Abstract
Purpose
The primary objective of this study was to use intracerebral microdialysis (ICMD) to determine the neuropharmacokinetics of bafetinib, a dual BCR-Abl/Lyn tyrosine kinase inhibitor that may have activity against gliomas.
Methods
A microdialysis catheter was placed into either peritumoral or enhancing brain tissue of 7 patients at time of tumor resection or biopsy. Twenty-four hours later, bafetinib was administered, 240 or 360 mg po, repeating the same dose 12 hours later. Dialysate samples were continuously collected for 24 hours, with plasma samples obtained in parallel. One to two weeks after finishing ICMD, patients were allowed to resume taking bafetinib continuously while being observed for toxicity and tumor response.
Results
Twenty-six dialysate samples per patient were collected (n=6) and analyzed for bafetinib by tandem mass spectrometry. Bafetinib concentrations in brain were below the lower limit of detection of the assay (0.1 ng/ml) in all samples except 1 from a single subject that was 0.52 ng/ml. The mean plasma bafetinib maximum concentrations after dose 1 and 2 were 143±99 and 247±73 ng/ml, respectively. Only 1 patient remained on treatment past 2 cycles, and no radiographic responses were seen.
Conclusions
Bafetinib does not sufficiently cross intact or disrupted blood-brain barrier, and therefore, systemic administration of bafetinib is not recommended when investigating this drug as a treatment for brain tumors. ICMD can be a valuable research tool in early drug development. Lead-in ICMD studies can be performed relatively quickly, requiring only a small number of patients, and without significantly disrupting standard cancer care.
Keywords: intracerebral microdialysis, bafetinib, neuropharmacokinetics, brain tumor
1. Introduction
Bafetinib is a potent second generation breakpoint cluster region—Abelson (BCR-ABL)/Lyn tyrosine kinase inhibitor (TKI)1,2 which is currently being investigated as a treatment for Philadelphia chromosome-positive leukemia. In addition to inhibiting autophosphorylation of ABL point mutations, this dual TKI selectively inactivates Lyn3 and Fyn, which are members of the sarcoma family kinases. Analysis of glioblastoma samples detected increased Lyn kinase activity,4 which may contribute to its malignant phenotype. Co-activation of Fyn kinase with epidermal growth factor receptors promotes tumor invasion and survival in glioblastoma.5In vitro studies of bafetinb alone or in combination with either temozolomide or erlotinib demonstrated activity against glioma cell lines.6 As a potent inhibitor of Lyn and Fyn , bafetinib may be effective in blocking the growth and spread of glioblastoma.
For other BCR-ABL TKIs, such as imatinib7-12 and dasatinib,13,14 conflicting data exist as to how well they cross the blood-brain barrier (BBB). With bafetinib, preclinical rodent studies showed that after oral administration, concentrations in rat brain were approximately 10% of plasmalevels.15,16 In mice, peak bafetinib concentrations in the brain occurred 2 hours after oral administration, achieving concentrations above the IC50 for leukemic cell lines.16 However, bafetinib, like imatinib,17,18 is a substrate for P-glycoprotein (P-gp),16 a transmembrane drug efflux pump found in BBB. To investigate the potential of bafetinib as a treatment for brain tumors, we performed an intracerebral microdialysis study to assess its neuropharmacokinetics in patients with recurrent high-grade gliomas.
2. Patients and Methods
2.1 Determination of the fractional recovery of bafetinib by the microdialysis catheter
A 70 Brain Microdialysis Catheter (membrane length 10 mm; shaft length 100 mm; semipermeable membrane molecular weight cut off of 20,000 Da; ref. no. P000050, M Dialysis, Solna, Sweden) was submerged in a 15 mL conical centrifuge tube containing bafetinib (200 ng/mL) in artificial cerebrospinal fluid (CSF) [Perfusion Fluid CNS, ref. no. P000151, M Dialysis, Solna, Sweden], at 37° C. Artificial CSF perfused the catheter at rates of 0.5 or 1.0 μL/min. Dialysate samples (30 μL) were collected at regular intervals and analyzed by liquid chromatography tandem mass spectrometry (LC-MS/MS).
