Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Oct 1.
Published in final edited form as: Cancer. 2012 Feb 27;118(19):4759–4767. doi: 10.1002/cncr.26541

Phase II study of Gleevec® plus hydroxyurea in adults with progressive or recurrent low-grade glioma1

David A Reardon 3,4,2, Annick Desjardins 2,5, James J Vredenburgh 2,5, James E Herndon II 6, April Coan 6, Sridharan Gururangan 2,3,4, Katherine B Peters 2,5, Roger McLendon 2,7, Sith Sathornsumetee 8, Jeremy N Rich 9, Eric S Lipp 2,3, Dorothea Janney 2,5, Henry S Friedman 2,3,4
PMCID: PMC3748946  NIHMSID: NIHMS319338  PMID: 22371319

Abstract

Background

We evaluated the efficacy of imatinib plus hydroxyurea in patients with progressive/recurrent low-grade glioma.

Methods

A total of 64 patients with recurrent/progressive low-grade glioma were enrolled in this single-center study that stratified patients into astrocytoma and oligodendroglioma cohorts. All patients received 500 mg of hydroxyurea twice a day. Imatinib was administered at 400 mg per day for patients not on EIAEDs and at 500 mg twice a day if on EIAEDs. The primary endpoint was progression-free survival at 12 months (PFS-12) and secondary endpoints were safety, median progression-free survival and radiographic response rate.

Results

Thirty-two patients were enrolled into each cohort. Eleven patients (17%) had prior radiotherapy and 24 (38%) had received prior chemotherapy. The median PFS and PFS-12 were 11 months and 39%, respectively. Outcome did not differ between the histologic cohorts. No patient achieved a radiographic response. The most common grade 3 or greater adverse events were neutropenia (11%), thrombocytopenia (3%) and diarrhea (3%).

Conclusions

Imatinib plus hydroxyurea was well tolerated among recurrent/progressive LGG patients but this regimen demonstrated negligible anti-tumor activity.

Keywords: imatinib, glioma, platelet derived growth factor, astrocytoma, oligodendroglioma

Introducation

Low-grade gliomas (LGG), a histologically diverse group of progressively infiltrative primary CNS tumors including astrocytoma, oligodendroglioma and oligoastrocytoma, most commonly affect children and young to middle-aged adults. Although some historical reports have misnomered these tumors as benign, most likely due to growth rates as low as 3-5 mm/year,1, 2 LGG can extensively infiltrate and cause progressive neurologic deficits, marked cognitive impairment and personality changes as well as recalcitrant seizures. In addition, many will eventually transform to high-grade histology.3 Median survival for most LGG subtypes is 5-10 years.4, 5

Optimal therapy for LGG remains undefined. Maximum safe resection can palliate symptoms and improve prognosis,6 but is not curative. Among newly diagnosed patients, post-surgical treatment is typically reserved for newly diagnosed patients with poor prognostic features including age over 40 years, poor performance status, tumors that are large or unresectable and non-oligodendroglial histology.7, 8 For such patients, as well as those with progressive or recurrent tumors, radiotherapy has historically been regarded as appropriate initial therapy. Although radiotherapy successfully achieves tumor control in most cases, it is also associated with potential late sequellae including cognitive decline, neuro-endocrine dysfunction, radiation-induced necrosis, vasculopathy, cortical atrophy, secondary malignancy and diminished quality of life.9-15 Due to these concerns, several chemotherapy options have been increasingly evaluated for LGG patients including PCV (procarbazine, carmustine and vincristine), carboplatin and temozolomide.16-24 Chemotherapy strategies have demonstrated sufficient benefit to generate ongoing, randomized phase III studies.

A general therapeutic strategy of significant interest in the current oncology era is the inhibition of biologically relevant growth factors and cell signaling mediators. Platelet-derived growth factor receptor-α (PDGFR-α) expression is increased in some LGG.25-29 In addition, PDGFR signaling is linked with several aspects of glioma biology including astrocytoma proliferation30 and neural stem/progenitor cell transformation.31

In the current manuscript we describe the first evaluation of a targeted therapeutic in the treatment of LGG. Imatinib mesylate (Gleevec®, formerly STI-571), a selective receptor tyrosine kinase inhibitor of PDGF receptors, Bcr-Abl, c-KIT and c-fms, has established activity against both hematologic and solid organ cancers.32 We performed a phase II study to evaluate the anti-tumor activity as well as safety of imatinib administered in combination with hydroxyurea, a ribonucleoside diphosphate reductase inhibitor, for patients with recurrent or progressive LGG.

Our results demonstrate that imatinib plus hydroxyurea is well tolerated but has negligible activity for patients with progressive or recurrent LGG.

Patients and Methods

Study design and treatment

We conducted an open-label, single center, phase II trial. The primary end point was 12-month progression-free survival (PFS) while secondary end points were overall survival (OS), median PFS, objective radiographic response rate and safety. Two patient cohorts, distinguished by tumor histology, were enrolled in parallel. Patients in cohort A had astrocytoma including the following histologic subtypes: fibrillary, gemistocytic, protoplasmic and pleomorphic xanthoastrocytoma. Patients in cohort B had oligodendroglioma or oligoastrocytoma. The dose of imatinib differed to account for the effect of CYP3A-inducing anti-epileptic drugs (EIAEDs) on imatinib metabolism33, 34: patients not on EIAEDs received 400mg once a day, while those on EIAEDs received 500mg twice a day. All patients received 500 mg of hydroxyurea twice a day. Medically appropriate efforts were used to maintain study-specific EIAED exposure for all patients.

Patients were assessed every 8 weeks and remained on study unless they withdrew consent, developed tumor progression or unacceptable toxicity.

