24.12.2012 Views

Annual Meeting Proceedings Part 1 - American Society of Clinical ...

Annual Meeting Proceedings Part 1 - American Society of Clinical ...

Annual Meeting Proceedings Part 1 - American Society of Clinical ...

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.

2031 Poster Discussion Session (Board #19), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

NCCTG N057K phase II trial <strong>of</strong> everolimus, temozolomide, and radiotherapy<br />

in patients with newly diagnosed glioblastoma: A North Central<br />

Cancer Treatment Group trial. Presenting Author: Daniel Ma, Mayo Clinic,<br />

Rochester, MN<br />

Background: The mammalian target <strong>of</strong> rapamycin (mTOR) functions within<br />

the PI3K/Akt pathway as a critical modulator <strong>of</strong> cell survival. Preclinical<br />

studies in GBM indicate that the combination <strong>of</strong> mTOR inhibitors, such as<br />

everolimus (RAD001), with either radiation therapy (RT) or temozolomide<br />

(TMZ) provide increased tumor cell killing. Methods: Newly diagnosed GBM<br />

pts were eligible for the study. RAD001 was dosed orally at 70 mg/week<br />

weekly, starting 1 week prior to RT/TMZ, and continued throughout<br />

RT/TMZ, adjuvant TMZ and then until progression. This was a single arm<br />

phase II design powered to detect a true overall survival at 12 months<br />

(OS12) <strong>of</strong> 73% (vs 58% in historical controls). Secondary endpoints were<br />

toxicity, response rate, and time to progression (TTP). A subgroup <strong>of</strong><br />

patients with measurable residual disease were eligible for the PET imaging<br />

component <strong>of</strong> the study, consisting <strong>of</strong> an 18FLT-PET/CT scan performed<br />

before and after the initial two doses <strong>of</strong> RAD001 but before the first dose <strong>of</strong><br />

RT or TMZ. Results: 103 patients were accrued to phase II <strong>of</strong> which 100<br />

were evaluable. The median age was 60.5 years (23-81), median ECOG PS<br />

was 1, 46 patients had GTR, 33 STR, and the remainder had biopsy at<br />

diagnosis. Treatment tolerance was acceptable: 17% patient had at least<br />

one grade 3 hematologic toxicity; 14% had at least one grade 4 hematologic<br />

toxicity, 42% had at least one grade 3 non-heme toxicity, while 12%<br />

had at least one grade 4 non-heme toxicity. No increased incidence <strong>of</strong><br />

infectious complications was observed and there were no treatment related<br />

deaths. Median PFS was 5.3 months (1.3-21.4), with 22 patients<br />

progression free. Mature OS data will be available at the meeting. MGMT<br />

methylation status analysis is ongoing. Of the pts who had evaluable<br />

FLT-PET data, three <strong>of</strong> eight (37.5%) had a drop in SUVmax �25% after<br />

two treatments <strong>of</strong> RAD001. Conclusions: RAD001 with standard <strong>of</strong> care<br />

chemoradiation had moderate toxicity. Serial 18FLT-PET was feasible for<br />

evaluating drug-induced changes in tumor proliferation following RAD001.<br />

Final outcome data and association <strong>of</strong> MGMT status with outcome will be<br />

reported at the meeting.<br />

2033 Poster Discussion Session (Board #21), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Final results <strong>of</strong> a randomized phase III trial <strong>of</strong> nimotuzumab for the<br />

treatment <strong>of</strong> newly diagnosed glioblastoma in addition to standard radiation<br />

and chemotherapy with temozolomide versus standard radiation and<br />

temoziolamide. Presenting Author: Manfred Westphal, Department <strong>of</strong><br />

