Eder JP, Ryan D, Lathia C, Lenz HJ. Safety and pharmacokinetics of the dual action Raf kinase and vascular endothelial growth factor receptor inhibitor, BAY 43-9006, in patients with advanced, refractory solid tumors. Clin Cancer Res. 2005;11:5472–5480. doi: 10.1158/1078-0432.CCR-04-2658. [DOI] [PubMed] [Google Scholar]
  • 52.Awada A, Hendlisz A, Gil T, Bartholomeus S, Mano M, de Valeriola D, Strumberg D, Brendel E, Haase CG, Schwartz B, Piccart M. Phase I safety and pharmacokinetics of BAY 43-9006 administered for 21 days on/7 days off in patients with advanced, refractory solid tumours. Br J Cancer. 2005;92:1855–1861. doi: 10.1038/sj.bjc.6602584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Lathia C, Lettieri J, Cihon F, Gallentine M, Radtke M, Sundaresan P. Lack of effect of ketoconazole-mediated CYP3A inhibition on sorafenib clinical pharmacokinetics. Cancer Chemother Pharmacol. 2006;57:685–692. doi: 10.1007/s00280-005-0068-6. [DOI] [PubMed] [Google Scholar]
  • 54.Keating GM, Santoro A. Sorafenib: a review of its use in advanced hepatocellular carcinoma. Drugs. 2009;69:223–240. doi: 10.2165/00003495-200969020-00006. [DOI] [PubMed] [Google Scholar]
  • 55.Moore M, Hirte HW, Siu L, Oza A, Hotte SJ, Petrenciuc O, Cihon F, Lathia C, Schwartz B. Phase I study to determine the safety and pharmacokinetics of the novel Raf kinase and VEGFR inhibitor BAY 43-9006, administered for 28 days on/7 days off in patients with advanced, refractory solid tumors. Ann Oncol. 2005;16:1688–1694. doi: 10.1093/annonc/mdi310. [DOI] [PubMed] [Google Scholar]
  • 56.Gilbert MR, Supko JG, Batchelor T, Lesser G, Fisher JD, Piantadosi S, Grossman S. Phase I clinical and pharmacokinetic study of irinotecan in adults with recurrent malignant glioma. Clin Cancer Res. 2003;9:2940–2949. [PubMed] [Google Scholar]
  • 57.Prados MD, Yung WK, Jaeckle KA, Robins HI, Mehta MP, Fine HA, Wen PY, Cloughesy TF, Chang SM, Nicholas MK, Schiff D, Greenberg HS, Junck L, Fink KL, Hess KR, Kuhn J. Phase 1 trial of irinotecan (CPT-11) in patients with recurrent malignant glioma: a North American Brain Tumor Consortium study. Neuro-oncology. 2004;6:44–54. doi: 10.1215/S1152851703000292. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Santisteban M, Buckner JC, Reid JM, Wu W, Scheithauer BW, Ames MM, Felten SJ, Nikcevich DA, Wiesenfeld M, Jaeckle KA, Galanis E North Central Cancer Treatment Group. Phase II trial of two different irinotecan schedules with pharmacokinetic analysis in patients with recurrent glioma: North Central Cancer Treatment Group results. J Neurooncol. 2009;92(2):165–175. doi: 10.1007/s11060-008-9749-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Raymond E, Brandes AA, Dittrich C, Fumoleau P, Coudert B, Clement PM, Frenay M, Rampling R, Stupp R, Kros JM, Heinrich MC, Gorlia T, Lacombe D, van den Bent MJ. Phase II study of imatinib in patients with recurrent gliomas of various histologies: a European Organisation for Research and Treatment of Cancer Brain Tumor Group Study. J Clin Oncol. 2008;26:4659–4665. doi: 10.1200/JCO.2008.16.9235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Reardon DA, Egorin MJ, Quinn JA, Rich JN, Sr, Gururangan I, Vredenburgh JJ, Desjardins A, Sathornsumetee S, Provenzale JM, Herndon JE, II, Dowell JM, Badruddoja MA, McLendon RE, Lagattuta TF, Kicielinski KP, Dresemann G, Sampson JH, Friedman AH, Salvado AJ, Friedman HS. Phase II study of imatinib mesylate plus hydroxyurea in adults with recurrent glioblastoma multiforme. J Clin Oncol. 2005;23:9359–9368. doi: 10.1200/JCO.2005.03.2185. [DOI] [PubMed] [Google Scholar]
  • 61.Wen PY, Yung WK, Lamborn KR, Dahia PL, Wang Y, Peng B, Abrey LE, Raizer J, Cloughesy TF, Fink K, Gilbert M, Chang S, Junck L, Schiff D, Lieberman F, Fine HA, Mehta M, Robins HI, DeAngelis LM, Groves MD, Puduvalli VK, Levin V, Conrad C, Maher EA, Aldape K, Hayes M, Letvak L, Egorin MJ, Capdeville R, Kaplan R, Murgo AJ, Stiles C, Prados MD. Phase I/II study of imatinib mesylate for recurrent malignant gliomas: North American Brain Tumor Consortium Study 99–08. Clin Cancer Res. 2006;12:4899–4907. doi: 10.1158/1078-0432.CCR-06-0773. [DOI] [PubMed] [Google Scholar]
  • 62.Prados MD, Lamborn KR, Chang S, Burton E, Butowski N, Malec M, Kapadia A, Rabbitt J, Page MS, Fedoroff A, Xie D, Kelley SK. Phase 1 study of erlotinib HCl alone and combined with temozolomide in patients with stable or recurrent malignant glioma. Neuro-oncology. 2006;8:67–78. doi: 10.1215/S1522851705000451. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES