Skip to main content
Top

07-10-2017 | Glioblastoma multiforme | Article

Phase I study of sorafenib and tipifarnib for recurrent glioblastoma: NABTC 05-02

Journal: Journal of Neuro-Oncology

Authors: Phioanh Leia Nghiemphu, Victoria Asuquo Ebiana, Patrick Wen, Mark Gilbert, Lauren E. Abrey, F. Lieberman, Lisa M. DeAngelis, H. Ian Robins, W. K. Alfred Yung, Susan Chang, Jan Drappatz, Minesh P. Mehta, Victor A. Levin, Kenneth Aldape, Janet E. Dancey, J. J. Wright, Michael Prados, John Kuhn, Timothy F. Cloughesy

Publisher: Springer US

Abstract

Recurrent glioblastoma (GBM) has a very low 6-month progression free survival (PFS) with currently available treatments. Combination chemotherapy to target multiple cell signaling pathways is currently being investigated in order to improve prognosis for recurrent disease. The purpose of this phase I study was to determine the maximum tolerated dose (MTD) for the combination of tipifarnib and sorafenib for the treatment of recurrent GBM. Patients with pathologically proven WHO grade IV GBM and radiographically proven tumor recurrence were eligible for this study. Treatments included sorafenib at twice daily and escalating dosages of tipifarnib. Dose-limiting toxicity (DLT) was determined over the first 28-days of treatments, and the MTD was determined in a 3 + 3 study design. We enrolled 24 patients, and 21 patients completed the MTD period. The study was stopped early with no MTD determination for excessive toxicities. The last dose level reached was sorafenib at 200 mg twice a day and tipifarnib 100 mg twice a day on an alternating week schedule. The DLTs included diarrhea, lipase elevation, hypophosphatemia, and arthralgia. The combination of sorafenib and tipifarnib has excessive toxicities and full single agent dosages could not be achieved in combination.

Introduction

Primary malignant central nervous system tumors cause more than 13,000 deaths per year in the United States [1]. The most common as well as the most aggressive primary brain tumor is glioblastoma (GBM). Despite optimal treatment with surgery, radiation therapy and temozolomide chemotherapy, almost 90% of patients with GBM will have tumor progression by 2 years [2]. Second line treatments with either chemotherapies or biological agents usually only achieve a 6-months progression free survival (PFS) in 15–16% of patients with GBM [3, 4], and slightly more, 29–45%, if treated with bevacizumab [5, 6]. Thus, more effective treatments at recurrence are needed.
Several pathways are implicated in the pathogenesis of GBM including multiple abnormalities in receptor tyrosine kinase pathways. For instance, the epidermal growth factor receptor is amplified in up to 70% of GBMs [7]. Mitogen binding of these receptors leads to activation of signal transduction cascades that include activation of the Ras genes [8]. Activated Ras further triggers the kinase activity of the protein kinase Raf and subsequently the mitogen-activated protein kinase (MAPK) pathway which are key controls in cell growth, proliferation, and survival [9]. Furthermore, overexpression of the Ras oncogene is also found in a large proportion of human cancers and Ras genes play a role in cell proliferation and differentiation [10].
Several clinical trials for the treatment of GBM have used agents targeting the RAS-MAPK pathway. One strategy attempts to use farnesyltransferase inhibitors (FTIs) to block the post-translational activation of RAS, including trials using the FTI tipifarnib. Tipifarnib (R115777, Zarnestra; Johnson & Johnson Pharmaceutical Research & Development LLC, Titusville, NJ) is a potent and selective nonpeptidomimetic FTI. Additional effects of this agent include inhibition of proliferation in tumors both with and without Ras mutations as well as effects on angiogenesis, apoptosis, and tumor microenvironment [1113]. The action of FTIs have been demonstrated in several pre-clinical studies to sensitize tumors to radiotherapy [11, 14] and inhibit glioma cell proliferation [15, 16].
In phase I studies, tipifarnib exhibits oral bioavailability with dose-proportional pharmacokinetics [17, 18]. Several trials in recurrent glioma found that tipifarnib has a toxicity profile and efficacy that depends on the types of antiepileptic drugs being taken by patients [19, 20], where patients taking enzyme inducing antiepileptic drugs (EIAEDs) would have different maximum tolerated dose (MTD) and type of dose limiting toxicity (DLT) than patients not taking EIAEDs [21]. These studies found that in patients taking EIAED, the MTD was 600 mg bid for 21 days every 4 weeks, double the MTD for patients not receiving EIAED. Also, the predominant DLT seen in the patients on EIAEDs was rash versus the myelosuppression seen in those patients not taking EIAEDs. Pharmacokinetic evaluation showed that the area under the plasma concentration–time curve (AUC) from 0 to 12 h at MTD was approximately halved in those receiving EIAEDs compared with those not receiving EIAEDs. Although limited pharmacodynamic evaluation revealed that at MTD, patients receiving EIAEDs had adequate inhibition of farnesylation in peripheral blood mononuclear cells, a later phase II clinical trial found that at MTD, patients not on EIAED had possibly better progression free survival (PFS) than those on EIAED, and the survival data showed limited efficacy in both these cohorts [20] (Fig. 1).
Without clinically significant benefits as a single agent, tipifarnib may have better efficacy when combined with other cytotoxic therapies or complementary targeted molecular agents. Blocking the RAS pathway further downstream with a Raf kinase inhibitor may increase inhibition of tumor growth in preclinical models [22], e.g. blockage of RAF may inhibit further activation down the MAPK pathway.
Sorafenib (BAY 43-9006, Nexavar; Bayer) is a novel bi-aryl urea that has been previously proven to inhibit RAF1 and b-RAF kinase family members. In addition, sorafenib demonstrated potent inhibition of several receptor tyrosine kinases involved in angiogenesis including VEGFR-2, PDGFR-β and VEGFR-3 [2224]. Sorafenib as a single agent has been evaluated in several Phase I studies in patients with advanced refractory solid tumors [25, 26] in various dosages and schedules, both intermittent and continuous. The most common drug-related toxicities have involved the gastro-intestinal tract (diarrhea, nausea, abdominal cramps) and skin (pruritus, rash and hand-foot syndrome). Other reported treatment-related adverse events were hepatic disorders (abnormal AST, ALT, bilirubin, and GGT) and elevation of pancreatic enzymes. Data from Phase I trials indicate that 400 mg BID is the MTD for sorafenib.
Since combination of an FTI inhibitor and a Raf inhibitor to block both upstream and downstream RAS/Raf/Mek pathway may better block tumor growth and improve survival for patients with GBM, we conducted a phase I clinical trial of sorafenib in combination with tipifarnib to determine the maximum tolerated dosages (MTD) for this combination. This study is part of the North American Brain Tumor Consortium study 05-02, which was a multi-arms study of different combinations of target inhibition to achieve better receptor tyrosine kinase inhibition, with sorafenib as the backbone (stable dose and agent in all arms) in combination with upstream RAS inhibition (tipifarnib, reported here) or with epidermal growh factor receptor inhibition (erlotinib) or PI-3-kinase pathway inhibition (temsirolimus). The primary objective of this study included finding the best phase I combination with the least toxicity and possibly some preliminary response to move on to a phase II efficacy study. This manuscript reports the results from the attempt to determine MTD of the combination of sorafenib and tipifarnib in patients with recurrent malignant glioma. The reports for the other arms of sorafenib with erlotinib or temsirolimus have been reported previously [27, 28].