2.2 Patient selection
To be eligible for participation in this pilot study, patients had to be ≥18 years old, have radiographic findings consistent with recurrent high-grade glioma, and be in need of tumor resection or biopsy. Other inclusion criteria were: a) Karnofsky performance status (KPS) ≥ 60%, b) recovery from toxicity of prior therapy, c) adequate bone marrow function (absolute neutrophil count ≥ 1500 cells/mm3 and platelet count ≥ 100,000 cells/mm3), hepatic function (total bilirubin ≤ 2.0 mg/dL, serum levels of aspartate aminotransferase and alanine aminotransferase ≤ 3 × the institutional upper limit of normal), and renal function (serum creatinine ≤ 1.5 × the institutional upper limit of normal), d), a minimum of 4 weeks from previous chemotherapy (6 weeks from a nitrosourea), and e) QTc interval <480 msec on electrocardiogram.
Patients were excluded from study participation if they a) were taking hepatic enzyme-inducing anticonvulsants within 2 weeks prior to enrollment, b) were receiving chemotherapy, radiation, or enrolled in another clinical trial, c) had a coagulopathy or were taking anticoagulant therapy or medications that inhibit platelet function, d) were pregnant or breast-feeding, or e) had a serious medical or psychiatric illness that could potentially interfere with the completion of study treatment.
Participants gave written informed consent. The study was approved by the City of Hope Institutional Review Board (IRB), conducted under an Investigational New Drug Application (IND# 110189), and registered at ClinicalTrials.gov (NCT01234740).
2.3 Treatment plan
During surgery, if frozen section indicated the presence of recurrent tumor, the neurosurgeon inserted a microdialysis catheter (70 Brain Microdialysis Catheter, M Dialysis, Solna, Sweden) into residual tumor or peritumoral tissue within 5-15 mm of the resection cavity. After a post-operative non-contrast computerized tomography (CT) scan of the brain confirmed the location of the catheter, a pump (107 Microdialysis Pump, ref. no. P000127, M Dialysis, Solna Sweden), perfused the catheter with artificial CSF at a rate of 0.5 μl/min. Contrast-enhanced brain magnetic resonance images (MRI) were obtained within 24 hours of surgery to serve as a baseline for assessing response and evaluate microdialysis catheter position (in enhancing versus peritumoral tissue) by creating a fused MRI/CT image.
At least 24 hours after surgery, study patients received an oral dose of bafetinib on an empty stomach: either 240 mg (patients 1-5), which was the maximum tolerated dose determined in a previous phase I study,19 or 360 mg (patient 6), with the same dose repeated 12 hours later. Dialysate samples were obtained hourly for 24 hours starting after the first dose. To stabilize bafetinib, 6 μL of 6% acetic acid was added to each microvial. Dialysate samples were immediately placed on dry ice and stored at ≤ −70°C until analysis. Plasma samples were obtained prior to each dose of bafetinib, and then 0.5, 1, 2, 3, 4, 6, 8, and 12 hours after each dose of bafetinib.
After completing the microdialysis portion of the study, patients were allowed to resume taking bafetinib after recovery from surgery. Brain MRIs were performed after every 2 cycles to assess response. All observed toxicities were graded using Common Terminology Criteria for Adverse Events version 4.0. MacDonald Criteria20 were used to assess radiographic responses.
2.4 Pharmacokinetic data analysis
Dialysate and plasma samples were analyzed for bafetinib using validated LC-MS/MS assays. Please see Supplementary Data for methodologic details.
2.5 Statistical analysis plan
The primary objective of this study was to evaluate the neuropharmacokinetics of bafetinib in patients with recurrent high-grade gliomas using intracerebral microdialysis and compare these data to plasma levels. Secondary objectives included describing toxicities and clinical benefit in this small cohort of patients with primary brain tumors. Based on our previous experience with using microdialysis to measure chemotherapy levels in the brain,21 we estimated that a sample size of 6-8 patients would be sufficient to characterize the neuropharmacokinetics of bafetinib.