Patient eligibility

Adult patients (≥ 18 years of age) were required to have histologically confirmed LGG that was progressive or recurrent based on radiographic or clinical features, the latter defined as new/worsening neurologic deficits excluding seizures. Patients with intrinsic optic pathway tumors were eligible without histologic confirmation as long as one of the following criteria were met: progressive vision loss documented by ophthalmologic examination; worsened proptosis; ≥ 2 mm increase in optic nerve diameter; or increased tumor distribution within the optic tract or optic radiations on neuroimaging. Measurable disease was required for all patients. Satisfactory hematologic (hemoglobin ≥ 10 g/dL, absolute neutrophil count (ANC) > 1500 cells/L, platelets > 100,000 cells/L), biochemical (serum creatinine < 1.5 mg/dL, BUN < 25 mg/dL, AST and bilirubin < 1.5 × upper limit of normal) and performance status (Karnofsky ≥ 60%) parameters were also required. Patients were required to be at least 2 weeks from prior surgery and at least 4 weeks from prior radiation or chemotherapy. All patients provided informed consent.

Key exclusion criteria included: prior imatinib or PDGFR-directed therapy; prior progressive disease or grade ≥ 3 toxicity to hydroxyurea treatment; grade ≥ 2 peripheral edema; pulmonary, pericardial or peritoneal effusions of any grade; patients with an excessive risk of an intracranial hemorrhage, or who had evidence of hemorrhage on pre-treatment MRI unless it was stable and grade 1; and concurrent warfarin administration. There was no limit on the number of prior therapies or episodes of progressive disease.

Assessments

Disease status was assessed after every other cycle using a modified version of the Macdonald criteria,35 to classify neurologic, corticosteroid and radiologic changes. In addition, the numeric cutoffs defined in the Macdonald criteria were also applied to the evaluation of the T2 or fluid attenuated inversion recovery (FLAIR) changes associated with the tumor to define response.

Safety assessments included weekly complete blood counts and monthly chemistry profiles. Adverse events were classified according to the Common Terminology Criteria for Adverse Events (CTCAE), version 3.0.

Treatment Modifications

Initiation of each 28-day treatment cycle required an ANC ≥ 1000/mm3, a platelet count of ≥ 100,000/mm3, SGOT ≤ 2.5 times the upper institutional norm, and a creatinine ≤ 1.5 times the upper institutional norm. Dose modifications were implemented for unacceptable toxicity defined as grade 4 neutropenia, grade ≥ 3 thrombocytopenia, grade ≥ 3 non-hematologic toxicity not attributable to underlying disease, concurrent medication or co-morbid event, or any toxicity requiring > two weeks to resolve to grade 1 or re-treatment criteria. Hydroxyurea was decreased to 500 mg once a day for the first episode of unacceptable hematologic toxicity. Thereafter additional episodes of unacceptable hematologic toxicity were treated with a decrease in total daily imatinib dosing of 100 mg/day for patients not on EIAEDs and 200 mg/day for patients on EIAEDs. For other unacceptable toxicities, the hydroxyurea dose was decreased by 250 mg/day and the dose of imatinib was decreased by 100 mg/day for patients not on EIAEDs and 200 mg/day for patients on EIAEDs.

Statistical considerations

The primary endpoint of the current study was progression-free survival at 12 months (PFS-12) for each cohort. Secondary endpoints included time to progression, overall survival (OS), objective response rate as well as safety and tolerability. Given a 12-PFS rate of 76% (95% confidence interval [CI]: 63% to 92%) for recurrent/progressive LGG patients treated with the standard 5-day regimen of temozolomide,20 a sample size goal of 32 patients per cohort was chosen to allow 90% power to differentiate between 12-PFS rates of 65% and 85% with a type I error rate of 0.082.

A “stopping rule” for unacceptable toxicity was incorporated for each stratum. Specifically, if 6 or more of the first 16 patients per stratum experienced unacceptable toxicity as defined above, further accrual would be suspended. In addition, if 9 or more of the total 32 patients experienced unacceptable toxicity, the treatment regimen would be considered to have an unacceptable toxicity profile. The type I and II error rates associated with this testing were 0.053 and 0.053, respectively.

Progression-free survival (PFS) and overall survival (OS) were measured from the cycle 1 start date and summarized using Kaplan-Meier estimator including 95% CIs.

Results

Patient characteristics

Sixty-four patients were enrolled between August 2005 and February 2008. Characteristics of patients at enrollment (Table 1) were comparable between the two cohorts except that oligodendroglioma patients were slightly older and were twice as long from the date of original diagnosis to study enrollment compared to the astrocytoma patients. In addition, a slightly higher percentage of oligodendroglioma patients had received prior chemotherapy. Approximately one-third of each cohort was on EIAEDs and approximately 17% of each cohort had received prior radiotherapy. A small subset of each cohort underwent either stereotactic biopsy or subtotal resection prior to enrollment and initiation of study therapy in order to confirm persistent LGG histology and demonstrate that these patients had not transformed to high-grade histology.

Table 1. Patient characteristics.