Patients and methods

Patient population

Eligible patients were ≥ 18 years of age with recurrent histologically confirmed WHO grade IV GBM or gliosarcoma. Patients must have unequivocal radiographic evidence of disease recurrence on either MRI or CT. A baseline MRI was obtained within 14 days of treatment on no or stable steroid dosage of at least 5 days. There were no restrictions on the number of recurrences or treatments, but patients must have progressed following prior radiotherapy and be at least 42 days from completion of radiotherapy. Eligibility criteria included Karnofsky performance score (KPS) ≥ 60 and adequate hematologic and organ function. Patients were excluded if they had received prior sorafenib, vatalanib, AEE-788, or any farnesyl transferase inhibitors, were receiving EIADs, had grade 2 or higher peripheral neuropathy, had histories of allergy to imidazoles, or were pregnant or breastfeeding. Patients were also excluded if they had significant medical illness or any disease that may obscure toxicity or dangerously alter drug metabolism. All patients with child bearing potential were required to use adequate contraception. The protocol and informed consent were approved by the institutional review boards of all participating institutions. All patients reviewed, signed, and provided written informed consent before enrollment.

Study design

This study was a part of a phase I sequential accrual design trial with three different combination therapy arms: Arm 1—sorafenib and erlotinib; Arm 2—sorafenib and temsirolimus; Arm 3—sorafenib and tipifarnib. Cohorts of 3 patients were enrolled starting with the first 3 patients in arm 1, then next 3 in arm 2, and so forth. This study design aimed to allow faster enrollment and more combination targeted therapies studied than the traditional 3 + 3 design of just one treatment modality. The most successful combination with the least toxicity would then have been expanded to a phase II study. We are reporting Arm 3 in this manuscript. The study is a phase I dose-escalation trial to establish the MTD of the combination of sorafenib and tipifarnib. The MTD was to be determined using the 3 + 3 trial design. The study was also designed to evaluate the pharmacokinetics of the combination therapy.

