19.11.2014 Views

The Outcomes of Concomitant Radiation Plus Temozolomide ... - NCI

The Outcomes of Concomitant Radiation Plus Temozolomide ... - NCI

The Outcomes of Concomitant Radiation Plus Temozolomide ... - NCI

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

±IU W / 3<br />

Journal <strong>of</strong> the Egyptian Nat. Cancer Inst., Vol. 21, No. 2, June: 107-119, 2009<br />

<strong>The</strong> <strong>Outcomes</strong> <strong>of</strong> <strong>Concomitant</strong> <strong>Radiation</strong> <strong>Plus</strong> <strong>Temozolomide</strong><br />

Followed by Adjuvant <strong>Temozolomide</strong> for Newly Diagnosed<br />

High Grade Gliomas: <strong>The</strong> Preliminary Results <strong>of</strong> Single<br />

Center Prospective Study<br />

HANAN SHAWKY, M.D.; ABDEL HALIM ABO HAMAR, M.D.; SAMAR GALAL, M.D.;<br />

FATMA ZAKARIA, M.D. and DALIA EL-SHORBAGY, M.D.<br />

<strong>The</strong> Department <strong>of</strong> Clinical Oncology, Tanta University Hospital.<br />

ABSTRACT<br />

Purpose: <strong>Temozolomide</strong> (TMZ) is an oral alkylating<br />

agent with demonstrated efficacy as second-line therapy<br />

for patients with recurrent anaplastic astrocytoma and<br />

glioblastoma multiforme (GBM). We reported the<br />

preliminary results <strong>of</strong> the treatment with concomitant<br />

radiation therapy (RT) plus TMZ followed by adjuvant<br />

TMZ therapy in patients with newly diagnosed high grade<br />

gliomas (HGG) to determine the safety, tolerability, and<br />

efficacy.<br />

Patients and Methods: Between January, 2006 and<br />

December, 2007, a total <strong>of</strong> 27 patients over the age <strong>of</strong> 18<br />

years with newly diagnosed, histologically confirmed<br />

HGG were assigned to receive oral TMZ (75 mg/m 2 /d x<br />

7 d/wk for 6 weeks, from the first to the last day <strong>of</strong> RT)<br />

with fractionated RT (60 Gy total dose: 2 Gy x 5 d/wk for<br />

6 weeks) followed by TMZ monotherapy (150 to 200<br />

mg/m 2 /d x 5 days, every 28 days for six cycles) at Clinical<br />

Oncology Department, Faculty <strong>of</strong> Medicine, Tanta<br />

University Hospital. <strong>The</strong> primary end point was overall<br />

survival; secondary end points were progression-free<br />

survival, safety and tolerability.<br />

Results: At a median follow-up period <strong>of</strong> 17 months<br />

(range; 5-30 months), the median progression-free survival<br />

(PFS) for all patients with HGG was 11 months, and the<br />

one-year PFS rate was 43.14%. <strong>The</strong> median overall survival<br />

(OS) was 19 months and the one-year OS rate was 81.2%.<br />

Patients with GBM were analyzed separately from HGG,<br />

and the median overall survival (OS) was 17 months, and<br />

the one-year OS rate was 83.3%. <strong>The</strong> median PFS was 10<br />

months, and the one-year PFS rate was 27.8%. <strong>The</strong> mean<br />

age was 50.2 years (standard deviation ±9.7284), and<br />

44.4% <strong>of</strong> patients had undergone biopsy only. <strong>The</strong>re was<br />

no mortality caused by drug toxicity. Patients younger<br />

than 50 years old and patients who underwent debulking<br />

Correspondence: Dr Hanan Shawky Gamal El-Deen, 37-<br />

Ibn Sinna Street, kafr El Zayat, Gharbya, hannshawky@<br />

yahoo.com<br />

surgery had the best survival outcome.<br />

Conclusion: <strong>The</strong> addition <strong>of</strong> TMZ to RT followed by<br />

adjuvant TMZ was well tolerated, and has shown promising<br />

activity in the treatment <strong>of</strong> newly diagnosed HGG. Further<br />

investigation is warranted.<br />

Key Words: <strong>Temozolomide</strong> – Radiotherapy – High grade<br />

glioma.<br />

INTRODUCTION<br />

Primary brain tumors comprise only<br />

approximately 2% <strong>of</strong> all malignant diseases [1].<br />

However, with an incidence <strong>of</strong> 5 per 100,000<br />

persons, more than 17,000 cases are diagnosed<br />

every year in the United States, with<br />

approximately 13,000 associated deaths [2]. In<br />

adults, the most common histologies are grade<br />

3 anaplastic astrocytoma (AA) and grade 4<br />

glioblastoma multiforme (GBM) [1,3]. <strong>The</strong><br />

standard management <strong>of</strong> malignant gliomas<br />

involves surgical resection to the extent that is<br />

safely feasible [4], followed by radiotherapy<br />

(RT) with or without adjuvant chemotherapy<br />

[5-10]. Despite this multidisciplinary approach,<br />

the prognosis for patients with GBM remains<br />

poor. <strong>The</strong> median survival rates for GBM are<br />

typically in the range <strong>of</strong> 9 to 12 months, with<br />

2-year survival rates in the range <strong>of</strong> only 8%<br />

to 12% [11-15].<br />

In the United States, adjuvant carmustine,<br />

a nitrosourea drug, is commonly prescribed<br />

[16,17]. Nitrosoureas are the main<br />

chemotherapeutic agents used in the treatment<br />

<strong>of</strong> malignant brain tumors; however, they have<br />

107


108<br />

<strong>The</strong> <strong>Outcomes</strong> <strong>of</strong> <strong>Concomitant</strong> <strong>Radiation</strong> <strong>Plus</strong> <strong>Temozolomide</strong><br />

shown only modest antitumor activity [11,13].<br />

Cooperative-group trials have investigated the<br />

addition <strong>of</strong> various chemotherapeutic regimens<br />

to radiotherapy [18-21], but no randomized phase<br />

3 trial <strong>of</strong> nitrosourea-based adjuvant<br />

chemotherapy has demonstrated a significant<br />

survival benefit as compared with radiotherapy<br />

alone, although there were more long-term<br />

survivors in the chemotherapy groups in some<br />

studies [22].<br />

<strong>Temozolomide</strong> (TMZ) is a novel, oral,<br />

second-generation, alkylating agent that has<br />

demonstrated antitumor activity as a single<br />

agent or in combination with other<br />

chemotherapeutic agents in the treatment <strong>of</strong><br />

recurrent and newly diagnosed GBM [23-32].<br />

After oral administration, temozolomide is<br />

rapidly absorbed with almost 100%<br />

bioavailability [33]. It readily crosses the bloodbrain<br />

barrier and achieves effective<br />

concentrations in the CNS with a reported<br />

plasma-CSF ratio <strong>of</strong> approximately 30% to 40%<br />

<strong>of</strong> those observed in the plasma [34,35]. <strong>The</strong><br />

approved conventional schedule is a daily dose<br />

<strong>of</strong> 150 to 200 mg per square meter <strong>of</strong> bodysurface<br />