3. Results
3.1 In vitro recovery results
In vitro recovery experiments determined that the fractional recovery of bafetinib at a flow rate of 0.5 μL/min was 85-90%, compared to 60-70% at a flow rate of 1 μL/min.
3.2 Patient characteristics
Seven subjects were enrolled from December 2010 to June 2011. Microdialysis data were successfully collected from 6 patients. The catheter would not flush after placement in 1, and that patient decided not to continue on study. Table 1 shows the demographics of the 6 patients from whom microdialysis data were obtained. All had recurrent high-grade gliomas and were previously treated with brain radiation and chemotherapy. During microdialysis, patients took non-hepatic enzyme inducing anticonvulsants (mainly levetiracetam). All patients received post-operative dexamethasone. The median total daily dose of dexamethasone was 12 mg (range 4-40 mg).
Table 1.
Patient characteristics
| Patient | Age (years) | Gender | Diagnosis | KPS | Prior Chemotherapy | Surgical Procedure |
|---|---|---|---|---|---|---|
| 1 | 68 | Male | GBM | 70 | TMZb, gene therapy study | Biopsy |
| 2 | 37 | Male | GBM | 60 | TMZ, isotretinoin, bevc, carboplatin | Resection |
| 3 | 38 | Female | PXAa with malignant features | 70 | TMZ | Resection |
| 4 | 30 | Female | Anaplastic astrocytoma | 90 | TMZ, carboplatin, etoposide, | Resection |
| 5 | 27 | Female | GBM | 80 | TMZ, bev | Biopsy |
| 6 | 31 | Male | Anaplastic oligoastrocytoma | 80 | carmustine wafer, TMZ | Biopsy |
PXA = pleomorphic xanthoastrocytoma. Although PXA is classified as a grade II glioma by the World Health Organization, when this patient's tumor recurred, it had anaplastic features consistent with a higher grade glioma, and so she was eligible to participate in the study.
TMZ = temozolomide
bev = bevacizumab
3.3 Brain interstitial levels of bafetinib
Although a highly sensitive and validated analytical method was used to measure bafetinib in dialysate (lower limit of quantitation [LLOQ] = 0.5 ng/ml and detection [LOD] = 0.1 ng/ml), analysis of the 156 dialysate samples (26 per patient) showed no detectable bafetinib in brain extracellular fluid (ECF) with the exception of a single sample from patient 2, which was obtained 5 hours after the first dose of study drug. The concentration of bafetinib was 0.52 ng/ml. Although this patient had the second highest maximum plasma concentration (Cmax) following dose 1 and a time to maximum plasma concentration (Tmax) of 2 hours, there was no clear relationship between plasma and ECF levels.
The microdialysis catheters in the first 4 study subjects were placed in non-enhancing peritumoral tissue. Figure 1A shows an example of a fused image of a study patient's immediate post-operative non-contrast CT scan (in which the catheter tip is visible) and a contrast-enhanced MRI performed approximately 24 hours after surgery. This merged image documents that the microdialysis catheter was placed in non-enhancing tissue where BBB is relatively intact.
Figure 1.
Fused images of a non-contrast head CT scan and a T1 post-contrast brain MRI of a study patient who underwent a gross total resection of tumor in the left frontal lobe. The CT scan was performed after the catheter was inserted to confirm correct placement of the catheter, and the MRI was performed on post-operative day 1. Images were autoregistered with mutual information matching algorithm using Varian Eclipse version 10.0 treatment planning system. The blended image is shown. A gold filament at the tip of the microdialysis catheter (arrow) is visible as bright signal on the CT scan but not visible on the T1 post-contrast MRI. Fused CT and T1 post-contrast MRI shows that the tip of the microdialysis catheter is placed in non-enhancing brain.
Because bafetinb was undetectable in nearly all dialysate samples from the first 4 patients, and since many drugs that cannot cross intact BBB may still pass through disrupted blood-tumor barrier, subsequent enrollment was restricted to patients undergoing biopsy only to assess bafetinib concentrations in enhancing tissue. Patients 5 and 6 had catheters placed within enhancing brain tissue (figure 1B). Furthermore, with IRB approval, patient 6 received a dose of 360 mg bid. Analysis of dialysate samples from both these patients, however, showed no detectable levels of bafetinib in brain ECF.