Neurosurgery, UKE Hamburg, Hamburg, Germany<br />

Background: The receptor for epidermal growth factor (EGF-R) has consistently<br />

been found expressed or overexpressed in human malignant glioma<br />

disease. Therapeutic targeting <strong>of</strong> the EGF-R has shown mixed responses<br />

which appear to be restricted by specific features <strong>of</strong> the tumor cells.Nimotuzumab<br />

binds preferentially to highly over expressing cells, a phase III trial<br />

was initiated to test efficacy in glioma. Methods: Nimotuzumab was tested<br />

in an open label, randomized, multicenter Phase III trial in patients with<br />

histologically confirmed, newly diagnosed glioblastoma. 12 consecutive<br />

weekly infusions <strong>of</strong> 400mg during standard radiotherapy with Temozolomide<br />

followed by biweekly infusions <strong>of</strong> 400mg arm A was randomized<br />

against standard radio chemotherapy alone arm B. Primary endpoint was<br />

PFS as determined by centralized neuro-imaging review. OS as secondary<br />

endpoint with quality <strong>of</strong> life, safety as additional parameters.MGMT and<br />

EGF-R were assessed where sufficient materials could be retrieved as well<br />

as tumor hypoxia. Results: Between 2007 and 2010, 149 patients were<br />

randomized and 142 were available for analysis. Stratification according to<br />

resection status resulted in 40 pts in the treatment arm and 41 in the<br />

control arm with residual tumor and 31 vs 30 pts without residual tumor.<br />

PFS: 12 month was 25.5% arm A vs 20.3% in arm B (p � 0.78) and OS:<br />

679 days vs 596 days. For non - methylated MGMT the difference was most<br />

notable: OS arm A 28 pats: 19.6 months vs arm B 28 pats 15.0 months<br />

EGF-R amplification, gave no clear signal, neither was there a statistically<br />

significant treatment effect between with or without residual tumor. Tumor<br />

hypoxia does not appear to have predictive value in the overall group but<br />

possibly in subgroups.AEs and SAEs did not reveal a new specific toxicity<br />

pr<strong>of</strong>ile beyond the known side effects from glioma treatment with current<br />

standard. Conclusions: Nimotuzumab shows a clear trend towards efficacy<br />

in MGMT non-methylated glioblastoma patients. Safety pr<strong>of</strong>ile and indications<br />

for subgroup efficacy potentially justify focused evaluation <strong>of</strong> antibody<br />

therapy against glioblastoma.<br />

Central Nervous System Tumors<br />

123s<br />

2032 Poster Discussion Session (Board #20), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

Phase I/randomized phase II trial <strong>of</strong> either dasatinib or placebo combined<br />

with standard chemoradiotherapy for newly diagnosed glioblastoma multiforme<br />

(GBM): Final results <strong>of</strong> phase I study. Presenting Author: Nadia N.<br />

Laack, Mayo Clinic, Rochester, MN<br />

Background: Dasatinib is a potent oral ATP competitive multi-targeted<br />

kinase inhibitor <strong>of</strong> multiple members <strong>of</strong> the Src kinase family, known to be<br />

involved in gliomagenesis and invasion. N0877 is a phase I/randomized<br />

phase II trial evaluating the combination <strong>of</strong> dasatinib, radiation (RT) and<br />

temozolomide (TMZ) in newly diagnosed GBM. The phase I portion has<br />

been completed and is the focus <strong>of</strong> this report. Methods: A cohorts-<strong>of</strong>-3<br />

design was used to assess the safety <strong>of</strong> dasatinib in combination with RT<br />

and concomitant TMZ, and establish the phase II dose <strong>of</strong> the combination.<br />

Dasatinib was given orally for 42 days, beginning with the first day <strong>of</strong> RT<br />

(total dose 60 Gy) and first dose <strong>of</strong> TMZ (75 mg/m2/d). A 24 - 42 day rest<br />

(cycle 2) followed the RT/TMZ/dasatinib. Patients (pts) were observed for<br />

DLT to the end <strong>of</strong> cycle 2. Patients then received 6 cycles (28 day cycles) <strong>of</strong><br />

dasatinib (once daily) and TMZ (days 1-5). At the completion <strong>of</strong> 6 cycles <strong>of</strong><br />