area for 5 days <strong>of</strong> every 28-day cycle.<br />

Daily therapy at a dose <strong>of</strong> 75 mg per square<br />

meter for up to seven weeks is safe; this level<br />

<strong>of</strong> exposure to temozolomide [36] depletes the<br />

DNA-repair enzyme O6-methylguanine-DNA<br />

methyltransferase (MGMT) [37]. This effect<br />

may be important because low levels <strong>of</strong> MGMT<br />

in tumor tissue are associated with longer<br />

survival among patients with glioblastoma [38,39]<br />

who are receiving nitrosourea-based adjuvant<br />

chemotherapy [40,41].<br />

<strong>The</strong> concurrent administration <strong>of</strong> TMZ and<br />

radiation has been explored in vitro and in vivo<br />

[42]. In vitro studies have shown that, depending<br />

on the cell line, treatment <strong>of</strong> glioma cells with<br />

TMZ and x-rays can have either an additive or<br />

synergistic effect. In a phase II study by Stupp,<br />

et al. [43], demonstrated that concomitant RT<br />

plus continuous daily TMZ followed by adjuvant<br />

TMZ is well tolerated and improves survival<br />

in patients with newly diagnosed GBM. Another<br />

completed phase III studies by the European<br />

Organisation for Research and Treatment <strong>of</strong><br />

Cancer (EORTC) had confirmed this data [23,44].<br />

On the basis <strong>of</strong> this evidence, we initiated<br />

this study to investigate the safety, tolerability,<br />

and survival <strong>of</strong> concomitant RT plus TMZ<br />

therapy followed by adjuvant TMZ therapy in<br />

patients with newly diagnosed HGG.<br />

PATIENTS AND METHODS<br />

Patients:<br />

Between January, 2006 and December, 2007,<br />

32 patients over the age <strong>of</strong> 18 years with newly<br />

diagnosed, histologically confirmed HGG, were<br />

the subjects <strong>of</strong> this study, <strong>of</strong> which 27 were<br />

assessable for response at Clinical Oncology<br />

Department, Faculty <strong>of</strong> Medicine, Tanta<br />

University Hospital. Five patients were<br />

ineligible, not treated, or incorrectly treated:<br />

reasons included treatment refusal (n=2) and<br />

hepatic insufficiency (n=3). Patients were<br />

required to have a Karn<strong>of</strong>sky performance status<br />

(KPS) <strong>of</strong> ≥70 and adequate hematologic, renal,<br />

and hepatic functions, defined as absolute<br />

neutrophil count 1.5x10 9 cells per liter; platelet<br />

count 100x10 9 cells per liter; hemoglobin more<br />

than 90 g/L; serum creatinine and total serum<br />

bilirubin 1.5 times the upper limit <strong>of</strong> normal;<br />

aspartate aminotransferase or alanine<br />

aminotransferase less than 2.5 times the upper<br />

limit <strong>of</strong> normal; and alkaline phosphatase less<br />

than 2.5 times the upper limit <strong>of</strong> normal. Study<br />

enrollment had to be within six weeks from<br />

diagnostic biopsy or resection. Eligible patients<br />

were also required to have no other severe<br />

underlying disease (including chronic hepatitis<br />

B or C infection). Exclusion criteria included<br />

any medical condition that could interfere with<br />

the oral administration <strong>of</strong> temozolomide or any<br />

previous or concurrent malignancies at other<br />

sites.<br />

Study design and treatment:<br />

Within six weeks after the histologic<br />

diagnosis <strong>of</strong> HGG, we assigned eligible patients<br />

to receive temozolomide (marketed as Temodal)<br />

at a dose <strong>of</strong> 75 mg per square meter per day,<br />

given 7 days per week from the first day <strong>of</strong><br />

radiotherapy until the last day <strong>of</strong> radiotherapy,<br />

in a fasting state, 1 hour before RT, and in the<br />

morning on days without RT. <strong>Concomitant</strong> focal<br />

RT was delivered once daily at 2 Gy per fraction,<br />

5 d/wk, for a total <strong>of</strong> 60 Gy. Adequate<br />

immobilization masks were required to ensure<br />

reproducibility. Treatment volumes were<br />

determined on the basis <strong>of</strong> preoperative contrastenhanced<br />

computed tomography (CT) or<br />

gadolinium-enhanced magnetic resonance


Hanan Shawky, et al. 109<br />

imaging (MRI) <strong>of</strong> the brain. Treatment volume<br />

generally included the contrast-enhancing lesion<br />

plus a 2- to 3-cm margin, depending on the<br />

location. <strong>Radiation</strong> therapy was delivered with<br />

60Co photons. After a 4-week break, patients<br />

were then to receive up to six cycles <strong>of</strong> adjuvant<br />

temozolomide according to the standard 5-day<br />

schedule every 28 days. <strong>The</strong> dose was 150 mg<br />

per square meter for the first cycle and was<br />

increased to 200 mg per square meter beginning<br />

with the second cycle, so long as there were no<br />

hematologic toxic effects. Prophylactic<br />

antiemetics were used only as required during<br />

concomitant RT plus temozolomide therapy.<br />

Prophylactic antiemetics, including<br />

metoclopramide or 5-hydroxytryptamine-3<br />

antagonists, were routinely prescribed once a<br />

day before adjuvant temozolomide.<br />

Anticonvulsants and corticosteroids were<br />

administered as needed.<br />

Surveillance and follow-up:<br />

<strong>The</strong> baseline examination included a<br />

complete medical history, physical examination,<br />

determination <strong>of</strong> performance status,<br />

hematology and clinical chemistry assessments,<br />

and gadolinium-enhanced MRI or contrastenhanced<br />

CT <strong>of</strong> the brain. During radiotherapy<br />

with temozolomide, complete blood counts were<br />

checked weekly, and blood chemistry was<br />

checked monthly. During adjuvant<br />

temozolomide therapy, patients underwent a<br />

monthly clinical evaluation and a comprehensive<br />

evaluation at the end <strong>of</strong> cycles 3 and 6. Tumor<br />

progression was defined according to the<br />

modified WHO criteria as an increase in tumor<br />

size by 25 percent, the appearance <strong>of</strong> new<br />

lesions, or an increased need for corticosteroids<br />

[45]. When there was tumor progression, patients<br />

were treated at the investigator's discretion, and<br />

the type <strong>of</strong> second-line therapy was recorded.<br />

Toxic effects were graded according to the<br />

National Cancer Institute Common Toxicity<br />

Criteria, version 2.0, with a score <strong>of</strong> 1 indicating<br />

mild adverse effects, a score <strong>of</strong> 2 moderate<br />

adverse effects, a score <strong>of</strong> 3 severe adverse<br />

effects, and a score <strong>of</strong> 4 life-threatening adverse<br />

effects.<br />

Statistical analysis:<br />

<strong>The</strong> primary end point was overall survival;<br />

secondary end points were progression-free<br />

survival, and safety. Toxicity was graded<br />

according to the common toxicity criteria<br />

(version 2.0). Safety and toxicity are reported<br />

for all treated patients.<br />

Toxic effects are reported separately for the<br />

radiotherapy period, defined as extending from<br />

day 1 <strong>of</strong> radiotherapy until 28 days after the<br />

last day <strong>of</strong> radiotherapy, or until the first day<br />

<strong>of</strong> adjuvant temozolomide therapy. <strong>The</strong><br />

adjuvant-therapy period was defined as<br />

extending from the first day <strong>of</strong> adjuvant<br />

temozolomide therapy until 35 days after day<br />

1 <strong>of</strong> the last cycle <strong>of</strong> temozolomide. Findings<br />

with respect to the quality <strong>of</strong> life are not reported<br />

here.<br />

Overall survival was calculated from the<br />

time <strong>of</strong> study entry until death or last followup<br />

according to the Kaplan-Meier method [46]<br />

with SPSS [Statistical package] (version 9.0).<br />

Mean and standard deviation were estimates <strong>of</strong><br />

quantitative data. Overall survival and<br />

progression-free survival were compared by the<br />

Kaplan-Meier method [46] with statistical<br />

significance assessed by the log-rank test. All<br />

p values were two-tailed; a value <strong>of</strong> ≤0.05 was<br />

considered significant.<br />

RESULTS<br />

Patient characteristics:<br />

Patients ≥18 years <strong>of</strong> age with newly<br />

diagnosed and histologically proven HGG were<br />

eligible for the study. Patient demographics and<br />

baseline disease characteristics for the eligible<br />

27 patients are listed in Table (1). <strong>The</strong> mean<br />

age was 50.2±9.7 years old, (range; 21-71 years<br />

old). <strong>The</strong> majority <strong>of</strong> patients had a Karn<strong>of</strong>sky<br />