Figure 1.
Fused images of a non-contrast head CT scan and a T1 post-contrast brain MRI of a study patient who underwent a biopsy of tumor in the right parietal lobe. The CT scan was performed after the catheter was inserted to confirm correct placement of the catheter, and the MRI was performed on post-operative day 1. Images were autoregistered with mutual information matching algorithm using Varian Eclipse version 10.0 treatment planning system. The blended image shows the gold filament of the catheter tip (arrow) as bright signal adjacent to enhancing brain tissue. The 3 mm gold tip is anterior to the 10 mm semi-permeable membrane (not visible on CT scan or MRI), which is in enhancing brain tissue.
3.4 Plasma levels of bafetinib
Table 2 and figure 2 summarize the bafetinib plasma pharmacokinetic data. A total of 102 plasma samples (17 per patient) were collected. The mean Cmax after dose 1 and 2 were 143±99 and 247±73 ng/ml respectively. The mean area-under-the-concentration-time curve to the last measured time point (AUC 0-12h) following dose 1 and 2 were 660±431 and 1174±534 ng/mlxhr, respectively. The average Tmax following dose 1 and 2 were 6±4 and 5±2 hr, respectively. As depicted in figure 2, there was significant inter-patient variability in the plasma concentrations. The bafetinib plasma pharmacokinetic results from this study are similar to previously published data.19
Table 2.
Plasma pharmacokinetics of bafetinib
| Dose #1 | Dose #2 | ||||||
|---|---|---|---|---|---|---|---|
| Patient | Dose (mg) | Cmax (ng/ml) | Tmax (hr) | AUC 0-12h (ng/ml × hr) | Cmax (ng/ml) | Tmax (hr) | AUC 0-12h (ng/ml × hr) |
| 1 | 240 | 112 | 4 | 623 | 235 | 3 | 921 |
| 2 | 240 | 237 | 2 | 873 | 183 | 3 | 585 |
| 3 | 240 | 65 | 8 | 374 | 340 | 4 | 1151 |
| 4 | 240 | 285 | 4 | 1396 | 231 | 8 | 1224 |
| 5 | 240 | 32 | 12 | 169 | 330 | 8 | 2164 |
| 6 | 360 | 129 | 3 | 522 | 164 | 4 | 1000 |
| AVG = | 143 | 6 | 660 | 247 | 5 | 1174 | |
| SD = | 99 | 4 | 431 | 73 | 2 | 534 | |
Figure 2.
Plot of the bafetinib plasma concentration versus time following the first 2 oral doses taken 12 hours apart. Solid circles are the mean concentrations at each time point, and the error bars are the standard deviations.
3.5 Safety and efficacy
All participants tolerated the intracerebral microdialysis well. Two developed grade 3 toxicities at least possibly related to bafetinib. One participant experienced a grade 3 elevation of alanine aminotransferase, and the other developed bacterial meningitis approximately 3 weeks after tumor resection and placement of the microdialysis catheter. Although the meningitis was likely a late post-operative complication, an association with bafetinib could not be excluded.
There were no radiographic responses to bafetinib. Three patients developed progressive disease within 2 months of starting study drug. One patient from whom dialysate samples were collected, experienced neurologic decline after surgery and did not resume bafetinib. Only 1 patient remained on study treatment past 2 months. He received 6 cycles of bafetinib, with serial brain MRIs showing stable disease. However, once the microdialysis data were available, the slides from this patient's biopsy were re-reviewed by the study neuropathologist who concluded that the pre-biopsy radiographic findings most likely represented treatment effect from radiation (pseudoprogression). Therefore, the stable disease during bafetinib treatment was likely due to lack of tumor progression at the time of study enrollment rather than inhibition of tumor growth by bafetinib. The last patient to enroll was taken off study 2 weeks into his first cycle of bafetinib when the study was closed based on the final analysis of dialysate samples showing no detectable levels of bafetinib in the brain.