Cohort A1 (n=32) Cohort B1 (n = 32)
Age (median; years) 39.3 46.2
 Range (years) 24.3 – 83.8 22.6 – 67.2
Gender
 Male 20 (63) 19 (60)
 Female 12 (38) 13 (40)
Histology
 Astrocytoma 32 (100) 0
 Oligodendroglioma 0 28 (88)
 Mixed Oligoastrocytoma 0 4 (12)
KPS
 91-100 18 (56) 21 (66)
 80 10 (31) 9 (28)
 60 – 70 4 (13) 2 (6)
AED Status
 EIAED 10 (31) 11 (34)
 Non EIAED 14 (44) 18 (56)
 No EIAED 8 (25) 3 (9)
Surgery, Prior to Enrollment
 Biopsy 4 (13) 9 (28)
 STR 0 2 (6)
 GTR 0 0
Time from Original Diagnosis
 Median (years) 2.53 5.84
 Range (years 0.03 – 22.21 0.04 – 14.54
Prior Conventional XRT 5 (16) 6 (18)
Stereotactic Radiosurgery 0 1 (3)
Prior Chemotherapy
 Temozolomide 10 (31) 11 (34)
 Carboplatin 0 1 (3)
 PCV 0 1 (3)
 BCNU Wafers 0 1 (3)
Number Prior Episodes of PD
 0 3 (9) 0
 1 23 (72) 21 (66)
 2 6 (19) 9 (28)
 ≥ 3 2(6)
Tumor > 6 cm 22 (69) 28 (88)
Tumor crossing midline 15 (47) 16 (50)
Presence of contrast enhancement 13 (41) 16 (50)
1

A: Astrocytoma cohort; B: oligodendroglioma cohort

Abbreviations: AED, anti-epileptic drugs; BCNU, carmustine; EIAED, CYP3A enzyme-inducing anti-epileptic drugs; GTR, gross total resection; KPS, Karnofsky performance status; PCV, procarbazine, carmustine and vincristine; PD, progressive disease; STR, subtotal resection; XRT, radiation therapy

Toxicity

Seven-hundred and fifty-seven cycles of therapy were administered as of April 1, 2011. The adverse events seen in the study were as expected for this population and this class of therapy. They were mostly mild and transient and gave no indication of target organ toxicity. Table2 summarizes adverse events felt to be at least possibly attributable to the study regimen. Of note, the frequency and severity of adverse events did not differ between patient cohorts or between patients on and not on EIAEDs. Three patients (4.6%) were taken off study therapy due to toxicity including two patients with grade 4 neutropenia and one patient with asymptomatic, grade 1 CNS hemorrhage noted on surveillance MRI. Most adverse events were grade 2. Among grade 3 events, single patients experienced anemia, fatigue, infection and weight loss while diarrhea and thrombocytopenia affected two patients each and five patients experienced neutropenia. Grade 4 events were limited to neutropenia (n=2) and diarrhea (n=1), respectively. There were no grade 5 attributable adverse events. Dose modification was required in five patients including 2 astrocytoma patients and three oligodendroglioma patients; of note, all five of these patients were not on EIAEDs.

Table 2. Summary of treatment toxicity.

Toxicity Cohort1 Grade 2 Grade 3 Grade 4
A B A B A B

Anemia 2 (6) 1 (3) 1 (3)
Anorexia 2 (6) 2 (6)
Creatinine increase 1 (3)
Diarrhea 5 (16) 3 (9) 1 (3) 1 (3) 1 (3)
Edema 2 (6)
Fatigue 12 (38) 13 (41) 1 (3)
Infection 4 (12) 3 (9) 1 (3)
Mucositis 1 (3)
Nausea 10 (31) 5 (16) 1 (3) 1 (3)
Neutropenia 2 (6) 2 (6) 3 (9) 1 (3) 1 (3)
Rash 5 (16) 2 (6)
Thrombocytopenia 2 (6)
Transaminase elevation 1 (3)
Weight loss 1 (3)
*

numbers in parentheses indicate percentage of treated patients

1

A: astrocytoma cohort; B: oligodendroglioma cohort

Outcome

Table 3 summarizes study outcome. As of April 1, 2011, median follow-up for all patients was 180 weeks and all patients have discontinued study therapy except for 1 oligodendroglioma patient who continues on treatment in cycle 51. Thirty-four patients (53%) elected to discontinue study therapy with stable disease while 27 patients (42%) came off study due to progressive disease. Three patients (5%) were taken off study therapy due to toxicity (discussed above).

Table 3. Summary of outcome.

Astrocytoma (n=32) Oligodendroglioma (n=32) All patients (n=64)
Median follow-up (weeks) 191.0 (174.0; 203.7)* 176.0 (147.0; 190.3) 179.9 (171.4; 192.1)
Progression-free survival
Median (weeks) 43.5 (31.7, 63.7) 43.3 (24.9, 53.6) 43.5 (33.0, 53.6)
12 months (%) 43.8 (26.5, 59.8) 34.4 (18.8, 50.6) 39.1 (27.2, 50.7)
24 months (%) 21.9 (9.6, 37.2) 21.9 (9.6, 37.2) 21.9 (12.7, 32.6)
36 months (%) 9.4 (2.4, 22.3) 15.6 (5.7, 30.0) 12.5 (5.8, 21.8)
Overall survival
Median (weeks) Not estimable Not estimable Not estimable
12 months (%) 93.8 (77.3, 98.4) 100 96.9 (88.1, 99.2)
24 months (%) 87.5 (70.0, 95.1) 90.6 (73.7, 96.9) 89.1 (78.4, 94.6)
36 months (%) 74.7 (55.7, 86.5) 75.4 (54.6, 87.7) 75.5 (62.6, 84.5)
*

Data in parentheses indicates 95% confidence intervals unless otherwise specified

Median PFS and PFS-12 for all patients were 43.5 weeks (95% CI: 33.0, 53.6) and 39.1% (95% CI: 27.2, 50.7). Of note, PFS did not differ between patients with astrocytoma or those with oligodendroglioma. Median OS was not reached for either study cohort. Best radiographic response was stable disease and none of the patients achieved a radiographic response. More than one episode of prior progression was associated with poorer PFS (p=0.005), while the presense of contrast enhancement had a borderline effect (p=0.07). In contrast, PFS was not related to age less than 40 years, KPS ≥ 90, maximum tumor unidimensional size greater than 6 centimeters, crossing the midline, oligodendroglioma histology or the concurrent administration of EIAEDs (Table 4).