Dosing and escalation

Tipifarnib and sorafenib were supplied by the Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute (Bethesda, MD). Tipifarnib was given once or twice a day, depending on the treatment cohort, with food. Sorafenib was given twice a day with or without food. If given with meals, patients were instructed to take sorafenib with a moderate to low-fat meal. For the first cycle of treatment, Cycle 1, only tipifarnib was given on day 1, and sorafenib would be initiated on day 2. Treatments were maintained until unacceptable toxicity, patient withdrawal, or disease progression.
Dose escalation was performed as per Table 2 in cohorts of three patients. Sorafenib dose remained the same in all cohorts at 200 mg twice daily. The starting level, level 0, dosing for tipifarnib was 200 mg twice a day. If no DLT occurred in that cohort, a subsequent cohort of three additional patients would be opened at the next higher dose level, level 1. If one patient experienced a DLT at level 0, three more patients would be added to that dose cohort. If two patients experienced DLT at level 0, the next three patients to be enrolled onto this treatment combination would be at a lower level, level − 1. The MTD was defined as the dose at which no more than 1/6 patients experienced a DLT with the next higher dose having exceeded that limit for DLTs, or if all levels did not have more than 1/6 patients with DLTs, the MTD would be the maximum planned dose level. A cycle was considered 28 days in length. DLT was determined over the first 28 days of treatment (cycle 1).
In the first version of the protocol, the starting dose for tipifarnib was given at 200 mg twice daily. Subsequently, the protocol was amended to revise the dosing schedule for a new level 0, “Revised level 0”, based on the results of a published study showing that higher tipifarnib dosing could be achieved with an alternate week schedule [29]. The Revised level 0 consisted of sorafenib at 200 mg twice daily and tipifarnib at 100 mg twice daily given on days 1–7 and 15–21 (alternate weeks) of a 28-days cycle.

Patient evaluation

Pretreatment evaluation included a medical history and physical examination, baseline tumor measurements by MRI or CT, baseline electrocardiograms, and laboratory tests for hematologies and chemistries obtained within 14 days prior to registration. Hematology was performed every week during the first cycle and then every 2 weeks for subsequent cycles. Chemistry panel and liver function tests were obtained every week for the first cycle and then every 4 weeks for subsequent cycles. Blood pressures and adverse events were evaluated weekly for the first cycle, and complete physical examination and neurologic examination were performed every 4 weeks. Brain imaging was performed prior to every other cycle.
DLT was evaluated according to the National Cancer Institute Common Toxicity Criteria version 3. DLT was defined as any grade 4 hematologic toxicity, grade 3 thrombocytopenia lasting greater than 7 days (per protocol), any grade 3 or 4 non-hematologic toxicity (except electrolyte imbalances, diarrhea, nausea, and vomiting which require maximal medical intervention before determined to be a DLT), any intolerable grade 2 non-hematological or grade 3 hematological toxicity requiring a dose reduction during the first 28 days of treatment, or any toxicity resulting in a treatment delay of > 1 week during the first 28 days of treatment.
Tumor response or progression was measured using Response Evaluation Criteria in Solid Tumor Group (RECIST) criteria. These criteria were used in order to allow for comparisons to previous trials, as this trial occurred before the more modern methods of trial response evaluations such as RANO criteria [30]. Tumor progression was defined as a new lesion representing tumor, clear clinical worsening, failure to return for evaluation due to death or deteriorating condition, 25% increase in tumor measurements, or clear worsening of any evaluable disease.

Pharmacokinetic evaluation

For both tipifarnib and sorafenib, whole blood samples (6 ml) were collected in heparinized (Na or Li) containing, nonseparator tubes by venipuncture (heparin lock) or by central venous catheter if in place. At the time of sampling, the first 1 ml of blood diluted with heparin or saline was discarded and the 6 ml sample then withdrawn. The blood sample was kept on ice and centrifuged within 30 min at 3000 rpm × 10–15 min. The plasma was removed, placed in appropriately labeled polypropylene storage tubes and stored at − 70 °C. Samples for pharmacokinetic analysis were collected on day 1–2 and day 15 of cycle 1 for tipifarnib, and day 2, 15, 28–29 for sorafenib. Samples were shipped frozen on dry ice to the study pharmacologist (J.K.). Quantitative analyses were performed as previously published using liquid chromatography method with tandem mass spectrometry [19, 27].

Statistical considerations

The primary end points for this tipifarnib dose-escalation, phase I study were to define DLT and determine the MTD for dosing in a phase II trial. The dose for patients was escalated as described, and DLT, MTD, and safety were evaluated. Using this dose-escalation scheme, the probabilities of escalating to the next dose level are based on the true rate of DLT at the current dose. Overall, if the true underlying proportion of DLTs was 30% at the current dose, there would be a 42% chance of escalating to the next dose. However, if the proportion of DLTs was 50%, the chance of escalation would only be 11%. Pharmacokinetic evaluation of the drug combinations, with determination of Cmax, Tmax AUC, as well as clearance and plasma half-life was obtained during the first treatment cycle in all phase I patients.