4. Discussion
Intracerebral microdialysis is a technique for continuously analyzing the concentration of a drug or biomolecule in the ECF of the brain without significantly disrupting tissue function.22,23 In this study, microdialysis was used to assess the neuropharmacokinetics of bafetinib prior to initiating an efficacy study in glioma patients. Our findings suggest that bafetinib does not sufficiently cross an intact or disrupted BBB, and therefore, systemically administered bafetinib is not recommended for testing in phase II brain tumor clinical trials. Inhibition of Lyn and Fyn kinases is a reasonable glioma treatment strategy, and exploring an alternative route of delivery for bafetinib may be worthy of further investigation.
In this study we assessed bafetinib concentrations in both peritumoral and enhancing brain tissue and determined that the average brain ECF: plasma bafetinib ratio in patients is likely to be < 0.05%. Studies in rats have suggested that bafetinib levels in the brain are approximately 10% of systemic levels.15,16 Since plasma protein binding of bafetinib in both humans and rats is 95%,24 the apparent species-related difference in CNS penetration cannot be explained by differences in the availability of free drug. Assuming a free drug fraction of 5% in patient plasma, a fractional recovery of 85%, and equilibration of free drug across the BBB, we would expect to measure bafetinib levels in brain ECF in the range of 1-10 ng/ml, which would have been easily detectable by our method (LOD = 0.1 ng/ml).
Interspecies differences do exist regarding BBB permeability, and the observed discrepancy in brain uptake of bafetinib between rodents and humans could be due to dissimilarities in levels of expression, and substrate specificity of various efflux transporters, and/or BBB structural differences .25-27 In addition, the radiometric method used in the rats15 can over-estimate drug levels in the brain due to the presence of residual blood in the tissue sample. The results of this study highlight the inadequacy of extrapolating data about CNS drug penetration from animal models to humans. Intracerebral microdialysis, which directly measures drug concentrations, is a preferred method for determining whether a drug can achieve therapeutic levels in the human brain.
Prior to performing intracerebral microdiaysis in patients, in vitro studies must be done to assess a drug's fractional recovery and determine the optimal conditions for in vivo recovery. Nonetheless, in vitro recovery may overestimate in vivo recovery because the experimental set up cannot simulate conditions that affect in vivo recovery.28 Several methods exist28,29 for in vivo catheter calibration, of which retrodialysis30 is the most simple and direct. However, because bafetinib is not available as an intravenous formulation, it was not feasible to perform retrodialysis in our study patients.
Not all anti-cancer agents are amenable to microdialysis. Factors that limit a drug's recovery include high molecular weight, aqueous insolubility, and lipophilicity. In addition, microdialysis provides information only about drug levels within the vicinity of the catheter. Since tissues can be heterogeneous, the levels may not reflect drug concentrations throughout the tumor.
Despite these methodological concerns, use of microdialysis to perform neuropharmacokinetic assessments is growing.21,31,32 We have previously applied this technique to describe changes in intracerebral levels of temozolomide,21 a drug whose ability to cross the BBB is well established. In that study, we demonstrated that the time course of temozolomide concentrations in brain interstitium was not the same as in plasma, which may have implications for the optimal timing of radiation when combined with temozolomide.
In this current study, we were able to conclude with only 6 patients that bafetinib does not achieve adequate concentrations in brain interstitium. In comparison, it required over 700 patients, within the context of multiple phase II trials33-37 as well as a phase III trial,38 to determine that imatinib had minimal activity against high-grade gliomas. While it is unclear whether imatinib lacked efficacy because of its inability to reach tumor in the brain, like bafetinib, the drug is highly protein-bound and a substrate for efflux transporters. Two studies7,12 reported detectable imatinib levels in post-treatment tumor samples; however, as previously discussed, tissue-based measurements may over-estimate CNS penetration due to the contribution of residual blood in the specimen. Moreover, tissue measurements can only give information about intratumoral drug levels at one timepoint, whereas microdialysis can provide information about the time course of changes in drug levels in the brain.