performance status (KPS) <strong>of</strong> ≥80. About 44%<br />

<strong>of</strong> the patients underwent biopsy only, 22.2%<br />

underwent gross total resection, and 18.5%<br />

underwent subtotal resection, however,<br />

immediate postoperative imaging was not<br />

performed in all patients. Histopthological slide<br />

revisions were confirmed the diagnosis <strong>of</strong><br />

glioblastoma in 66.7% <strong>of</strong> the reviewed cases;<br />

25.9% had anaplastic astrocytoma (WHO grade<br />

III), and in 7.4% <strong>of</strong> the patients the diagnosis<br />

was oligodendroglioma (WHO grade III). <strong>The</strong><br />

mean time from diagnosis to the start <strong>of</strong> therapy<br />

with RT plus temozolomide was 2.7 weeks,<br />

standard deviation ±0.9842, (range, 1-5 weeks).<br />

Table (2) summarizes the details <strong>of</strong><br />

treatment. Among the 27 patients who were<br />

assigned to receive concomitant radiotherapy


110<br />

<strong>The</strong> <strong>Outcomes</strong> <strong>of</strong> <strong>Concomitant</strong> <strong>Radiation</strong> <strong>Plus</strong> <strong>Temozolomide</strong><br />

plus temozolomide, 17 (63%) completed both<br />

radiotherapy and temozolomide as planned. Ten<br />

patients (37%) prematurely discontinued<br />

adjuvant temozolomide because <strong>of</strong> toxic effects<br />

(in 3 patients), disease progression (in 5), or<br />

other reasons (in 2). <strong>The</strong> majority <strong>of</strong> patients<br />

completed their RT within the prescribed 6<br />

weeks (42±3 days). Unplanned interruptions in<br />

RT were usually brief (median, four days) and<br />

interruptions due to the toxicity <strong>of</strong> therapy<br />

Table (1): Demographic characteristics <strong>of</strong> the 27 patients<br />

with high grade gliomas at baseline.<br />

Characteristic<br />

Age (years):<br />

Mean<br />

Range<br />


Hanan Shawky, et al. 111<br />

was discontinued early because <strong>of</strong> progressive<br />

disease in 5 patients (18.5%)]. Only 5 patients<br />

discontinued adjuvant temozolomide because<br />

<strong>of</strong> toxic effects. Beginning with cycle 2, the<br />

dose <strong>of</strong> temozolomide was increased to 200 mg<br />

per square meter in 70.4% <strong>of</strong> patients.<br />

Seventeen patients (63%) received all<br />

concomitant and adjuvant temozolomide as<br />

planned in the protocol. <strong>The</strong> response rate,<br />

including complete remission and partial<br />

remission, was 55.6%.<br />

Safety and tolerability:<br />

We analyzed adverse events separately<br />

during radiotherapy with concomitant<br />

temozolomide, the adjuvant-therapy period, and<br />

the entire study period (from study entry until<br />

disease progression or last follow-up).<br />

Hematologic toxicity and infection:<br />

<strong>Concomitant</strong> phase <strong>of</strong> treatment. During the<br />

concomitant RT plus temozolomide phase, grade<br />

3 or 4 neutropenia occurred in 2 patients (7.4%)<br />

(Table 3), and grade 3 or 4 thrombocytopenia<br />

occurred in 2 patients (7.4%), with 1 patient<br />

experiencing platelet counts <strong>of</strong> less than 10,000<br />

cells per cubic millimeter. Grade 3 or 4<br />

lymphocytopenia occurred in 3 patients (11.1%).<br />

Two patients had infections that required<br />

hospitalization and treatment interruption.<br />

Analysis indicated that 1 <strong>of</strong> the 2 patients<br />

developed pneumonia. This patient was<br />

receiving corticosteroids and experienced grade<br />

4 neutropenia and lymphocytopenia at the time<br />

<strong>of</strong> infection. One patient required surgical<br />

revision <strong>of</strong> a scar infection and osteomyelitis 3<br />

weeks after start <strong>of</strong> RT. However, this patient's<br />

blood counts were within normal limits during<br />

treatment.<br />

Adjuvant temozolomide: During the adjuvant<br />

temozolomide phase, grade 3 or 4 neutropenia<br />

or thrombocytopenia occurred in 7.4% and<br />

18.5% <strong>of</strong> patients, respectively. Five patients<br />

required a dose reduction or delay because <strong>of</strong><br />

grade 3 or 4 thrombocytopenia.<br />

Non-hematologic toxicities:<br />

Non-hematologic toxicities were mild to<br />

moderate (Table 4). During the CCRT phase,<br />

prophylactic antiemetics were required in 59.3%<br />

<strong>of</strong> patients; however, only 7 patients (25.9%)<br />

received antiemetics for longer than the first<br />

Table (3): Hematologic toxicities and infection in the 27 patients with high grade<br />

gliomas.<br />

RT with concomitant TMZ<br />

Adjuvant TMZ<br />

Adverse Event<br />

Grade 3<br />

Grade 4<br />

Grade 3<br />

Grade 4<br />

No. %<br />

No. %<br />

No.<br />

%<br />

No.<br />

%<br />

Anemia<br />

Neutropenia<br />

Thrombocytopenia<br />

Lymphocytopenia<br />

Infection<br />

1<br />

1<br />

1<br />

2<br />

1<br />

3.7<br />

3.7<br />

3.7<br />

7.4<br />

3.7<br />

0<br />

1<br />

1<br />

2<br />

1<br />

0<br />

3.7<br />

3.7<br />

3.7<br />

3.7<br />

1<br />

1<br />

2<br />

3<br />

0<br />

3.7<br />

3.7<br />

7.4<br />

11.1<br />

0<br />

0<br />

1<br />

3<br />

3<br />

0<br />

0<br />

3.7<br />

11.1<br />

11.1<br />

0<br />

Table (4): Non-hematologic toxicities in the 27 patients with high grade gliomas.<br />

Adverse Event<br />

RT with concomitant TMZ<br />

Grade 2 Grade 3 Grade 4 Grade 2<br />

Adjuvant TMZ<br />

Grade 3 Grade 4<br />

No.<br />

%<br />

No.<br />

%<br />

No. % No. %<br />

No. % No. %<br />

Nausea/vomiting<br />

Rash<br />

Fatigue<br />

6<br />

0<br />

4<br />

22.2<br />

0<br />

14.8<br />

1<br />

1<br />

1<br />

3.7<br />

3.7<br />

3.7<br />

0<br />

0<br />

0<br />

0<br />

0<br />

0<br />

7<br />

0<br />

4<br />

25.9<br />

0<br />

14.8<br />

2<br />

0<br />

1<br />

7.4<br />

0<br />

3.7<br />

0<br />

0<br />

0<br />

0<br />

0<br />

0


112 <strong>The</strong> <strong>Outcomes</strong> <strong>of</strong> <strong>Concomitant</strong> <strong>Radiation</strong> <strong>Plus</strong> <strong>Temozolomide</strong><br />

week <strong>of</strong> the concomitant treatment. During the<br />

adjuvant temozolomide phase, 33.3% <strong>of</strong> patients<br />

required antiemetic therapy. One patient<br />

experienced a treatment-induced rash that<br />

resulted in early discontinuation <strong>of</strong><br />

temozolomide after 7 days <strong>of</strong> CCRT. Moderate<br />

to severe fatigue was reported in 5 patients<br />

during the CCRT phase (grade 3; one patient)<br />

and in 5 patients during the adjuvant<br />

temozolomide phase (grade 3; one patient).<br />

<strong>The</strong> short duration <strong>of</strong> follow-up precludes<br />

definitive assessment <strong>of</strong> late radiation toxicity;<br />

only 9 patients were alive with a follow-up<br />

longer than 24 months. However, signs <strong>of</strong><br />

leukoencephalopathy, without evident clinical<br />

impairment, were apparent on MRI in all <strong>of</strong><br />

these patients. One patient developed<br />

intracranial hypertension, refractory seizures,<br />

and loss <strong>of</strong> vision 25 months after beginning<br />

RT. <strong>The</strong> loss <strong>of</strong> vision may in part be due to<br />