Results

Patient characteristics

A total of 24 patients were enrolled between December 2007 and December 2009 (Table 1) into Arm 3 of this study of sorafenib and tipifarnib. The other 2 arms also completed enrollment during this time, but not reported here. All patients had pathologically confirmed GBM prior to treatment; two patients had a previous diagnosis of a WHO grade III glioma with later transformation to glioblastoma. All patients failed treatments with both radiotherapy and temozolomide chemotherapy. Most of the patients were treated in the 1st or 2nd recurrence. Patients were enrolled in three different dose levels in cohorts of three, with replacement if a patient did not meet criteria for safety evaluations for DLT as defined above (Table 2). In general, most patients ended study because of disease progression, with one due to clinical decline without clear tumor growth, six for unacceptable toxicities, and four withdrew consent to continue study participation. We do not have progression free survival (PFS) data on those who withdrew consents or stopped due to adverse events. For the rest of the patients, median PFS was 55 days, and none had event free survival at 6 months. To date, all 24 patients have expired with a median overall survival (OS) of 4.38 months (data not shown).
Table 1
Patient characteristics
Number
24
Male
16
Female
8
Age, median (range)
57 (27–76)
1st recurrence
18
2nd recurrence
5
3rd recurrence
1
KPS (median, range)
85 (60–100)
Table 2
Dose levels and number of enrollment and DLTs
Dose level
Sorafenib dose (mg)
Tipifarnib dose (mg)
Number enrolled
Number DLT
Number replaced
0
200 bid
100 BID d1–21
10
4
0
− 1
200 bid
100 QD d1–21
9
3
3
Revised 0
200 bid
100 BID d1–7, 15–21
5
3
0
For cycle 1 only, Sorafenib was started on day 2
DLT dose limiting toxicity, bid twice a day, qd once a day, d1–21 taken days 1–21 of 28 days, d1–7/15–21 taken days 1–7, break days 8–14, then taken again days 15–21 of 28-days

Toxicities

A summary of grade 3 and 4 toxicities is provided in Table 3. At starting dose level 0, ten patients were evaluated, four of whom experienced DLTs, including lipase elevation, grade 2 hypertension, diarrhea, and emesis. This arm enrolled additional patients as the toxicities were unexpected, and the study team enrolled additional patients to confirm these toxicities.
Table 3
Grade 3 and 4 events attributable to combination treatment
Dose levels
0
− 1
Revised 0
Total% (N = 24)
Abdominal pain
1
  
4
Arthralgia
 
1
 
4
Diarrhea
2
  
8
Dysphasia
  
1
4
Encephalopathy
  
1
4
Fatigue
2
  
8
Fever
  
1
4
Headache
  
1
4
Hyponatremia
 
1
 
4
Hypophosphatemia
2
2
 
17
Infective myositis
1
  
4
Lipase increased
1
1
2
17
Lymphocyte count decreased
4
3
 
29
Serum amylase increased
1
  
4
Thromboembolic event
  
1
4
Vomiting
1
  
4
Number of patients with a grade 3 or 4 toxicity related to either sorafenib, tipifarnib or combination of both. The first three columns refer to number of patients with grade 3 or 4 toxicity per dose level (please refer to Table 2 for dosage of drugs per dose level). Last column is the total percent of patients with a grade 3 or 4 toxicity at all dose levels
On the lower dose level − 1, two of the first three patients had to be replaced due to noncompliance with treatments. Six additional patients were evaluated (one patient had progression in the original cohort). Three of these patients experienced DLTs including grade 4 lipase elevation, grade 3 arthralgia, and hypophosphatemia.
At revised level 0 with every other week tipifarnib dosing, the first two enrolled patients developed serious adverse events (SAEs) with one grade 3 encephalopathy, headache, fever, dysphasia, and one with grade 4 thromboembolic event and grade 3 lipase elevation. Although these events were not all attributable to study drugs, safety concerns regarding frequent SAEs from this combination led to a decision to stop further enrollment into this study.

Pharmacokinetic data

Pharmacokinetic data were obtained in most patients in the first 2 dose levels. PK studies were not performed when level 0 was revised. The levels of sorafenib and tipifarnib are reported in Table 4 (and Supplementary tables for per patient data). The tipifarnib levels are comparable to historical data of single agent tipifarnib in patients not taking EIAED at MTD of 300 mg twice daily [19], and the AUC do not seem to change significantly between daily or twice daily dosing when accounting for the range of AUCs seen.
Table 4
Pharmacokinetic data
(A) Mean levels of sorafenib/N-oxide
Tipifarnib dose level
Day
Cpmax (µg/ml)
AUC 0–12 (µg × h/ml)
100 mg QD
15
3.34/0.43 (± 1.31/0.23)
30.59/4.16 (± 14.9/1.76)
28
3.43/1.17 (± 1.46/0.85)
41.43/7.83 (± 28.82/6.6)
100 mg BID
15
4.17/0.56 (± 2.99/0.41)
12.17/0.60 (± 16.3/0.8)
28
4.53–0.66 (± 2.38/0.32)
36.45/5.39 (± 17.2/2.71)
(B) Peak plasma concentrations of tipifarnib
Tipifarnib dose level
Day
Cpmax (ng/ml)
100 mg QD
1
209.5 ± 135.85
15
169.5 ± 186
100 mg D1, 100 mg BID D2–21
1
132.17 ± 65.96
15
233.6 ± 84.83
Historicala
1
634 ± 374
15
(C) Plasma concentrations of tipifarnib
Tipifarnib dose level
Day
AUC0-12 (ng × h/ml)
100 mg QD
1
814.5 ± 347.57
15
706 ± 644.88
AUC (ng × h/ml)
903.5 ± 377.43
100 mg D1, 100 mg BID D2–21
1
631.67 ± 431.12
15
390.25 ± 468.8
AUC (ng × h/ml)
761.5 ± 468.8
Historicala
AUC (ng × h/ml)
380 ± 217
(D) Half-life of tipifarnib
Tipifarnib dose level
T1/2 (h)
100 mg QD
6.57 ± 1.18
100 mg D1, 100 mg BID D2–21
3.35 ± 1.29
Historicala
3.66 ± 1.18
AUC area under the curve, Cpmax peak plasma concentration
aLevels of patients not taking EIAED, at Tipifarnib of 300 mg bid, from previous study. Cloughesy et al. [19]