This neuropharmacokinetic study of bafetinib illustrates the valuable role intracerebral microdialysis can play in early drug development. Such studies can be done within the context of standard cancer treatment—while patients are undergoing a craniotomy or brain biopsy. Although intracerebral microdialysis must take place in the hospital, collection of dialysate samples is done in ambulatory patients while recovering from surgery, often without prolonging hospitalization. Since only a small number of participants are needed, these studies can be performed relatively inexpensively and quickly. Lead-in microdialysis studies, such as the one described here, should be considered to assess the neuropharmacokinetics of a drug at its established maximum tolerated dose prior to initiating efficacy studies in brain tumor patients.
Supplementary Material
Acknowledgements
We thank Rachel Magnusson, R.N., for her meticulous management of the microdialysis catheters.
Role of the Funding Source CytRx Corporation provided financial support for this study and coordinated the analysis of the plasma samples. The study sponsor was not involved in the design of the protocol, the collection and interpretation of the data, the writing of the manuscript, or the decision to submit the manuscript for publication.
Financial support: CytRx Corporation
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Kimura S, Naito H, Segawa H, et al. NS-187, a potent and selective dual Bcr-Abl/Lyn tyrosine kinase inhibitor, is a novel agent for imatinib-resistant leukemia. Blood. 2005;106:3948–54. doi: 10.1182/blood-2005-06-2209. [DOI] [PubMed] [Google Scholar]
- 2.Goldman J, Melo J. Chronic Myeloid Leukemia – Advances in Biology and New Approaches to Treatment. N Engl J Med. 2003;349:1451–64. doi: 10.1056/NEJMra020777. [DOI] [PubMed] [Google Scholar]
- 3.Santos FP, Kantarjian H, Cortes J, Quintas-Cardama A. Bafetinib, a dual Bcr-Abl/Lyn tyrosine kinase inhibitor for the potential treatment of leukemia. Curr Opin Investig Drugs. 2010;11:1450–65. [PubMed] [Google Scholar]
- 4.Stettner MR, Wang W, Nabors LB, et al. Lyn kinase activity is the predominant cellular SRC kinase activity in glioblastoma tumor cells. Cancer Res. 2005;65:5535–43. doi: 10.1158/0008-5472.CAN-04-3688. [DOI] [PubMed] [Google Scholar]
- 5.Lu KV, Zhu S, Cvrljevic A, et al. Fyn and SRC are effectors of oncogenic epidermal growth factor receptor signaling in glioblastoma patients. Cancer Res. 2009;69:6888–98. doi: 10.1158/0008-5472.CAN-09-0347. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Velcheti V, Peterson B, Levitt D, Prakash O. Preclinical evaluation of bafetinib for treatment of glioblastoma multiforme. J Clin Oncol, ASCO Annual Meeting Proceedings. 2010;28(suppl):2082. [Google Scholar]
- 7.Holdhoff M, Supko JG, Gallia GL, et al. Intratumoral concentrations of imatinib after oral administration in patients with glioblastoma multiforme. J Neurooncol. 2010;97:241–5. doi: 10.1007/s11060-009-0008-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Bihorel S, Camenisch G, Lemaire M, Scherrmann JM. Influence of breast cancer resistance protein (Abcg2) and p-glycoprotein (Abcb1a) on the transport of imatinib mesylate (Gleevec) across the mouse blood-brain barrier. J Neurochem. 2007;102:1749–57. doi: 10.1111/j.1471-4159.2007.04808.x. [DOI] [PubMed] [Google Scholar]
- 9.Leis JF, Stepan DE, Curtin PT, et al. Central nervous system failure in patients with chronic myelogenous leukemia lymphoid blast crisis and Philadelphia chromosome positive acute lymphoblastic leukemia treated with imatinib (STI-571). Leuk Lymphoma. 2004;45:695–8. doi: 10.1080/10428190310001625728. [DOI] [PubMed] [Google Scholar]