prior RT. Subsequent work-up indicated a spinal<br />

dissemination <strong>of</strong> the disease with positive CSF<br />

cytology and no evidence <strong>of</strong> local recurrence.<br />

A second patient developed neurologic<br />

deterioration with progressive short-term<br />

memory loss and hemiplegia 17 months after<br />

beginning RT. At 26 months, this patient was<br />

still alive without evidence <strong>of</strong> tumor progression.<br />

<strong>The</strong> remaining patients with follow-up longer<br />

than 18 months are doing well without any<br />

clinical signs <strong>of</strong> neurologic impairment.<br />

Thromboembolic events occurred in 3<br />

patients (11.1%). Two patients died <strong>of</strong> cerebral<br />

hemorrhage in the absence <strong>of</strong> a coagulation<br />

disorder or thrombocytopenia.<br />

Survival:<br />

At the time <strong>of</strong> this analysis, 19 patients had<br />

died. <strong>The</strong> median duration <strong>of</strong> follow-up was 17<br />

months, (range, 5-30 months). On the basis <strong>of</strong><br />

Kaplan-Meier estimates, the median overall<br />

survival for the all patients with high grade<br />

gliomas (n=27) was 19 months (95% confidence<br />

interval, 13.58-24.42) (Table 5). About 33.3%<br />

<strong>of</strong> our patients were alive at 2 years, (Table 5).<br />

<strong>The</strong> median progression-free survival was<br />

11 months (95% confidence interval, 8.56-<br />

13.44) (Table 5). <strong>The</strong> two-year progression-free<br />

survival rate was 26.1%.<br />

<strong>The</strong> 18 patients with glioblastoma were<br />

analyzed separately from the other patients with<br />

high grade gliomas, and the median overall<br />

survival was 17 months (95% confidence<br />

interval, 13.9-20.1). <strong>The</strong> one-year and two-year<br />

overall survival rate were 83.3% and 38.1%<br />

respectively (Fig. 1).<br />

<strong>The</strong> median progression-free survival for<br />

the 18 patients with glioblastoma was 10 months<br />

(95% confidence interval, 8.99-11.01) (Fig. 2).<br />

<strong>The</strong> six-month progression-free survival rate<br />

was 72.22%.<br />

Prognostic factors:<br />

We analyzed the median overall survival<br />

and survival rates <strong>of</strong> the eligible patient<br />

populations in relation to prognostic indicators.<br />

In patients younger than 50 years old, the median<br />

survival was not reached at 24 months, with<br />

66.7% <strong>of</strong> these patients still alive at 24 months.<br />

In patients 50 years old, the 24 months overall<br />

survival was only 35.7% months (p=0.005),<br />

(Fig. 3). <strong>The</strong> prognosis by surgical respectability<br />

was also analyzed in these eligible patients.<br />

Patients who underwent gross total resection,<br />

near-total removal and subtotal resection had<br />

24 months overall survival <strong>of</strong> 57%. However,<br />

for patients who underwent partial removal or<br />

biopsy, the 24 months overall survival was<br />

26.8% (p=0.0017), (Fig. 4).<br />

As regard to pathological type, the 24 months<br />

overall survival for patients with glioblastoma<br />

and anaplastic astrocytoma (WHO grade III),<br />

was 38.1% and 77.8% respectively (p=0.0025),<br />

(Fig. 5).<br />

Survival according to other possible<br />

prognostic factors were included, Karn<strong>of</strong>sky<br />

Table (5): Overall Survival and progression-free survival<br />

<strong>of</strong> all patients with high grade gliomas (n=27).<br />

Variable<br />

Overall Survival (months):<br />

Median<br />

95% confidence interval<br />

12-month<br />

24-month<br />

Progression-free survival<br />

(months):<br />

Median<br />

95% confidence interval<br />

12-month<br />

24-month<br />

Survival<br />

19.00 months<br />

13.58-24.42 months<br />

81.16%<br />

33.3%<br />

11.00<br />

8.56-13.44 months<br />

43.14%<br />

26.14%


Hanan Shawky, et al. 113<br />

Oveall Survival<br />

1.0<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

Survival Function<br />

Censored<br />

Progrssion free Survival<br />

1.0<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

Survival Function<br />

Censored<br />

0.0<br />

0 6 12 18 24 30 36<br />

Time (Months)<br />

Fig. (1): Overall survival <strong>of</strong> patients with glioblastoma<br />

(n =18).<br />

1.0<br />

0.8<br />

0.0<br />

0 6 12 18 24 30 36<br />

Time (Months)<br />

Fig. (2): Progression-free survival <strong>of</strong> patients with glioblastoma<br />

(n =18).<br />

1.0<br />

0.8<br />

Oveall Survival<br />

0.6<br />

0.4<br />

0.2<br />

Oveall Survival<br />

0.6<br />

0.4<br />

0.2<br />

0.0<br />

0 6 12 18 24 30 36<br />

Time (Months)<br />

Age<br />


114 <strong>The</strong> <strong>Outcomes</strong> <strong>of</strong> <strong>Concomitant</strong> <strong>Radiation</strong> <strong>Plus</strong> <strong>Temozolomide</strong><br />

Oveall Survival<br />

1.0<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0.0<br />

0 6 12 18 24 30 36<br />

Time (Months)<br />

Sex<br />

Female<br />

Female-censored<br />

Fig. (7): Overall Survival <strong>of</strong> patients with high grade gliomas<br />