TMZ � dasatinib, pts stay on dasatinib only (28 day cycles) until<br />

progressive disease. Results: Phase I component is complete with 13<br />

patients (3 at dose level 0, 3 at dose level 0-A, 7 at dose level 1). One<br />

patient in dose level 1 had to be replaced due to the development <strong>of</strong><br />

unrelated medical issues. One DLT (grade 4 pancytopenia) was observed in<br />

1 patient at dose level 0 (50mg bid) and one DLT (grade 3 rash) was<br />

observed in 1 patient at dose level 1 (150mg qd). MTD <strong>of</strong> dasatinib was<br />

determined to be 150mg daily. Most common adverse events throughout<br />

the entire study period were hematologic with the most common toxicity<br />

being lymphopenia (grade 3 in 69% <strong>of</strong> patients, grade 4 in 8%). Other<br />

toxicities attributed to treatment and occurring in � 10% <strong>of</strong> patients<br />

included anemia (31%), neutropenia (15%), and fatigue(15%). Best<br />

response was stable disease in 10 patients, progressive disease in 1<br />

patient, and not evaluable in 2. Conclusions: MTD for dasatinib in<br />

combination with TMZ and RT in newly diagnosed GBM patients is 150mg<br />

daily. Toxicity was primarily hematologic with minimal non-hematologic<br />

events. This dose is currently being used in the ongoing placebo-controlled,<br />

randomized phase II trial.<br />

2034 Poster Discussion Session (Board #22), Fri, 1:00 PM-5:00 PM and<br />

4:30 PM-5:30 PM<br />

APG101_CD_002: A phase II, randomized, open-label, multicenter study<br />

<strong>of</strong> weekly APG101 plus reirradiation versus reirradiation in the treatment <strong>of</strong><br />

patients with recurrent glioblastoma. Presenting Author: Martin Bendszus,<br />

Department <strong>of</strong> Neuroradiology, University Hospital Heidelberg, Heidelberg,<br />

Germany<br />

Background: Preclinical data indicate antiinvasive activity <strong>of</strong> APG101, an<br />

intravenous CD95 ligand-binding fusion protein, as well as synergistic<br />

activity together with radiotherapy in glioblastoma. Methods: Patients with<br />

recurrent glioblastoma after prior standard radiochemotherapy with temozolomide<br />

(� 1 second-line chemotherapy) were considered for re-irradiation<br />

provided a tumor diameter 1-4 cm and time since the end radiotherapy � 8<br />

months. Patients were randomized 1:2 between radiotherapy (36 Gy; 5<br />

times 2 Gy per week) or radiotherapy plus APG101 at 400 mg weekly flat<br />

dose to be continued until progression. Radiotherapy plans were centrally<br />

evaluated. Primary endpoint was 6-months progression free survival (PFS-<br />

6). MRIs were performed every 6-weeks and centrally read. Sample size <strong>of</strong><br />

the investigational treatment arm according to a two-stage design <strong>of</strong> Simon<br />

required 55 patients. A control arm <strong>of</strong> 28 patients was implemented to<br />

validate the assumptions on PFS-6 in a cohort <strong>of</strong> patients treated with<br />

reirradiation alone. Results: Between 12/09 and 09/11, 84 pts in 25<br />

centers were randomized and treated, preliminary data <strong>of</strong> the current report<br />

are available on 71 patients (49 APG01 � irradiation, 22 irradiation<br />

alone). Median age was 57 years, median KPS 90%. The maximal tumor<br />

diameter was � 2-5 cm in 34 patients and � 2.5 cm in 37 patients. No<br />

SUSARs have to be reported. Nine patients achieved PFS-6 in the APG101<br />

arm and none in the radiotherapy arm. Conclusions: APG101_CD_002 is<br />

the first trial evaluating CD95-mediated pathway inhibition as a therapeutic<br />

strategy. This trial is also the prospective trial on reirradiation in<br />

glioblastoma. Side effects <strong>of</strong> the combination were minimal, and treatment<br />

could be delivered as planned. The experimental arm met the primary<br />

endpoint. The approach to block rather than stimulate the CD95 system is<br />

breaking a paradigm. Our data suggest that CD95 inhibition by APG101<br />

should be evaluated in the management <strong>of</strong> newly diagnosed glioblastoma in<br />

combination with standard radiochemotherapy. Further molecular data and<br />

updated results will be presented.<br />

Visit abstract.asco.org and search by abstract for the full list <strong>of</strong> abstract authors and their disclosure information.

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

Saved successfully!

Ooh no, something went wrong!