Discussion

This phase I study demonstrated that the combination of tipifarnib and sorafenib was toxic even at a dose of 200 mg twice a day of sorafenib and 100 mg twice a day every other week of tipifarnib. These doses are lower than those reached by prior phase I studies of single agent sorafenib or tipifarnib, including a study of single agent tipifarnib in patients with malignant glioma not receiving EIAED where the MTD was 300 mg twice daily [19, 31]. Since we were unable to achieve even single agent dosage levels, we terminated the trial early and did not attempt to identify a MTD.
Given that malignant gliomas can have multiple aberrant molecular targets, combination targeted therapies are more likely to be effective than a single agent [32]. Single agent molecular targeted trials in recurrent GBM have shown little improvement in survival [16]. However, to date, combination therapeutics, including those targeting the EGFR pathway and/or PI3K pathway have yet to show a survival improvement over single agents [15]. Furthermore, as was seen in this phase I trial, the efficacy of combination therapies may also be limited by failure to achieve expected dosing levels because of increased toxicities due to overlapping target inhibition. Even in those patients who did not experience dose limiting toxicities, survival was low, and no further survival analysis was performed due to the low PFS and OS.
Our subjects experienced toxicities well-known in the side effect profiles of both drugs, including rash and diarrhea for tipifarnib, hypertension and hypophosphatemia for sorafenib, and elevated lipase for both, resulting in more frequent DLTs in combination than previously reported as monotherapy. The combination also seems to have higher frequencies of fatigue, altered mental status, and mucositis than expected with single agents. Other combinatorial studies for treatments of GBM have also resulted in increased toxicities without improved efficacy, possibly from pharmacokinetic interactions between the two drugs [28, 33]. Because we did not see a change in the expected pharmacokinetics of these two agents when used in combination, the toxicities may be due to overlapping target inhibition rather than toxic levels of the drugs themselves.
Future clinical studies on combination molecular treatments should consider other strategies to achieve increased breadth of target inhibition without additional toxicity. Choice of agents for combination treatments should be based on both rational molecular targets and complementary side effect profiles. Promiscuous kinase inhibitors such as sorafenib might make particularly poor partners for combination therapy [34, 35]. Other options include using alternative drug schedules, such as pulsatile dosing to achieve better CNS drug level [36, 37] with the potential to limit exposure to prolonged toxicities, as seen with the currently enrolling trials using pulsatile dosing of erlotinib or lapatinib for GBM [38, 39].

Funding

This study was sponsored by NIH Grant U01 CA-062412. Tipifarnib and sorafenib were provided by the National Cancer Institute, NIH.