according to sex.<br />

performance status (KPS) (p=


Hanan Shawky, et al. 115<br />

In this study, lymphocytopenia is <strong>of</strong>ten<br />

observed with TMZ treatment but may, in part,<br />

be due to the frequent administration <strong>of</strong><br />

corticosteroids as was described in another<br />

phase I trial [36]. Although lymphocytopenia<br />

occurs frequently, it is not typically associated<br />

with clinical sequelae. However, one <strong>of</strong> the 27<br />

patients we treated with concomitant RT plus<br />

TMZ therapy developed pneumonia. <strong>The</strong><br />

frequency <strong>of</strong> opportunistic infections in a similar<br />

patient population treated with RT alone is<br />

unknown.<br />

Nausea and vomiting, the most frequently<br />

reported non-hematologic adverse events, were<br />

also mild to moderate and could be readily<br />

controlled with the administration <strong>of</strong> standard<br />

antiemetics. Non-hematologic toxicity observed<br />

with temozolomide treatment was in agreement<br />

with the report published by Stupp, et al. [43].<br />

Late toxicity resulting from exposure to<br />

alkylating agents or combined modality<br />

treatment remains a concern. <strong>Concomitant</strong> RT<br />

plus TMZ therapy did not increase late toxicities<br />

associated with RT during our follow-up period;<br />

however, follow-up remains too short to make<br />

any conclusions with regard to late toxicities<br />

resulting from treatment with TMZ.<br />

Some reports <strong>of</strong> concomitant RT plus TMZ<br />

followed by adjuvant TMZ therapy for GBM<br />

were published. In 2005, the efficacy <strong>of</strong><br />

postoperative TMZ radiochemotherapy in<br />

malignant glioma was reported in Germany [65].<br />

According to that report, median PFS time was<br />

7.3 months for primary glioblastoma, treated<br />

with concomitant RT plus TMZ. That study also<br />

reported that the median overall survival time<br />

for patients with glioblastoma was 14.6 months.<br />

Another randomized prospective study <strong>of</strong><br />

concomitant RT plus TMZ was reported in 2005<br />

proving that this protocol is more effective than<br />

RT alone in patients with newly diagnosed<br />

glioblastoma [66]. This randomized study<br />

compared concomitant RT plus TMZ with RT<br />

alone in patients with newly diagnosed<br />

glioblastoma. It reported that the median overall<br />

survival time for patients with glioblastoma<br />

was 14.6 months with RT plus TMZ and 12.1<br />

months with RT alone. <strong>The</strong> 2-year overall<br />

survival rate was 26.5% with RT plus TMZ and<br />

10.4% with RT alone. That study confirmed the<br />

effectiveness <strong>of</strong> concomitant RT plus TMZ for<br />

glioblastoma patients.<br />

Survival results in our study are encouraging.<br />

Indeed, the median PFS time <strong>of</strong> 10 months, the<br />

median overall survival <strong>of</strong> 17 months and the<br />

2-year overall survival rate <strong>of</strong> 38.1% for the 18<br />

patients with glioblastoma in our series treated<br />

with concomitant RT plus TMZ followed by<br />

adjuvant TMZ therapy compares favorably with<br />

the other previous reported protocols. At present<br />

concomitant RT plus TMZ followed by adjuvant<br />

TMZ therapy is widely accepted as the current<br />

standard care for patients with glioblastoma<br />

[23,32,42,44,67-72].<br />

Unlike glioblastoma, there is no definite<br />

consensus about the standard regimen for WHO<br />

grade III gliomas, such as anaplastic astrocytoma<br />

and anaplastic oligodendroglioma. For example,<br />

a phase III trial <strong>of</strong> RT plus chemotherapy using<br />

procarbazine, lomustine and vincristine (PCV)<br />

to treat anaplastic oligodendroglioma was<br />

published in 2006 [73]. This study concluded<br />

that PCV plus RT did not prolong the survival<br />

<strong>of</strong> patients with anaplastic oligodendroglioma<br />

and the longer PFS was associated with<br />

significant toxicity. <strong>The</strong>refore, it is essential to<br />

verify the role <strong>of</strong> TMZ in the treatment WHO<br />

grade III glioma. Since the above-mentioned<br />

landmark study <strong>of</strong> Stupp, et al. [66] in 2005,<br />

there have been many phase II and III clinical<br />

studies <strong>of</strong> the treatment <strong>of</strong> HGG with TMZ in<br />

adults [67,68,74-77]. <strong>The</strong>se trials have reported<br />

good outcomes, and our present results for<br />

patients with WHO grade III and IV gliomas<br />

are also favorable, with tolerable toxicity. In<br />

our study the median PFS time was 11 months,<br />

and the median overall survival was 19 months<br />

for the 27 patients with HGG treated with<br />

concomitant RT plus TMZ followed by adjuvant<br />

TMZ therapy. Our results are consistent with<br />

most <strong>of</strong> these reports, confirming that<br />

concomitant RT plus TMZ followed by adjuvant<br />

TMZ therapy <strong>of</strong>fers good clinical outcomes in<br />

the treatment <strong>of</strong> HGG.<br />

Subanalyses performed to determine the<br />

existence <strong>of</strong> prognostic factors in the patient<br />

population under evaluation revealed that<br />

baseline KPS was an important prognostic factor<br />

that correlated meaningfully with median<br />

survival (p=


116<br />

<strong>The</strong> <strong>Outcomes</strong> <strong>of</strong> <strong>Concomitant</strong> <strong>Radiation</strong> <strong>Plus</strong> <strong>Temozolomide</strong><br />

substantially better survival rates than patients<br />

who had a biopsy only. <strong>The</strong>se prognostic factors<br />

observed in our patient population under<br />

evaluation were in agreement with the report<br />

published by Stupp, et al. [43].<br />

In conclusion, this is the first report <strong>of</strong> results<br />

<strong>of</strong> concomitant RT plus TMZ followed by<br />

adjuvant TMZ therapy in the treatment <strong>of</strong> HGG<br />

in Clinical Oncology Department, Tanta<br />

University Hospital, Faculty <strong>of</strong> Medicine, Tanta<br />

University, Egypt. This study demonstrated<br />

concomitant RT plus TMZ followed by adjuvant<br />

TMZ therapy, is a promising regimen for<br />

patients with HGG and we propose concomitant<br />

RT plus TMZ followed by adjuvant TMZ<br />

therapy as an alternative approach with tolerable<br />

toxicities for patients with WHO grade III<br />

gliomas, as well as for those with glioblastoma,<br />

nevertheless, the challenge remains to improve<br />

clinical outcomes further. To confirm this, a<br />

multicenter, meta-analysis and a randomized<br />

trial with a large number <strong>of</strong> patients are required<br />

in the near future. Many questions remain<br />

unanswered regarding the applications <strong>of</strong> this<br />

regimen to lower grade gliomas and the optimal<br />

combination <strong>of</strong> radiotherapy and temozolomide.<br />

REFERENCES<br />

1- Wen PY, Kesari S. Malignant Gliomas in Adults.<br />

NEJM. 2008, 359: 492-507.<br />

2- Ries LAG, Eisner MP, Kosary CL, Hankey BF, Miller<br />

BA, Clegg LX, et al. (eds). SEER Cancer Statistics.<br />

2001, 1973-1998. Bethesda, MD, National Cancer<br />

Institute.<br />

3- Kleihues P, Burger PC, Scheithauer BW. <strong>The</strong> new<br />

WHO classification <strong>of</strong> brain tumours. Brain Pathol.<br />

1993, 3: 255-68.<br />

4- Pichlmeier U, Bink A, Schackert G, Stummer W, the<br />

ALA Glioma Study Group. Resection and survival in<br />

glioblastoma multiforme: An RTOG recursive<br />

partitioning analysis <strong>of</strong> ALA study patients. Neurooncol.<br />

2008, 10(6): 1025-34.<br />

5- Sigmond J, Honeywell RJ, Postma TJ, Dirven CMF,<br />

de Lange SM, van der Born K, et al. Gemcitabine<br />

uptake in glioblastoma multiforme: potential as a<br />

radiosensitizer. Ann Oncol. 2009, 20: 182- 7.<br />

6- Vredenburgh JJ, Desjardins A, Reardon DA, Friedman<br />

HS. Experience with irinotecan for the treatment <strong>of</strong><br />

malignant glioma. Neuro Oncol. 2009, 11: 80-91.<br />

7- Stupp R, Hegi ME, van den Bent MJ, Mason WP,<br />

Weller M, Miriman<strong>of</strong>f RO, et al. Changing paradigmsan<br />

update on the multidisciplinary management <strong>of</strong><br />

malignant glioma. Oncologist. 2006, 11(2): 165-80.<br />

8- Gilbert MR. New treatments for malignant gliomas:<br />

careful evaluation and cautious optimism required.<br />

Ann Intern Med. 2006, 144 (5): 371-3.<br />

9- ReardonDA, Rich JN, Friedman HS, Bigner DD.<br />

Recent Advances in the Treatment <strong>of</strong> Malignant<br />

Astrocytoma. J. Clin. Oncol. 2006, 24 (8): 1253-65.<br />

10- Attenello FJ, Mukherjee D, Datoo G, McGirt MJ,<br />

Bohan E, Weingart JD, et al. Use <strong>of</strong> Gliadel (BCNU)<br />

Wafer in the Surgical Treatment <strong>of</strong> Malignant Glioma:<br />

A 10-Year Institutional Experience. Ann. Surg. Oncol.<br />

2008, 15: 2887-93.<br />

11- Galanis E, Buckner J. Chemotherapy for high-grade<br />

gliomas. Br J Cancer. 2000, 82: 1371-80.<br />

12- Levin V, Leibel S, Gutin P. Neoplasms <strong>of</strong> the central<br />

nervous system, in DeVita V Jr, Hellman S, Rosenberg<br />

S (eds): Cancer: Principles and Practice <strong>of</strong> Oncology.<br />

Philadelphia, PA, Lippincott-Raven. 1997, pp 2022-82.<br />

13- DeAngelis LM. Brain tumors. N Engl J Med. 2001,<br />

344: 114-23.<br />

14- Buckner JC. Factors influencing survival in highgrade<br />

gliomas. Semin Oncol. 2003, 30 (19): 10-14.<br />

15- Curran WJ, Scott CB, Horton J, Nelson JS, Weinstein<br />

AS, Fischbach AJ, et al. Recursive partitioning analysis<br />

<strong>of</strong> prognostic factors in three <strong>Radiation</strong> <strong>The</strong>rapy<br />