- 10.le Coutre P, Kreuzer KA, Pursche S, et al. Pharmacokinetics and cellular uptake of imatinib and its main metabolite CGP74588. Cancer Chemother Pharmacol. 2004;53:313–23. doi: 10.1007/s00280-003-0741-6. [DOI] [PubMed] [Google Scholar]
- 11.Neville K, Parise RA, Thompson P, et al. Plasma and cerebrospinal fluid pharmacokinetics of imatinib after administration to nonhuman primates. Clin Cancer Res. 2004;10:2525–9. doi: 10.1158/1078-0432.ccr-03-0155. [DOI] [PubMed] [Google Scholar]
- 12.Razis E, Selviaridis P, Labropoulos S, et al. Phase II study of a neoadjuvant imatinib in glioblastoma: evaluation of clinical and molecular effects of the treatment. Clin Cancer Res. 2009;15:6258–66. doi: 10.1158/1078-0432.CCR-08-1867. [DOI] [PubMed] [Google Scholar]
- 13.Chen Y, Agarwal S, Shaik NM, Chen C, Yang Z, Elmquist WF. P-glycoprotein and breast cancer resistance protein influence brain distribution of dasatinib. J Pharmacol Exp Ther. 2009;330:956–63. doi: 10.1124/jpet.109.154781. [DOI] [PubMed] [Google Scholar]
- 14.Porkka K, Koskenvesa P, Lundan T, et al. Dasatinib crosses the blood-brain barrier and is an efficient therapy for central nervous system Philadelphia chromosome-positive leukemia. Blood. 2008;112:1005–12. doi: 10.1182/blood-2008-02-140665. [DOI] [PubMed] [Google Scholar]
- 15.Final Report for Study No. B-5 (English Report): Tissue concentrations of radioactivity after a single oral administration of 14C-CML-187 to male rats. Nippon Shinyaku Co, Ltd.; Kyoto, Japan: Feb 03, 2005. [Google Scholar]
- 16.Asumi Y, Shinya K, Satohiro M, et al. INNO-406, a novel BCR-ABL/Lyn dual tyrosine kinase inhibitor, suppresses the growth of Ph + leukemia cells in the central nervous system, and cyclosporine A augments its in vivo activity. Blood. 2007;109:306–14. doi: 10.1182/blood-2006-03-013250. [DOI] [PubMed] [Google Scholar]
- 17.Dai H, Marbach P, Lemaire M, Hayes M, Elmquist WF. Distribution of STI-571 to the brain is limited by P-glycoprotein-mediated efflux. J Pharmacol Exp Ther. 2003;304:1085–92. doi: 10.1124/jpet.102.045260. [DOI] [PubMed] [Google Scholar]
- 18.Hamada A, Miyano H, Watanabe H, Saito H. Interaction of imatinib mesylate with human P-glycoprotein. J Pharmacol Exp Ther. 2003;307:824–8. doi: 10.1124/jpet.103.055574. [DOI] [PubMed] [Google Scholar]
- 19.Kantarjian H, le Coutre P, Cortes J, et al. Phase 1 study of INNO-406, a dual Abl/Lyn kinase inhibitor, in Philadelphia chromosome-positive leukemias after imatinib resistance or intolerance. Cancer. 2010;116:2665–72. doi: 10.1002/cncr.25079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Macdonald DR, Cascino TL, Schold SC, Jr, Cairncross JG. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8:1277–80. doi: 10.1200/JCO.1990.8.7.1277. [DOI] [PubMed] [Google Scholar]
- 21.Portnow J, Badie B, Chen J, Liu A, Blanchard S, Synold TW. The neuropharmacokinetics of temozolomide in patients with resectable brain tumors: potential implications for the current approach to chemoradiation. Clin Cancer Res. 2009;15:7092–8. doi: 10.1158/1078-0432.CCR-09-1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.de Lange EC, de Boer BA, Breimer DD. Microdialysis for pharmacokinetic analysis of drug transport to the brain. Adv Drug Deliv Rev. 1999;36:211–27. doi: 10.1016/s0169-409x(98)00089-1. [DOI] [PubMed] [Google Scholar]
- 23.Blakeley J, Portnow J. Microdialysis for assessing intratumoral drug disposition in brain cancers: a tool for rational drug development. Expert Opin Drug Metab Toxicol. 2010;6:1477–91. doi: 10.1517/17425255.2010.523420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Bafetinib Investigator's Brochure. (Edition 5) 2010 Jan; [Google Scholar]