Compliance with ethical standards

Conflict of interest

Dr. Chang is a consultant to Edge and Blaze and serves as research support to Route and Novartis. Dr. Dancey receives funding from AstraZeneca, Pfizer, and Merck for her institution. Dr. Wen provides research support to Agios, Angiochem, AstraZeneca, Genentech, Roche, Glaxosmith Kline, Immunocellular Therapeutics, Karyopharm, Merck, Novartis, Oncoceotics, Sanofi-Aventis. Dr. Wen serves on the advisory board for AstraZeneca, Cavion, Genentech/Roche, Insys, Monteris, Novogen, Novartis, Regeneron Vascular Biogenics, VBI Vaccines. Dr. Wen is a speaker for Merck and serves on the Data Monitoring Safety Board for Tocagen and Monteris. Dr. DeAngelis serves on the scientific advisory boards of Sapience, Roche, and Tocagen.
Literature
1.
American Cancer Society (2010) Cancer Facts and Figs. 2010. American Cancer Society, Atlanta
2.
Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJB, Belanger K, Brandes AA, Marosi C, Bogdahn U, Curschmann J, Janzer RC, Ludwin SK, Gorlia T, Allgeier A, Lacombe D, Cairncross JG, Eisenhauer E, Mirimanoff RO, The European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups and the National Cancer Institute of Canada Clinical Trials Group (2005) Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med 352 (10):987–996. doi:10.​1056/​NEJMoa043330 CrossRefPubMed
3.
Wong E, Hess K, Gleason M, Jaeckle K, AP K, MD P, VA L, WK Y (1999) Outcomes and prognostic factors in recurrent glioma patients enrolled onto phase II clinical trials. J Clin Oncol 17(8):2572CrossRefPubMed
4.
Lamborn KR, Yung WKA, 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, North American Brain Tumor Consortium Study (2008) Progression-free survival: an important end point in evaluating therapy for recurrent high-grade gliomas. Neuro Oncol 10 (2):162–170. doi:10.​1215/​15228517-2007-062 CrossRefPubMedPubMedCentral
5.
Friedman HS, Prados M, Wen PY, Mikkelsen T, Schiff D, Abrey LE, Yung WK, Paleologos N, Nicholas MK, Jensen R, Vredenburgh J, Huang J, Zheng M, Cloughesy T (2009) Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 27(28):4733–4740CrossRefPubMed
6.
Kreisl TN, Zhang W, Odia Y, Shih JH, Butman JA, Hammoud D, Iwamoto FM, Sul J, Fine HA (2011) A phase II trial of single-agent bevacizumab in patients with recurrent anaplastic glioma. Neuro Oncol 13(10):1143–1150. doi:10.​1093/​neuonc/​nor091 CrossRefPubMedPubMedCentral
7.
Maher EA, Furnari FB, Bachoo RM, Rowitch DH, Louis DN, Cavenee WK, DePinho RA (2001) Malignant glioma: genetics and biology of a grave matter. Genes Dev 15(11):1311–1333CrossRefPubMed
8.
Feldkamp MM, Lala P, Lau N, Roncari L, Guha A (1999) Expression of activated epidermal growth factor receptors, Ras-guanosine triphosphate, and mitogen-activated protein kinase in human glioblastoma multiforme specimens. Neurosurgery 45(6):1442–1453CrossRefPubMed
9.
Dancey J, Sausville EA (2003) Issues and progress with protein kinase inhibitors for cancer treatment. Nat Rev Drug Discov 2(4):296–313. doi:10.​1038/​nrd1066 CrossRefPubMed
10.
Guha A (1998) Ras activation in astrocytomas and neurofibromas. Can J Neurol Sci 25(4):267–281CrossRefPubMed
11.
Delmas C, Heliez C, Cohen-Jonathan E, End D, Bonnet J, Favre G, Toulas C (2002) Farnesyltransferase inhibitor, R115777, reverses the resistance of human glioma cell lines to ionizing radiation. Int J Cancer 100(1):43–48. doi:10.​1002/​ijc.​10439 CrossRefPubMed
12.
End DW, Smets G, Todd AV, Applegate TL, Fuery CJ, Angibaud P, Venet M, Sanz G, Poignet H, Skrzat S, Devine A, Wouters W, Bowden C (2001) Characterization of the antitumor effects of the selective farnesyl protein transferase inhibitor R115777 in vivo and in vitro. Cancer Res 61(1):131–137PubMed
13.
Feldkamp MM, Lau N, Roncari L, Guha A (2001) Isotype-specific Ras. GTP-levels predict the efficacy of farnesyl transferase inhibitors against human astrocytomas regardless of Ras mutational status. Cancer Res 61(11):4425–4431PubMed
14.
Delmas C, End D, Rochaix P, Favre G, Toulas C, Cohen-Jonathan E (2003) The farnesyltransferase inhibitor R115777 reduces hypoxia and matrix metalloproteinase 2 expression in human glioma xenograft. Clin Cancer Res 9(16 Pt 1):6062–6068PubMed