Oncology Group malignant glioma trials. J Natl Cancer<br />

Inst. 1993, 85: 704-10.<br />

16- Walker MD, Alexander E Jr, Hunt WE, MacCarty CS,<br />

Mahaley MS Jr, Mealey J Jr, et al. Evaluation <strong>of</strong><br />

BCNU and/or radiotherapy in the treatment <strong>of</strong><br />

anaplastic gliomas: a cooperative clinical trial. J<br />

Neurosurg. 1978, 49: 333-43.<br />

17- Walker MD, Green SB, Byar DP, Alexander E,<br />

Batzdorf U, Brooks WH, et al. Randomized<br />

comparisons <strong>of</strong> radiotherapy and nitrosoureas for the<br />

treatment <strong>of</strong> malignant glioma after surgery. N Engl<br />

J Med. 1980, 303 (23): 1323-29.<br />

18- Medical Research Council Brain Tumor Working<br />

Party. Randomized trial <strong>of</strong> procarbazine, lomustine,<br />

and vincristine in the adjuvant treatment <strong>of</strong> high-grade<br />

astrocytoma: a Medical Research Council trial. J Clin<br />

Oncol. 2001, 19: 509-18.<br />

19- Shapiro WR, Green SB, Burger PC, Mahaley MS Jr,<br />

Selker RG, VanGilder JC, et al. Randomized trial <strong>of</strong><br />

three chemotherapy regimens and two radiotherapy<br />

regimens and two radiotherapy regimens in postoperative<br />

treatment <strong>of</strong> malignant glioma: Brain Tumor Cooperative<br />

Group trial 8001. J Neurosurg. 1989, 71: 1-9.<br />

20- Chang CH, Horton J, Schoenfeld D, Salazer O, Perez-<br />

Tamayo R, Kramer S, et al. Comparison <strong>of</strong><br />

postoperative radiotherapy and combined postoperative<br />

radiotherapy and chemotherapy in the multidisciplinary<br />

management <strong>of</strong> malignant gliomas: a joint <strong>Radiation</strong><br />

<strong>The</strong>rapy Oncology Group and Eastern Cooperative<br />

Oncology Group study. Cancer. 1983, 52 (6): 997-<br />

1007.<br />

21- Green SB, Byar DP, Walker MD, Pistenmaa DA,<br />

Alexander E Jr, Batzdorf U, et al. Comparisons <strong>of</strong>


Hanan Shawky, et al. 117<br />

carmustine, procarbazine, and high-dose<br />

methylprednisolone as additions to surgery and<br />

radiotherapy for the treatment <strong>of</strong> malignant glioma.<br />

Cancer Treat Rep. 1983, 67: 121-32.<br />

22- Stupp R, Hegi ME. Recent developments in the<br />

management <strong>of</strong> malignant glioma. In: Perry MC, ed.<br />

ASCO 2003 educational book. Alexandria, Va.:<br />

American Society <strong>of</strong> Clinical Oncology. 2003, 779-<br />

88.<br />

23- Stupp R, Hegi ME, Mason WP, van den Bent MJ,<br />

Taphoorn MJ, Janzer RC, et al. Effects <strong>of</strong> Radiotherapy<br />

with <strong>Concomitant</strong> and Adjuvant <strong>Temozolomide</strong> versus<br />

Radiotherapy alone on survival in Glioblastoma in<br />

a Randomized Phase III study: 5-year Analysis <strong>of</strong> the<br />

EORTC-<strong>NCI</strong>C trial. <strong>The</strong> Lancet Oncology. 2009, 10<br />

(5): 459-66.<br />

24- Herrlinger U, Rieger J, Koch D, Loeser S, Blaschke<br />

B, Kortmann R-D, et al. Phase II Trial <strong>of</strong> Lomustine<br />

<strong>Plus</strong> <strong>Temozolomide</strong> Chemotherapy in Addition to<br />

Radiotherapy in Newly Diagnosed Glioblastoma:<br />

UKT-03. J. Clin. Oncol. 2006, 24 (27): 4412-7.<br />

25- Wick A, Felsberg J, Steinbach JP, Herrlinger U, Platten<br />

M, Blaschke B, et al. Efficacy and Tolerability <strong>of</strong><br />

<strong>Temozolomide</strong> in an Alternating Weekly Regimen in<br />

Patients With Recurrent Glioma. J. Clin. Oncol. 2007,<br />

25 (22): 3357-61.<br />

26- Kesari S, Schiff D, Henson JW, Muzikansky A, Gigas<br />

DC, Doherty L, et al. Phase II study <strong>of</strong> temozolomide,<br />

thalidomide, and celecoxib for newly diagnosed<br />

glioblastoma in adults. Neuro Oncol. 2008, 10:<br />

300-8.<br />

27- Wakabayashi T, Kayama T, Nishikawa R, Takahashi<br />

H, Yoshimine T, Hashimoto N, et al. A Multicenter<br />

Phase I Trial <strong>of</strong> Interferon-{beta} and <strong>Temozolomide</strong><br />

Combination <strong>The</strong>rapy for High-grade Gliomas<br />

(INTEGRA Study). Jpn. J. Clin. Oncol. 2008, 38:<br />

715-8.<br />

28- Brown PD, Krishnan S, Sarkaria JN, Wu W, Jaeckle<br />

KA, Uhm JH, et al. Phase I/II Trial <strong>of</strong> Erlotinib and<br />

<strong>Temozolomide</strong> With <strong>Radiation</strong> <strong>The</strong>rapy in the<br />

Treatment <strong>of</strong> Newly Diagnosed Glioblastoma<br />

Multiforme: North Central Cancer Treatment Group<br />

Study N0177 J. Clin. Oncol. 2008, 26: 5603-9.<br />

29- Prados MD, Chang SM, Butowski N, DeBoer R,<br />

Parvataneni R, Carliner H, et al. Phase II Study <strong>of</strong><br />

Erlotinib <strong>Plus</strong> <strong>Temozolomide</strong> During and After<br />

<strong>Radiation</strong> <strong>The</strong>rapy in Patients With Newly Diagnosed<br />

Glioblastoma Multiforme or Gliosarcoma. J. Clin.<br />

Oncol. 2009, 27: 579-84.<br />

30- van den Bent MJ, Brandes AA, Rampling R,<br />

Kouwenhoven MCM, Kros JM, Carpentier AF, et al.<br />

Randomized Phase II Trial <strong>of</strong> Erlotinib Versus<br />

<strong>Temozolomide</strong> or Carmustine in Recurrent<br />

Glioblastoma: EORTC Brain Tumor Group Study<br />

26034. J. Clin. Oncol. 2009, 27: 1268-74.<br />

31- Glas M, Happold C, Rieger J, Wiewrodt D, Bahr O,<br />

Steinbach JP, et al. Long-Term Survival <strong>of</strong> Patients<br />

With Glioblastoma Treated With Radiotherapy and<br />

Lomustine <strong>Plus</strong> <strong>Temozolomide</strong>. J. Clin. Oncol. 2009,<br />