Table 4. Assessment of factors relative to progression-free survival.

Group Total # Failed Median survival in weeks (95% CI) 6-month survival (95% CI) 12-month survival (95% CI) 24-month survival (95% CI) Logrank p-value
Maximum tumor size ≤ 6cm 16 15 46.6 (8.1, 63.7) 68.8% (40.5%, 85.6%) 43.8% (19.8%, 65.6%) 12.5% (2.1%, 32.8%) 0.6061
Maximum tumor size > 6cm 48 43 41.4 (31.7, 53.6) 68.8% (53.6%, 79.8%) 37.5% (24.1%, 50.9%) 25% (13.9%, 37.8%)
No tumor crossing the midline 33 28 49 (31.9, 64.3) 72.7% (54.1%, 84.8%) 45.5% (28.2%, 61.2%) 30.3% (15.9%, 46.1%) 0.0917
Tumor crossing the midline 31 30 41 (24, 44.9) 64.5% (45.2%, 78.5%) 32.3% (16.9%, 48.6%) 12.9% (4.1%, 27%)
No presence of contrast 35 30 51.4 (41, 63.7) 80% (62.6%, 89.9%) 48.6% (31.4%, 63.7%) 28.6% (14.9%, 43.8%) 0.0673
Presence of contrast 29 28 37.7 (23.6, 46.4) 55.2% (35.6%, 71%) 27.6% (13.1%, 44.3%) 13.8% (4.3%, 28.6%)
Age ≥ 40 years 38 34 45.9 (31.7, 56) 68.4% (51.1%, 80.7%) 42.1% (26.4%, 57%) 26.3% (13.7%, 40.8%) 0.3583
Age < 40 years 26 24 42.1 (24.6, 53.6) 69.2% (47.8%, 83.3%) 34.6% (17.5%, 52.5%) 15.4% (4.8%, 31.5%)
KPS < 90 25 23 43 (24.9, 55.9) 64% (42.2%, 79.4%) 32% (15.2%, 50.2%) 20% (7.3%, 37.2%) 0.5713
KPS ≥ 90 39 35 46.4 (31.7, 63.7) 71.8% (54.9%, 83.3%) 43.6% (27.9%, 58.3%) 23.1% (11.4%, 37.1%)
Astrocytoma 32 29 43.5 (31.7, 63.7) 71.9% (52.9%, 84.3%) 43.8% (26.5%, 59.8%) 21.9% (9.6%, 37.2%) 0.9434
Oligodendroglioma 32 29 43.3 (24.9, 53.6) 65.6% (46.6%, 79.3%) 34.4% (18.8%, 50.6%) 21.9% (9.6%, 37.2%)
No use of EIAEDs 43 39 44 (33, 56) 67.4% (51.3%, 79.3%) 41.9% (27.1%, 55.9%) 23.3% (12%, 36.6%) 0.8876
Use of EIAEDs 21 19 41 (24.6, 55.9) 71.4% (47.2%, 86%) 33.3% (14.9%, 53.1%) 19% (5.9%, 37.7%)
≤ 1 PD 49 43 49 (41.1, 63.7) 75.5% (60.9%, 85.3%) 46.9% (32.6%, 60%) 26.5% (15.2%, 39.3%) 0.0045
>1 PD 15 15 25.1 (15.6, 41) 46.7% (21.2%, 68.7%) 13.3% (2.2%, 34.6%) 6.7% (0.4%, 26%)

Abbreviations: CI, confidence interval; cm, centimeter; EIAED, CYP3A-enzyme inducing antiepileptic drugs; KPS, Karnofsky performance status; PD, progressive disease

Discussion

Low-grade glioma accounts for approximately 15% of adult primary central nervous system tumors. Large series have identified clinical factors associated with poor prognosis including age over 40 years, poor performance status, tumors that are large or unresectable and non-oligodendroglial histology.7 More recently biologic factors have been linked with outcome including histology,5 co-deletion of chromosomes 1p19q,36 methylation of methylguanine methyltransferase,19, 21 and IDH1/2 mutation.37-39

Beyond radiotherapy, optimal therapy for patients with poor prognostic factors or those with recurrent/progressive tumors have not been clearly identified. Several series have demonstrated that chemotherapy regimens such as PCV, and more recently including temozolomide, have activity in this setting.17-21, 40-45

The primary rationale for evaluating imatinib in this study is its inhibitory capacity against PDGFR. Low-grade gliomas, including both astrocytomas and oligodendrogliomas frequently express PDGFR while some of these tumors also express PDGF, implicating a potential autocrine and/or paracrine loop of dysregulated signaling.26-28, 46 Several important findings also implicate PDGFR signaling in LGG physiology47 including that exogenous PDGF induces astrocytoma proliferation30 and that PDGFR activation has been linked with transformation of neural stem/progenitor cells into glial tumors31, 48-51 In addition, PDGF dominant-negative mutants can revert the transformed phenotype of human astrocytoma cells.52

Imatinib has not been previously evaluated for patients with grade II glioma, however, preclinical in vitro assays have demonstrated activity against astrocytoma lines.53 In addition, imatinib was anecdotally reported to elicit marked regression of a recurrent pilocytic astrocytoma (WHO grade I) in a pediatric patient.54

Hydroxyurea, a ribonucleoside diphosphate reductase inhibitor, exerts a cell-cycle specific effect during the S-phase of mitosis, thereby blocking DNA synthesis. Although hydroxyurea has is not known to have single-agent activity against malignant gliomas, it has been incorporated into several therapeutic regimens for primary CNS tumor patients.55-59 Hydroxyurea exhibits rapid absorption from the gastrointestinal tract following oral dosing and readily crosses the blood-brain barrier. In the current study regimen for LGG patients, we hypothesized that the cytotoxicity of protracted hydroxyurea dosing may augment the anti-PDGFR activity of imatinib yielding enhanced anti-tumor activity than could be achieved by either agent alone. Of note, modest activity of imatinib plus hydroxyurea was initially observed among recurrent high-grade glioma patients;33, 60, 61 however, these results were not confirmed in subsequent multi-center studies.62, 63 Several factors may have contributed to these disappointing results including the relatively low level of PDGFR activation in primary GBM tumors.