15.
Wick W, Weller M, Weiler M, Batchelor T, Yung AW, Platten M (2011) Pathway inhibition: emerging molecular targets for treating glioblastoma. Neuro Oncol 13(6):566–579. doi:10.​1093/​neuonc/​nor039 CrossRefPubMedPubMedCentral
16.
Gilbert MR (2011) Recurrent glioblastoma: a fresh look at current therapies and emerging novel approaches. Semin Oncol 38(Suppl 4):S21–S33. doi:10.​1053/​j.​seminoncol.​2011.​09.​008 CrossRefPubMed
17.
Hudes G, Schol J, Baab J (1999) Phase I clinical and pharmacokinetic trial of the farnesyltransferase inhibitor R115777 in a 21-day dosing schedule. Paper presented at the proceedings of the annual meeting of the Amercian Society of Clinical Oncology, May 1999
18.
Zujewski J, Horak ID, Bol CJ, Woestenborghs R, Bowden C, End DW, Piotrovsky VK, Chiao J, Belly RT, Todd A, Kopp WC, Kohler DR, Chow C, Noone M, Hakim FT, Larkin G, Gress RE, Nussenblatt RB, Kremer AB, Cowan KH (2000) Phase I and pharmacokinetic study of farnesyl protein transferase inhibitor R115777 in advanced cancer. J Clin Oncol 18(4):927–941CrossRefPubMed
19.
Cloughesy TF, Kuhn J, Robins HI, Abrey L, Wen P, Fink K, Lieberman FS, Mehta M, Chang S, Yung A, DeAngelis L, Schiff D, Junck L, Groves M, Paquette S, Wright J, Lamborn K, Sebti SM, Prados M (2005) Phase I trial of tipifarnib in patients with recurrent malignant glioma taking enzyme-inducing antiepileptic drugs: a North American Brain Tumor Consortium Study. J Clin Oncol 23(27):6647–6656. doi:10.​1200/​JCO.​2005.​10.​068 CrossRefPubMed
20.
Cloughesy TF, Wen PY, Robins HI, Chang SM, Groves MD, Fink KL, Junck L, Schiff D, Abrey L, Gilbert MR, Lieberman F, Kuhn J, DeAngelis LM, Mehta M, Raizer JJ, Yung WK, Aldape K, Wright J, Lamborn KR, Prados MD (2006) Phase II trial of tipifarnib in patients with recurrent malignant glioma either receiving or not receiving enzyme-inducing antiepileptic drugs: a North American Brain Tumor Consortium Study. J Clin Oncol 24(22):3651–3656. doi:10.​1200/​JCO.​2006.​06.​2323 CrossRefPubMed
21.
Kuhn J, Prados M, Robins H (2000) Phase I trial of R115777 (Zarnestra) in patients with recurrent malignant glioma taking enzyme inducting antiepileptic drugs (EIAED): a North American Brain Tumor Consortium (NABTC) report. In: Proceedings of the American Society of Clinical Oncology, pp 86a, Abstract no. 342
22.
Wilhelm S, Chien DS (2002) BAY 43-9006: preclinical data. Curr Pharm Des 8(25):2255–2257CrossRefPubMed
23.
Dy GK, Adjei AA (2002) Novel targets for lung cancer therapy: part II. J Clin Oncol 20(13):3016–3028. doi:10.​1200/​JCO.​2002.​02.​112 CrossRefPubMed
24.
Dy GK, Adjei AA (2002) Novel targets for lung cancer therapy: part I. J Clin Oncol 20(12):2881–2894. doi:10.​1200/​JCO.​2002.​11.​145 CrossRefPubMed
25.
Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H, Chen C, Zhang X, Vincent P, McHugh M, Cao Y, Shujath J, Gawlak S, Eveleigh D, Rowley B, Liu L, Adnane L, Lynch M, Auclair D, Taylor I, Gedrich R, Voznesensky A, Riedl B, Post LE, Bollag G, Trail PA (2004) BAY 43-9006 exhibits broad spectrum oral antitumor activity and targets the RAF/MEK/ERK pathway and receptor tyrosine kinases involved in tumor progression and angiogenesis. Cancer Res 64(19):7099–7109. doi:10.​1158/​0008-5472.​CAN-04-1443 CrossRefPubMed
26.
Heim M, Scharifi M, Zisowsky J, Jaehde U, Voliotis D, Seeber S, Strumberg D (2005) The Raf kinase inhibitor BAY 43-9006 reduces cellular uptake of platinum compounds and cytotoxicity in human colorectal carcinoma cell lines. Anticancer Drugs 16(2):129–136CrossRefPubMed
27.
Lee EQ, Kuhn J, Lamborn KR, Abrey L, DeAngelis LM, Lieberman F, Robins HI, Chang SM, Yung WK, Drappatz J, Mehta MP, Levin VA, Aldape K, Dancey JE, Wright JJ, Prados MD, Cloughesy TF, Gilbert MR, Wen PY (2012) Phase I/II study of sorafenib in combination with temsirolimus for recurrent glioblastoma or gliosarcoma: North American Brain Tumor Consortium study 05-02. Neuro Oncol 14(12):1511–1518. doi:10.​1093/​neuonc/​nos264 CrossRefPubMedPubMedCentral
28.