27: 1257-61.<br />

32- Beier D, Rohrl S, Pillai DR, Schwarz S, Kunz-<br />

Schughart L A, Leukel P, et al. <strong>Temozolomide</strong><br />

Preferentially Depletes Cancer Stem Cells in<br />

Glioblastoma. Cancer Res. 2008, 68: 5706-15.<br />

33- Newlands ES, Blackledge GR, Slack JA, Rustin GJ,<br />

Smith DB, Stuart NS, et al. Phase I trial <strong>of</strong><br />

temozolomide (CCRG 81045: M&B 39831: NSC<br />

362856). Br J Cancer. 1992, 65: 287-91.<br />

34- Marzolini C, Decosterd LA, Shen F, Gander M,<br />

Leyvraz S, Bauer J, et al. Pharmacokinetics <strong>of</strong><br />

temozolomide in association with fotemustine in<br />

malignant melanoma and malignant glioma patients:<br />

Comparison <strong>of</strong> oral, intravenous, and hepatic intraarterial<br />

administration. Cancer Chemother Pharmacol.<br />

1998, 42: 433-40.<br />

35- Stupp R, Ostermann S, Leyvraz S, Csajka C, Buclin<br />

T. Cerebrospinal fluid levels <strong>of</strong> temozolomide as a<br />

surrogate marker for brain penetration. Proc Am Soc<br />

Clin Oncol. 2001, 20: 59a, (abstr 232).<br />

36- Brock CS, Newlands ES, Wedge SR, Bower M, Evans<br />

H, Colquhoun I, et al. Phase I trial <strong>of</strong> temozolomide<br />

using an extended continuous oral schedule. Cancer<br />

Res. 1998, 58: 4363-7.<br />

37- Tolcher AW, Gerson SL, Denis L, Geyer C, Hammond<br />

LA, Patnaik A, et al. Marked inactivation <strong>of</strong> O6-<br />

alkylguanine-DNA alkyltransferase activity with<br />

protracted temozolomide schedules. Br J Cancer.<br />

2003, 88: 1004-11.<br />

38- Hegi ME, Liu L, Herman JG, Stupp R, Wick W, Weller<br />

M, et al. Correlation <strong>of</strong> O6-Methylguanine<br />

Methyltransferase (MGMT) Promoter Methylation<br />

With Clinical <strong>Outcomes</strong> in Glioblastoma and Clinical<br />

Strategies to Modulate MGMT Activity. J. Clin.<br />

Oncol.2008, 26 (25): 4189-99.<br />

39- Brandes AA, Franceschi E, Tosoni A, Blatt V, Pession<br />

A, Tallini G, et al. MGMT Promoter Methylation<br />

Status Can Predict the Incidence and Outcome <strong>of</strong><br />

Pseudoprogression After <strong>Concomitant</strong><br />

Radiochemotherapy in Newly Diagnosed Glioblastoma<br />

Patients. J. Clin. Oncol. 2008, 26 (13): 2192-7.<br />

40- Esteller M, Garcia-Foncillas J, Andion E, Goodman<br />

SN, Hidalgo OF, Vanaclocha V, et al. Inactivation <strong>of</strong><br />

the DNA-repair gene MGMT and the clinical response<br />

<strong>of</strong> gliomas to alkylating agents. N Engl J Med. 2000,<br />

343: 1350-4. [Erratum, N Engl J Med. 2000, 343:<br />

1740].<br />

41- Hegi ME, Diserens AC, Godard S, Dietrich P-Y, Regli<br />

L, Ostermann S, et al. Clinical trial substantiates the<br />

predictive value <strong>of</strong> O-6-methylguanine-DNA<br />

methyltransferase promoter methylation in<br />

glioblastoma patients treated with temozolomide. Clin<br />

Cancer Res. 2004, 10: 1871-4.<br />

42- Chakravarti A, Erkkinen MG, Nestler U, Stupp R,<br />

Mehta M, Aldape K, et al. <strong>Temozolomide</strong>-Mediated<br />

<strong>Radiation</strong> Enhancement in Glioblastoma: A Report<br />

on Underlying Mechanisms. Clin. Cancer Res. 2006,<br />

12 (15): 4738-46.<br />

43- Stupp R, Dietrich P-Y, Ostermann Kraljevic S, Pica<br />

A, Maillard I, Maeder P, et al. Promising survival for


118<br />

<strong>The</strong> <strong>Outcomes</strong> <strong>of</strong> <strong>Concomitant</strong> <strong>Radiation</strong> <strong>Plus</strong> <strong>Temozolomide</strong><br />

patients with newly diagnosed glioblastoma<br />

multiforme treated with concomitant radiation plus<br />

temozolomide followed by adjuvant temozolomide.<br />

J Clin Oncol. 2002, 20 (5): 1375- 82.<br />

44- Miriman<strong>of</strong>f RO, Gorlia T, Mason W, Van den Bent<br />

MJ, Kortmann RD, Fisher B, et al. Radiotherapy and<br />

<strong>Temozolomide</strong> for Newly Diagnosed Glioblastoma:<br />

Recursive Partitioning Analysis <strong>of</strong> the EORTC<br />

26981/22981-<strong>NCI</strong>C CE3 Phase III Randomized Trial.<br />

J. Clin. Oncol. 2006, 24 (16): 2563-9.<br />

45- Macdonald DR, Cascino TL, Schold SC Jr, Cairncross<br />

JG. Response criteria for phase II studies <strong>of</strong> supratentorial<br />

malignant glioma. J Clin Oncol. 1990, 8: 1277-80.<br />

46- Kaplan EL, Meier P. Nonparametric estimation from<br />

incomplete observations. J Am Stat Assoc. 1958, 53:<br />

457-81.<br />

47- Kristiansen K, Hagen S, Kollevold T, Torvik A, Holme<br />

I, Nesbakken R, et al. Combined modality therapy <strong>of</strong><br />

operated astrocytomas grade III and IV: Confirmation<br />

<strong>of</strong> the value <strong>of</strong> postoperative irradiation and lack <strong>of</strong><br />

potentiation <strong>of</strong> bleomycin on survival time-A<br />

prospective multicenter trial <strong>of</strong> the Scandinavian<br />

Glioblastoma Study Group. Cancer. 1981, 47: 649-<br />

52.<br />

48- Shapiro WR. Chemotherapy <strong>of</strong> malignant gliomas:<br />

studies <strong>of</strong> the BTCG. Rev Neurol. 1992, 148: 428-<br />

34.<br />

49- Grossman SA, O'Neill A, Grunnet M, Mehta M,<br />

Pearlman JL, Wagner H, et al. Phase III study<br />

comparing three cycles <strong>of</strong> infusional carmustine and<br />

cisplatin followed by radiation therapy with radiation<br />

therapy and concurrent carmustine in patients with<br />

newly diagnosed supratentorial glioblastoma<br />

multiforme: Eastern Cooperative Oncology Group<br />

Trial 2394. J Clin Oncol. 2003, 21: 1485-91.<br />

50- Deutsch M, Green SB, Strike TA, Burger PC,<br />

Robertson JT, Selker RG, et al. Results <strong>of</strong> a randomized<br />

trial comparing BCNU plus radiotherapy,<br />

streptozotocin plus radiotherapy, BCNU plus<br />

hyperfractionated radiotherapy, and BCNU following<br />

misonidazole plus radiotherapy in the postoperative<br />

treatment <strong>of</strong> malignant glioma. Int J Radiat Oncol<br />

Biol Phys. 1989, 16: 1389-96.<br />

51- Fisher BJ, Scott C, Macdonald DR, Coughlin C, Curran<br />

WJ. Phase I study <strong>of</strong> topotecan plus cranial radiation<br />

for glioblastoma multiforme: Results <strong>of</strong> <strong>Radiation</strong><br />

<strong>The</strong>rapy Oncology Group Trial 9507. J Clin Oncol.<br />

2001, 19: 1111-7.<br />

52- Del Rowe J, Scott C, Werner-Wasik M, Bahary JP,<br />

Curran WJ, Urtasun RC, et al. Single-arm, open-label<br />

phase II study <strong>of</strong> intravenously administered<br />

tirapazamine and radiation therapy for glioblastoma<br />

multiforme. J Clin Oncol. 2000, 18: 1254-9.<br />

53- Kleinberg L, Grossman SA, Piantadosi S, Zeltzman<br />

M, Wharam M. <strong>The</strong> effects <strong>of</strong> sequential versus<br />