In the current study, imatinib and hydroxyurea was well-tolerated among recurrent LGG patients. The majority of adverse events were grade 2 while grade 3 events were uncommon and grade 4 toxicities were rare. The toxicity profile of this regimen appears comparable to that reported for various temozolomide regimens among recurrent/progressive LGG patients. 19-21, 40, 41

However, outcome on the current study appears inferior to that reported for temozolomide. We observed no radiographic responses whereas the radiographic response rate reported among various temozolomide regimens is 20-61%.19-21, 24, 40, 41 One potential factor contributing to this difference may be differences in criteria used to define radiographic response. For example, we required a partial response to include a 50% reduction of the FLAIR bi-dimensional product as well as any enhancing disease, whereas some of the other studies only assessed enhancing tumor. The accurate ascertainment of tumor growth control can be challenging to achieve among tumors where the natural history reflects indolent growth, such as LGG. In addition, the use of a numeric cut-off to define progression, such as a 25% increase in tumor dimension, can be misleading in that this practice allows tumors to be classified as stable, when in fact, they are progressively growing. Recently published recommendations for response assessment among LGG patients provide guidelines that will help ensure optimal and consistent assessment approaches for future LGG studies.64

In addition, the median PFS and PFS-12 rate observed on our study, 11 months and 39% respectively, were lower than most of the results reported for temozolomide therapy (Table 4).19-21, 24, 40, 41 We evaluated several clinical factors previously shown to predict outcome for LGG patients and most were not associated with PFS, most likely reflecting the overall negative outcome on our study. Unfortunately, due to lack of archival tumor material, we were unable to assess whether PDGF/PDGFR expression or genetic abnormalities such as chromosome 1p/19q or IDH 1/2 mutations correlated with outcome. Another factor contributing to the low activity observed on the current study is that the distribution of imatinib into the brain in preclinical studies has been shown to be limited by P-glycoprotein mediated efflux.65 Although imatinib has been shown to achieve intratumoral concentrations that are comparable to those measured in plasma among malignant glioma patients where the blood-brain-barrier is relatively defective,66 the blood-brain barrier was likely intact for most of the patients in the current study given their low-grade histology. 67Although our study demonstrated that imatinib, combined with hydroxyurea had negligible anti-tumor activity, PDGF and PDGFR remain noteworthy potential therapeutic targets for recurrent/progressive LGG and studies evaluating other PDGF/PDGFR inhibitors should be considered.

Table 5.

Outcome on the current study relative to temozolomide studies for LGG.

Report (citation) Design Number of patients Failed prior XRT (%) Failed prior chemotherapy (%) ORR (%) Median PFS (months) PFS-12 (%)
Current study Prospective 64 11 (17) 24 (38) 0 11 39
Taal19 Prospective 58 58 (100) 0 25 (54)1 8 25
Kesari21 Prospective 44 12 (27) 0 9 (20) 38 91
Quinn20 Prospective 46 7 (15) 10 (22) 28 (61) 22 76
Tosoni41 Prospective 30 0 0 9 (30) 22 73
Pace40 Prospective 43 30 (70) 16 (37) 20 (47) 10 47
Kaloshi24 Retrospective 149 0 0 22 (15) 28 80
1

46 of 58 patients were evaluable for response;

Abbreviations: LGG, low-grade glioma; ORR, overall radiographic response rate; PFS, progression-free survival; PFS-12, progression-free survival at 12 months; XRT, radiation therapy;

Acknowledgments

In addition to the above investigators, we wish to thank Wendy Gentry for her assistance in the preparation of this manuscript.

Footnotes

Conflict of Interest Disclosre: The authors made no disclosures.

1

Grant Support: Grants 5 P50 NS20023-29 and 4 R37 CA011898-42 from NIH, Department of Health and Human Services, Bethesda, Maryland and a research grant from Novartis Pharmaceuticals.