Prados M, Gilbert M, Kuhn J, Lamborn K, Cloughesy T, Lieberman F, Puduvalli V, Robins HI, Lassman A, Wen PY (2009) Phase I/II study of sorefenib and erlotinib for patients with recurrent glioblastoma (GBM) (NABTC 05-02). J Clin Oncol 27:15 (suppl; abstr 2005)CrossRef
29.
Kirschbaum M, Synold T, Stein AS, Tuscano J, Zain JM, Popplewell L, Karanes C, O’Donnell MR, Pulone B, Rincon A, Wright J, Frankel P, Forman SJ, Newman EM (2011) A phase 1 trial dose escalation study of tipifarnib on a week-on, week-off schedule in relapsed, refractory or high-risk myeloid leukemia. Leukemia 25(10):1543–1547. doi:10.​1038/​leu.​2011.​124 CrossRefPubMedPubMedCentral
30.
Wen PY, Chang SM, Van den Bent MJ, Vogelbaum MA, Macdonald DR, Lee EQ (2017) Response assessment in neuro-oncology clinical trials. J Clin Oncol 35(21):2439–2449. doi:10.​1200/​JCO.​2017.​72.​7511 CrossRefPubMed
31.
Strumberg D, Richly H, Hilger RA, Schleucher N, Korfee S, Tewes M, Faghih M, Brendel E, Voliotis D, Haase CG, Schwartz B, Awada A, Voigtmann R, Scheulen ME, Seeber S (2005) Phase I clinical and pharmacokinetic study of the Novel Raf kinase and vascular endothelial growth factor receptor inhibitor BAY 43-9006 in patients with advanced refractory solid tumors. J Clin Oncol 23(5):965–972. doi:10.​1200/​JCO.​2005.​06.​124 CrossRefPubMed
32.
Stommel JM, Kimmelman AC, Ying H, Nabioullin R, Ponugoti AH, Wiedemeyer R, Stegh AH, Bradner JE, Ligon KL, Brennan C, Chin L, DePinho RA (2007) Coactivation of receptor tyrosine kinases affects the response of tumor cells to targeted therapies. Science 318(5848):287–290. doi:10.​1126/​science.​1142946 CrossRefPubMed
33.
Reardon DA, Cloughesy T, Rich J, Alfred Yung WK, Yung L, DiLea C, Huang J, Dugan M, Mietlowski W, Maes A, Conrad C (2012) Pharmacokinetic drug interaction between AEE788 and RAD001 causing thrombocytopenia in patients with glioblastoma. Cancer Chemother Pharmacol 69(1):281–287. doi:10.​1007/​s00280-011-1754-1 CrossRefPubMed
34.
Galanis E, Anderson SK, Lafky JM, Uhm JH, Giannini C, Kumar SK, Kimlinger TK, Northfelt DW, Flynn PJ, Jaeckle KA, Kaufmann TJ, Buckner JC (2013) Phase II study of bevacizumab in combination with sorafenib in recurrent glioblastoma (N0776): a north central cancer treatment group trial. Clin Cancer Res 19(17):4816–4823. doi:10.​1158/​1078-0432.​CCR-13-0708 CrossRefPubMed
35.
Peereboom DM, Ahluwalia MS, Ye X, Supko JG, Hilderbrand SL, Phuphanich S, Nabors LB, Rosenfeld MR, Mikkelsen T, Grossman SA, New Approaches to Brain Tumor Therapy Consortium (2013) NABTT 05-02: a phase II and pharmacokinetic study of erlotinib and sorafenib for patients with progressive or recurrent glioblastoma multiforme. Neuro Oncol 15 (4):490–496. doi:10.​1093/​neuonc/​nos322 CrossRefPubMedPubMedCentral
36.
Clarke JL, Pao W, Wu N, Miller VA, Lassman AB (2010) High dose weekly erlotinib achieves therapeutic concentrations in CSF and is effective in leptomeningeal metastases from epidermal growth factor receptor mutant lung cancer. J Neurooncol 99(2):283–286. doi:10.​1007/​s11060-010-0128-6 CrossRefPubMedPubMedCentral
37.
Vivanco I, Robins HI, Rohle D, Campos C, Grommes C, Nghiemphu PL, Kubek S, Oldrini B, Chheda MG, Yannuzzi N, Tao H, Zhu S, Iwanami A, Kuga D, Dang J, Pedraza A, Brennan CW, Heguy A, Liau LM, Lieberman F, Yung WKA, Gilbert MR, Reardon DA, Drappatz J, Wen PY, Lamborn KR, Chang SM, Prados MD, Fine HA, Horvath S, Wu N, Lassman AB, DeAngelis LM, Yong WH, Kuhn JG, Mischel PS, Mehta MP, Cloughesy TF, Mellinghoff IK (2012) Differential sensitivity of glioma-versus lung cancer–specific EGFR mutations to EGFR kinase inhibitors. Cancer Discov. doi:10.​1158/​2159-8290.​cd-11-0284 PubMedPubMedCentral
38.
EGFR Inhibition Using High Dose Administration of Erlotinib Weekly for Recurrent malignant Gliomas with EGFR Variant III Mutation (2011) ClinicalTrials.gov. http://​clinicaltrials.​gov/​ct2/​show/​NCT01257594?​term=​erlotinib+AND+ne​w+York+AND+gliob​lastoma&​rank=​5. Accessed Feb 2012
39.
Yu A, Faiq N, Green S, Lai A, Green R, Hu J, Cloughesy TF, Mellinghoff I, Nghiemphu PL (2017) Report of safety of pulse dosing of lapatinib with temozolomide and radiation therapy for newly-diagnosed glioblastoma in a pilot phase II study. J Neurooncol. doi:10.​1007/​s11060-017-2533-6 PubMedCentral