- 25.Bauer M, Zeitlinger M, Karch R, et al. Pgp-mediated interaction between (R)-[11C]verapamil and tariquidar at the human blood-brain barrier: a comparison with rat data. Clin Pharmacol Ther. 2012;91:227–33. doi: 10.1038/clpt.2011.217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Uchida Y, Ohtsuki S, Katsukura Y, et al. Quantitative targeted absolute proteomics of human blood-brain barrier transporters and receptors. J Neurochem. 2011;117:333–45. doi: 10.1111/j.1471-4159.2011.07208.x. [DOI] [PubMed] [Google Scholar]
- 27.Syvanen S, Lindhe O, Palner M, et al. Species differences in blood-brain barrier transport of three positron emission tomography radioligands with emphasis on P-glycoprotein transport. Drug Metab Dispos. 2009;37:635–43. doi: 10.1124/dmd.108.024745. [DOI] [PubMed] [Google Scholar]
- 28.Chu J, Gallo JM. Application of microdialysis to characterize drug disposition in tumors. Adv Deliv Rev. 2000;45:243–53. doi: 10.1016/s0169-409x(00)00115-0. [DOI] [PubMed] [Google Scholar]
- 29.Zhou Q, Gallo JM. In vivo microdialysis for PK and PD studies of anticancer drugs. AAPS J. 2005;7:E659–67. doi: 10.1208/aapsj070366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Wang Y, Wong SL, Sawchuk RJ. Microdialysis calibration using retrodialysis and zero-net flux: application to a study of the distribution of zidovudine to rabbit cerebrospinal fluid and thalamus. Pharm Res. 1993;10:1411–19. doi: 10.1023/a:1018906821725. [DOI] [PubMed] [Google Scholar]
- 31.Bergenheim AT, Roslin M, Ungerstedt U, Waldenstrom A, Henriksson R, Ronquist G. Metabolic manipulation of glioblastoma in vivo by retrograde microdialysis of L-2, 4 diaminobutyric acid (DAB). J Neuroooncol. 2006;80:285–93. doi: 10.1007/s11060-006-9186-1. [DOI] [PubMed] [Google Scholar]
- 32.Blakeley JO, Olson J, Grossman SA, He X, Weingart J, Supko JG. Effect of blood brain barrier permeability in recurrent high grade gliomas on the intratumoral pharmacokinetics of methotrexate: a microdialysis study. J Neurooncol. 2009;91:51–8. doi: 10.1007/s11060-008-9678-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Raymond E, Brandes AA, Dittrich C, et al. 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;26:4659–65. doi: 10.1200/JCO.2008.16.9235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wen PY, Yung WK, Lamborn KR, et al. Phase I/II study of imatinib mesylate for recurrent malignant gliomas: North American Brain Tumor Consortium Study 99-08. Clin Cancer Res. 2006;12:4899–907. doi: 10.1158/1078-0432.CCR-06-0773. [DOI] [PubMed] [Google Scholar]
- 35.Reardon DA, Egorin MJ, Quinn JA, et al. Phase II study of imatinib mesylate plus hydroxyurea in adults with recurrent glioblastoma multiforme. J Clin Oncol. 2005;23:9359–68. doi: 10.1200/JCO.2005.03.2185. [DOI] [PubMed] [Google Scholar]
- 36.Reardon DA, Dresemann G, Taillibert S, et al. Multicentre phase II studies evaluating imatinib plus hydroxyurea in patients with progressive glioblastoma. Br J Cancer. 2009;15:1995–2004. doi: 10.1038/sj.bjc.6605411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Desjardins A, Quinn JA, Vredenburgh JJ, et al. Phase II study of imatinib mesylate and hydroxyurea for recurrent grade III malignant gliomas. J Neurooncol. 2007;83:53–60. doi: 10.1007/s11060-006-9302-2. [DOI] [PubMed] [Google Scholar]
- 38.Dresemann G, Weller M, Rosenthal MA, et al. Imatinib in combination with hydroxyurea versus hydroxyurea alone as oral therapy in patients with progressive pretreated glioblastoma resistant to standard dose temozolomide. J Neurooncol. 2010;96:393–402. doi: 10.1007/s11060-009-9976-3. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