References

  • 1.Mandonnet E, Delattre JY, Tanguy ML, et al. Continuous growth of mean tumor diameter in a subset of grade II gliomas. Ann Neurol. 2003;53:524–528. doi: 10.1002/ana.10528. [DOI] [PubMed] [Google Scholar]
  • 2.van den Bent MJ, Afra D, de Witte O, et al. Long-term efficacy of early versus delayed radiotherapy for low-grade astrocytoma and oligodendroglioma in adults: the EORTC 22845 randomised trial. Lancet. 2005;366:985–990. doi: 10.1016/S0140-6736(05)67070-5. [DOI] [PubMed] [Google Scholar]
  • 3.Wessels PH, Weber WE, Raven G, et al. Supratentorial grade II astrocytoma: biological features and clinical course. Lancet Neurology. 2003;2:395–403. doi: 10.1016/s1474-4422(03)00434-4. [DOI] [PubMed] [Google Scholar]
  • 4.Claus EB, Black PM. Survival rates and patterns of care for patients diagnosed with supratentorial low-grade gliomas: data from the SEER program, 1973-2001. Cancer. 2006;106:1358–1363. doi: 10.1002/cncr.21733. [DOI] [PubMed] [Google Scholar]
  • 5.Okamoto Y, Di Patre PL, Burkhard C, et al. Population-based study on incidence, survival rates, and genetic alterations of low-grade diffuse astrocytomas and oligodendrogliomas. Acta Neuropathol. 2004;108:49–56. doi: 10.1007/s00401-004-0861-z. [DOI] [PubMed] [Google Scholar]
  • 6.Smith JS, Chang EF, Lamborn KR, et al. Role of extent of resection in the long-term outcome of low-grade hemispheric gliomas. J Clin Oncol. 2008;26:1338–1345. doi: 10.1200/JCO.2007.13.9337. [DOI] [PubMed] [Google Scholar]
  • 7.Pignatti F, van den Bent M, Curran D, et al. Prognostic factors for survival in adult patients with cerebral low-grade glioma. J Clin Oncol. 2002;20:2076–2084. doi: 10.1200/JCO.2002.08.121. [DOI] [PubMed] [Google Scholar]
  • 8.Chang EF, Smith JS, Chang SM, et al. Preoperative prognostic classification system for hemispheric low-grade gliomas in adults. J Neurosurg. 2008;109:817–824. doi: 10.3171/JNS/2008/109/11/0817. [DOI] [PubMed] [Google Scholar]
  • 9.Douw L, Klein M, Fagel SS, et al. Cognitive and radiological effects of radiotherapy in patients with low-grade glioma: long-term follow-up. Lancet Neurology. 2009;8:810–818. doi: 10.1016/S1474-4422(09)70204-2. [DOI] [PubMed] [Google Scholar]
  • 10.Olson JD, Riedel E, DeAngelis LM. Long-term outcome of low-grade oligodendroglioma and mixed glioma. Neurology. 2000;54:1442–1448. doi: 10.1212/wnl.54.7.1442. [DOI] [PubMed] [Google Scholar]
  • 11.Klein M, Heimans JJ, Aaronson NK, et al. Effect of radiotherapy and other treatment-related factors on mid-term to long-term cognitive sequelae in low-grade gliomas: a comparative study. Lancet. 2002;360:1361–1368. doi: 10.1016/s0140-6736(02)11398-5. [DOI] [PubMed] [Google Scholar]
  • 12.Vigliani MC, Sichez N, Poisson M, et al. A prospective study of cognitive functions following conventional radiotherapy for supratentorial gliomas in young adults: 4-year results. Int J Radiat Oncol Biol Phys. 1996;35:527–533. doi: 10.1016/s0360-3016(96)80015-0. [DOI] [PubMed] [Google Scholar]
  • 13.Kiebert GM, Curran D, Aaronson NK, et al. Quality of life after radiation therapy of cerebral low-grade gliomas of the adult: results of a randomised phase III trial on dose response (EORTC trial 22844) EORTC Radiotherapy Co-operative Group. Eur J Cancer. 1998;34:1902–1909. doi: 10.1016/s0959-8049(98)00268-8. [DOI] [PubMed] [Google Scholar]
  • 14.Surma-aho O, Niemela M, Vilkki J, et al. Adverse long-term effects of brain radiotherapy in adult low-grade glioma patients. Neurology. 2001;56:1285–1290. doi: 10.1212/wnl.56.10.1285. [DOI] [PubMed] [Google Scholar]
  • 15.Brown PD, Buckner JC, Uhm JH, et al. The neurocognitive effects of radiation in adult low-grade glioma patients. Neuro Oncol. 2003;5:161–167. doi: 10.1215/S1152-8517-02-00043-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wen PY, DeAngelis LM. Chemotherapy for low-grade gliomas: emerging consensus on its benefits. Neurology. 2007;68:1762–1763. doi: 10.1212/01.wnl.0000266866.13748.a9. [DOI] [PubMed] [Google Scholar]
  • 17.Buckner JC, Gesme D, Jr, O'Fallon JR, et al. Phase II trial of procarbazine, lomustine, and vincristine as initial therapy for patients with low-grade oligodendroglioma or oligoastrocytoma: efficacy and associations with chromosomal abnormalities. J Clin Oncol. 2003;21:251–255. doi: 10.1200/JCO.2003.06.023. [DOI] [PubMed] [Google Scholar]
  • 18.Hoang-Xuan K, Capelle L, Kujas M, et al. Temozolomide as initial treatment for adults with low-grade oligodendrogliomas or oligoastrocytomas and correlation with chromosome 1p deletions. J Clin Oncol. 2004;22:3133–3138. doi: 10.1200/JCO.2004.10.169. [DOI] [PubMed] [Google Scholar]
  • 19.Taal W, Dubbink HJ, Zonnenberg CB, et al. First-line temozolomide chemotherapy in progressive low-grade astrocytomas after radiotherapy: molecular characteristics in relation to response. Neuro Oncol. 2011;13:235–241. doi: 10.1093/neuonc/noq177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Quinn JA, Reardon DA, Friedman AH, et al. Phase II trial of temozolomide in patients with progressive low-grade glioma. J Clin Oncol. 2003;21:646–651. doi: 10.1200/JCO.2003.01.009. [DOI] [PubMed] [Google Scholar]