concurrent chemotherapy and radiotherapy on survival<br />

and toxicity in patients with newly diagnosed highgrade<br />

astrocytoma. Int J Radiat Oncol Biol Phys.<br />

1999, 44: 535-43.<br />

54- Brem H, Piantadosi S, Burger PC, Walker M, Selker<br />

R, Vick NA, et al. Placebo-controlled trial <strong>of</strong> safety<br />

and efficacy <strong>of</strong> intraoperative controlled delivery by<br />

biodegradable polymers <strong>of</strong> chemotherapy for recurrent<br />

gliomas: <strong>The</strong> Polymer-Brain Tumor Treatment Group.<br />

Lancet. 1995, 345: 1008-12.<br />

55- Westphal M, Hilt DC, Bortey E, Delavault P, Olivares<br />

R, Warnke PC, et al. A phase 3 trial <strong>of</strong> local<br />

chemotherapy with biodegradable carmustine (BCNU)<br />

wafers (Gliadel wafers) in patients with primary<br />

malignant glioma. Neuro-Oncology. 2003, 5 (2): 79-<br />

88.<br />

56- Friedman HS, Kerby T, Calvert H. <strong>Temozolomide</strong> and<br />

treatment <strong>of</strong> malignant glioma. Clin Cancer Res. 2000,<br />

6: 2585-97.<br />

57- Reid JM, Stevens DC, Rubin J, Ames MM.<br />

Pharmacokinetics <strong>of</strong> 3-methyl-(triazen-1-yl)imidazole-<br />

4-carboximide following administration <strong>of</strong><br />

temozolomide to patients with advanced cancer. Clin<br />

Cancer Res. 1997, 3: 2393-8.<br />

58- van den Bent MJ, Taphoorn MJB, Brandes AA, Menten<br />

J, Stupp R, Frenay M, et al. Phase II Study <strong>of</strong> First-<br />

Line Chemotherapy With <strong>Temozolomide</strong> in Recurrent<br />

Oligodendroglial Tumors: <strong>The</strong> European Organization<br />

for Research and Treatment <strong>of</strong> Cancer Brain Tumor<br />

Group Study 26971. J. Clin. Oncol. 2003, 21 (13):<br />

2525-8.<br />

59- Newlands ES, Stevens MFG, Wedge SR, Wheelhouse<br />

RT, Brock C. <strong>Temozolomide</strong>: a review <strong>of</strong> its discovery,<br />

chemical properties, pre-clinical development and<br />

clinical trials. Cancer Treat Rev. 1997, 23: 35-61.<br />

60- Stupp R, Gander M, Leyvraz S, Newlands E. Current<br />

and future developments in the use <strong>of</strong> temozolomide<br />

for the treatment <strong>of</strong> brain tumours. Lancet Oncol,<br />

2001;2: 552 - 60.<br />

61- Khan RB, Raizer JJ, Malkin MG, Bazylewicz KA,<br />

Abrey LE. A phase II study <strong>of</strong> extended low-dose<br />

temozolomide in recurrent malignant gliomas. Neurooncol.<br />

2002, 4 (1): 39 - 43.<br />

62- Yung WK, Prados MD, Yaya-Tur R, Rosenfeld SS,<br />

Brada M, Friedman HS, et al. Multicenter phase II<br />

trial <strong>of</strong> temozolomide in patients with anaplastic<br />

astrocytoma or anaplastic oligoastrocytoma at first<br />

relapse. J Clin Oncol. 1999, 17 (9): 2762- 71.<br />

63- Yung WK, Albright RE, Olson J, Fredericks R, Fink<br />

K, Prados MD, et al. A phase II study <strong>of</strong> temozolomide<br />

vs. procarbazine in patients with glioblastoma<br />

multiforme at first relapse. Br J Cancer. 2000, 83:<br />

588- 93.<br />

64- Brada M, Hoang-Xuan K, Rampling R, Dietrich P-Y,<br />

Dirix LY, Macdonald D, et al. Multicenter phase II<br />

trial <strong>of</strong> temozolomide in patients with glioblastoma<br />

multiforme at first relapse. Ann Oncol. 2001, 12: 259-<br />

66.<br />

65- Kocher M, Kunze S, Eich HT, Semrau R, Muller RP.<br />

Efficacy and toxicity <strong>of</strong> postoperative temozolomide<br />

radiochemotherapy in malignant glioma. Strahlenther<br />

Onkol. 2005, 181: 157-63.


Hanan Shawky, et al. 119<br />

66- Stupp R, Mason WP, van den Bent MJ, Weller M,<br />

Fisher B, Taphoorn MJ, et al. Radiotherapy plus<br />

<strong>Concomitant</strong> and Adjuvant <strong>Temozolomide</strong> for<br />

Glioblastoma. N Engl J Med. 2005, 352: 987-96.<br />

67- Stupp R, Hegi ME, Gilbert MR, Chakravarti A.<br />

Chemoradiotherapy in Malignant Glioma: Standard<br />

<strong>of</strong> Care and Future Directions. J. Clin. Oncol. 2007,<br />

25 (26): 4127-36.<br />

68- Perry J, Laperriere N, Zuraw L, Chmbers A, Spith<strong>of</strong>f<br />

K, Cairncross JG. Adjuvant chemotherapy for adults<br />

with malignant glioma: a systemic review. Can J<br />

Neurol Sci. 2007, 34: 402-10.<br />

69- Sathornsumetee S, Rich JN and Reardon DA. Diagnosis<br />

and treatment <strong>of</strong> high-grade astrocytoma. Neurol Clin.<br />

2007, 25: 1111-39.<br />

70- Cohen MH, Johnson JR and Pazdur R. Food and Drug<br />

Administration Drug Approval Summary:<br />

<strong>Temozolomide</strong> <strong>Plus</strong> <strong>Radiation</strong> <strong>The</strong>rapy for the<br />

Treatment <strong>of</strong> Newly Diagnosed Glioblastoma<br />

Multiforme. Clin. Cancer Res. 2005, 11 (19): 6767-<br />

71.<br />

71- Athanassiou H, Synodinou M, Maragoudakis E,<br />

Paraskevaidis M, Verigos C, Misailidou D, et al.<br />

Randomized Phase II Study <strong>of</strong> <strong>Temozolomide</strong> and<br />

Radiotherapy Compared With Radiotherapy Alone in<br />

Newly Diagnosed Glioblastoma Multiforme. J. Clin.<br />

Oncol. 2005, 23 (10): 2372-7.<br />

72- Glas M, Happold C, Rieger J, Wiewrodt D, Bahr O,<br />

Steinbach JP, et al. Long-Term Survival <strong>of</strong> Patients<br />

With Glioblastoma Treated With Radiotherapy and<br />

Lomustine <strong>Plus</strong> <strong>Temozolomide</strong>. J. Clin. Oncol. 2009,<br />

27: 1257-61.<br />

73- Cairncross G, Berkey B, Shaw E, Jenkins R,<br />

Scheithauer B, Brachman D, et al. Phase III trial <strong>of</strong><br />

chemotherapy plus radiotherapy compared with<br />

radiotherapy alone for pure and mixed anaplastic<br />

oligodendroglioma: Intergroup <strong>Radiation</strong> <strong>The</strong>rapy<br />

Oncology Group Trial 9402. J Clin Oncol. 2006, 24:<br />

2707-14.<br />

74- Corsa P, Parisi S, Raguso A, Troiano M, Perrone A,<br />

Cossa S, et al.. <strong>Temozolomide</strong> and Radiotherapy as<br />

first-line treatment <strong>of</strong> high-grade gliomas. Tumori.<br />

2006, 92: 299-305.<br />

75- Oshiro OS, Tsugu H, Komatsu F, Ohmura T, Ohta M,<br />

Sakamoto S, et al. Efficacy <strong>of</strong> <strong>Temozolomide</strong><br />

Treatment in Patients with High-grade Glioma.<br />

Anticancer Res. 2009, 29: 911-7.<br />

76- Wick W, Platten M and Weller M. New (alternative)<br />

temozolomide regimens for the treatment <strong>of</strong> glioma.<br />

Neuro-oncol, 2009, 11 (1): 69-79.<br />

77- Vogelbaum MA, Berkey B, Peereboom D, Macdonald<br />

D, Giannini C, Suh JH, et al. Phase II trial <strong>of</strong><br />

preirradiation and concurrent temozolomide in patients<br />

with newly diagnosed anaplastic oligodendrogliomas<br />

and mixed anaplastic oligoastrocytomas: RTOG<br />

BR0131. Neuro Oncol. 2009, 11: 167-75.

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!