  • 21.Kesari S, Schiff D, Drappatz J, et al. Phase II study of protracted daily temozolomide for low-grade gliomas in adults. Clin Cancer Res. 2009;15:330–337. doi: 10.1158/1078-0432.CCR-08-0888. [DOI] [PubMed] [Google Scholar]
  • 22.Gururangan S, Cavazos CM, Ashley D, et al. Phase II study of carboplatin in children with progressive low-grade gliomas. J Clin Oncol. 2002;20:2951–2958. doi: 10.1200/JCO.2002.12.008. [DOI] [PubMed] [Google Scholar]
  • 23.Peyre M, Cartalat-Carel S, Meyronet D, et al. Prolonged response without prolonged chemotherapy: a lesson from PCV chemotherapy in low-grade gliomas. Neuro Oncol. 2010;12:1078–1082. doi: 10.1093/neuonc/noq055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Kaloshi G, Benouaich-Amiel A, Diakite F, et al. Temozolomide for low-grade gliomas: predictive impact of 1p/19q loss on response and outcome. Neurology. 2007;68:1831–1836. doi: 10.1212/01.wnl.0000262034.26310.a2. [DOI] [PubMed] [Google Scholar]
  • 25.Nister M, Enblad P, Backstrom G, et al. Platelet-derived growth factor (PDGF) in neoplastic and non-neoplastic cystic lesions of the central nervous system and in the cerebrospinal fluid. Br J Cancer. 1994;69:952–956. doi: 10.1038/bjc.1994.184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Guha A, Dashner K, Black PM, et al. Expression of PDGF and PDGF receptors in human astrocytoma operation specimens supports the existence of an autocrine loop. Int J Cancer. 1995;60:168–173. doi: 10.1002/ijc.2910600206. [DOI] [PubMed] [Google Scholar]
  • 27.Hermanson M, Funa K, Hartman M, et al. 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]
  • 28.Martinho O, Longatto-Filho A, Lambros MB, et al. 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]
  • 29.Di Rocco F, Carroll RS, Zhang J, et al. Platelet-derived growth factor and its receptor expression in human oligodendrogliomas. Neurosurgery. 1998;42:341–346. doi: 10.1097/00006123-199802000-00080. [DOI] [PubMed] [Google Scholar]
  • 30.Ranza E, Facoetti A, Morbini P, et al. Exogenous platelet-derived growth factor (PDGF) induces human astrocytoma cell line proliferation. Anticancer Res. 2007;27:2161–2166. [PubMed] [Google Scholar]
  • 31.Jackson EL, Garcia-Verdugo JM, Gil-Perotin S, et al. PDGFR alpha-positive B cells are neural stem cells in the adult SVZ that form glioma-like growths in response to increased PDGF signaling. Neuron. 2006;51:187–199. doi: 10.1016/j.neuron.2006.06.012. [DOI] [PubMed] [Google Scholar]
  • 32.Waller CF. Imatinib mesylate. Recent Results Cancer Res. 2010;184:3–20. doi: 10.1007/978-3-642-01222-8_1. [DOI] [PubMed] [Google Scholar]
  • 33.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;23:9359–9368. doi: 10.1200/JCO.2005.03.2185. [DOI] [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;12:4899–4907. doi: 10.1158/1078-0432.CCR-06-0773. [DOI] [PubMed] [Google Scholar]
  • 35.Macdonald DR, Cascino TL, Schold SC, Jr, et al. Response criteria for phase II studies of supratentorial malignant glioma. Journal of Clinical Oncology. 1990;8:1277–1280. doi: 10.1200/JCO.1990.8.7.1277. [DOI] [PubMed] [Google Scholar]
  • 36.Jenkins RB, Blair H, Ballman KV, et al. A t(1;19)(q10;p10) mediates the combined deletions of 1p and 19q and predicts a better prognosis of patients with oligodendroglioma. Cancer Res. 2006;66:9852–9861. doi: 10.1158/0008-5472.CAN-06-1796. [DOI] [PubMed] [Google Scholar]
  • 37.Dubbink HJ, Taal W, van Marion R, et al. IDH1 mutations in low-grade astrocytomas predict survival but not response to temozolomide. Neurology. 2009;73:1792–1795. doi: 10.1212/WNL.0b013e3181c34ace. [DOI] [PubMed] [Google Scholar]
  • 38.Houillier C, Wang X, Kaloshi G, et al. IDH1 or IDH2 mutations predict longer survival and response to temozolomide in low-grade gliomas. Neurology. 2010;75:1560–1566. doi: 10.1212/WNL.0b013e3181f96282. [DOI] [PubMed] [Google Scholar]
  • 39.Sanson M, Marie Y, Paris S, et al. Isocitrate dehydrogenase 1 codon 132 mutation is an important prognostic biomarker in gliomas. J Clin Oncol. 2009;27:4150–4154. doi: 10.1200/JCO.2009.21.9832. [DOI] [PubMed] [Google Scholar]
  • 40.Pace A, Vidiri A, Galie E, et al. Temozolomide chemotherapy for progressive low-grade glioma: clinical benefits and radiological response. Ann Oncol. 2003;14:1722–1726. doi: 10.1093/annonc/mdg502. [DOI] [PubMed] [Google Scholar]
  • 41.Tosoni A, Franceschi E, Ermani M, et al. Temozolomide three weeks on and one week off as first line therapy for patients with recurrent or progressive low grade gliomas. J Neurooncol. 2008;89:179–185. doi: 10.1007/s11060-008-9600-y. [DOI] [PubMed] [Google Scholar]
  • 42.Brada M, Viviers L, Abson C, et al. Phase II study of primary temozolomide chemotherapy in patients with WHO grade II gliomas. Ann Oncol. 2003;14:1715–1721. doi: 10.1093/annonc/mdg371. [DOI] [PubMed] [Google Scholar]
  • 43.Brandes AA, Basso U, Vastola F, et al. Carboplatin and teniposide as third-line chemotherapy in patients with recurrent oligodendroglioma or oligoastrocytoma: a phase II study. Ann Oncol. 2003;14:1727–1731. doi: 10.1093/annonc/mdg494. [DOI] [PubMed] [Google Scholar]
  • 44.Galanis E, Buckner JC, Burch PA, et al. Phase II trial of nitrogen mustard, vincristine, and procarbazine in patients with recurrent glioma: North Central Cancer Treatment Group results. J Clin Oncol. 1998;16:2953–2958. doi: 10.1200/JCO.1998.16.9.2953. [DOI] [PubMed] [Google Scholar]
  • 45.Soffietti R, Nobile M, Ruda R, et al. Second-line treatment with carboplatin for recurrent or progressive oligodendroglial tumors after PCV (procarbazine, lomustine, and vincristine) chemotherapy: a phase II study. Cancer. 2004;100:807–813. doi: 10.1002/cncr.20042. [DOI] [PubMed] [Google Scholar]
  • 46.Maxwell M, Naber SP, Wolfe HJ, et al. Coexpression of platelet-derived growth factor (PDGF) and PDGF-receptor genes by primary human astrocytomas may contribute to their development and maintenance. J Clin Invest. 1990;86:131–140. doi: 10.1172/JCI114675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Calzolari F, Malatesta P. Recent insights into PDGF-Induced gliomagenesis. Brain Pathol. 20:527–538. doi: 10.1111/j.1750-3639.2009.00335.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Assanah M, Lochhead R, Ogden A, et al. Glial progenitors in adult white matter are driven to form malignant gliomas by platelet-derived growth factor-expressing retroviruses. J Neurosci. 2006;26:6781–6790. doi: 10.1523/JNEUROSCI.0514-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Shih AH, Holland EC. Platelet-derived growth factor (PDGF) and glial tumorigenesis. Cancer Lett. 2006;232:139–147. doi: 10.1016/j.canlet.2005.02.002. [DOI] [PubMed] [Google Scholar]
  • 50.Shih AH, Dai C, Hu X, et al. 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]
  • 51.Dai C, Celestino JC, Okada Y, et al. PDGF autocrine stimulation dedifferentiates cultured astrocytes and induces oligodendrogliomas and oligoastrocytomas from neural progenitors and astrocytes in vivo. Genes Dev. 2001;15:1913–1925. doi: 10.1101/gad.903001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Shamah SM, Stiles CD, Guha A. Dominant-negative mutants of platelet-derived growth factor revert the transformed phenotype of human astrocytoma cells. Mol Cell Biol. 1993;13:7203–7212. doi: 10.1128/mcb.13.12.7203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Ranza E, Bertolotti A, Facoetti A, et al. Influence of imatinib mesylate on radiosensitivity of astrocytoma cells. Anticancer Res. 2009;29:4575–4578. [PubMed] [Google Scholar]
  • 54.McLaughlin ME, Robson CD, Kieran MW, et al. Marked regression of metastatic pilocytic astrocytoma during treatment with imatinib mesylate (STI-571, Gleevec): a case report and laboratory investigation. J Pediatr Hematol Oncol. 2003;25:644–648. doi: 10.1097/00043426-200308000-00012. [DOI] [PubMed] [Google Scholar]
  • 55.Geyer JR, Zeltzer PM, Boyett JM, et al. Survival of infants with primitive neuroectodermal tumors or malignant ependymomas of the CNS treated with eight drugs in 1 day: a report from the Childrens Cancer Group. J Clin Oncol. 1994;12:1607–1615. doi: 10.1200/JCO.1994.12.8.1607. [DOI] [PubMed] [Google Scholar]
  • 56.Kaba SE, Kyritsis AP, Hess K, et al. TPDC-FuHu chemotherapy for the treatment of recurrent metastatic brain tumors. J Clin Oncol. 1997;15:1063–1070. doi: 10.1200/JCO.1997.15.3.1063. [DOI] [PubMed] [Google Scholar]
  • 57.Kyritsis AP, Yung WK, Jaeckle KA, et al. Combination of 6-thioguanine, procarbazine, lomustine, and hydroxyurea for patients with recurrent malignant gliomas. Neurosurgery. 1996;39:921–926. doi: 10.1097/00006123-199611000-00006. [DOI] [PubMed] [Google Scholar]
  • 58.Levin VA. The place of hydroxyurea in the treatment of primary brain tumors. Semin Oncol. 1992;19:34–39. [PubMed] [Google Scholar]
  • 59.Prados MD, Larson DA, Lamborn K, et al. Radiation therapy and hydroxyurea followed by the combination of 6-thioguanine and BCNU for the treatment of primary malignant brain tumors. Int J Radiat Oncol Biol Phys. 1998;40:57–63. doi: 10.1016/s0360-3016(97)00566-x. [DOI] [PubMed] [Google Scholar]
  • 60.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]
  • 61.Dresemann G. Imatinib and hydroxyurea in pretreated progressive glioblastoma multiforme: a patient series. Ann Oncol. 2005;16:1702–1708. doi: 10.1093/annonc/mdi317. [DOI] [PubMed] [Google Scholar]
  • 62.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]
  • 63.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;101:1995–2004. doi: 10.1038/sj.bjc.6605411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.van den Bent M, Wefel J, Schiff D, et al. Response assessment in neuro-oncology (a report of the RANO group): assessment of outcome in trials of diffuse low-grade gliomas. Lancet Oncol. 2011 doi: 10.1016/S1470-2045(11)70057-2. [DOI] [PubMed] [Google Scholar]
  • 65.Dai H, Marbach P, Lemaire M, et al. Distribution of STI-571 to the brain is limited by P-glycoprotein-mediated efflux. J Pharmacol Exp Ther. 2003;304:1085–1092. doi: 10.1124/jpet.102.045260. [DOI] [PubMed] [Google Scholar]
  • 66.Holdhoff M, Supko JG, Gallia GL, et al. Intratumoral concentrations of imatinib after oral administration in patients with glioblastoma multiforme. J Neurooncol. 2010;97:241–245. doi: 10.1007/s11060-009-0008-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.U.S. Department of Health and Human Services CfDE, and Research FaDa: FDA Project on Cancer Drug Aproval Endpoint.

RESOURCES