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

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

2008 Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Rituximab, methotrexate (MTX), procarbazine, and vincristine (R-MPV)<br />

followed by consolidation high-dose chemotherapy (HDC) and autologous<br />

stem-cell transplant (ASCT) for newly diagnosed primary CNS lymphoma<br />

(PCNSL). Presenting Author: Antonio Marcilio Padula Omuro, Memorial<br />

Sloan-Kettering Cancer Center, New York, NY<br />

Background: In our previous study in newly diagnosed PCNSL, induction<br />

chemotherapy with MTX and cytarabine followed by consolidation HDC<br />

(carmustine, etoposide, cytarabine, melphalan [BEAM]) with ASCT without<br />

radiotherapy resulted in only 50% <strong>of</strong> pts transplanted, reflecting low<br />

efficacy <strong>of</strong> induction chemotherapy, and short intent-to-treat (ITT) median<br />

PFS (�6m). In this phase II trial, we sought to optimize this strategy by<br />

utilizing a more effective induction regimen (R-MPV) and a more aggressive<br />

HDC regimen (Soussain et al). Methods: Pts received 5-7 cycles <strong>of</strong> R-MPV<br />

(MTX: 3.5g/m2) and if a partial or complete response was achieved, HDC<br />

with thiothepa, cyclophosphamide and busulfan was given, followed by<br />

ASCT and no radiotherapy. The primary endpoint was ITT 1 year event-free<br />

survival (promising: 75%, non-promising: 50%; 90% power, significance�0.05).<br />

Follow-up included comprehensive neuropsychological evaluation.<br />

Results: Accrual has been completed (N�32 pts, median age 57<br />

[range 23-67], median KPS�80). Following R-MPV, 17 pts achieved a CR,<br />

13 pts a PR and two pts progressed. A total <strong>of</strong> 25 (78%) pts were<br />

transplanted; the reasons for not receiving transplant were progressive<br />

disease (N�2), poor performance status/ physician’s decision (N� 2),<br />

mobilization failure (N�1) and consent withdrawn (N� 2). One pt who<br />

withdrew consent relapsed and received HDCASCT for salvage. Two (8%)<br />

pts died from early complications <strong>of</strong> ASCT (Stevens-Johnson: one, sepsis:<br />

one) and one pt experienced a fatal late colitis <strong>of</strong> unknown etiology. In the<br />

ITT population, the median EFS and OS have not been reached after a<br />

median follow-up <strong>of</strong> 22 months. The 1 year EFS was 78% (95%CI 58-90)<br />

and the 2y OS was 76% (95% CI 54-89). No pt has developed delayed<br />

neurotoxicity. Conclusions: R-MPV induction regimen resulted in improved<br />

response rates, allowing 78% <strong>of</strong> pts to receive HDC-ASCT. Although more<br />

toxic, this regimen resulted in excellent disease control and survival in the<br />

ITT population, far exceeding the efficacy <strong>of</strong> our previous transplant study.<br />

The primary endpoint was met, warranting further investigation.<br />

2009 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Effects <strong>of</strong> cediranib, a VEGF signaling inhibitor, in combination with<br />

chemoradiation on tumor blood flow and survival in newly diagnosed<br />

glioblastoma. Presenting Author: Elizabeth Robins Gerstner, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: Anti-angiogenic therapy is hypothesized to synergize with<br />

radiation and chemotherapy by improving tumor blood flow. We evaluated<br />

the tolerability, efficacy and potential mechanism <strong>of</strong> action <strong>of</strong> radiation,<br />

temozolomide, and cediranib in newly diagnosed glioblastoma patients.<br />

Methods: Newly diagnosed glioblastoma patients were treated with radiation,<br />

temozolomide, and cediranib followed by monthly temozolomide for 6<br />

cycles and daily cediranib until tumor progression or toxicity as part <strong>of</strong> an<br />

IRB-approved, Phase Ib/II clinical trial. MRI scans including measurement<br />

<strong>of</strong> cerebral blood flow were performed at baseline, weekly during the 6<br />

weeks <strong>of</strong> chemoradiation and then monthly. Radiographic response was<br />

determined by RANO criteria. Results: Six patients were enrolled in the<br />

phase Ib part <strong>of</strong> the study with cediranib 30 mg daily in combination with<br />

temozolomide and radiation. No dose-limiting toxicities were identified.<br />

Forty patients were enrolled in the phase II part <strong>of</strong> the study. Among the<br />

entire cohort <strong>of</strong> 46 patients, median age was 57 (range 35-74), median<br />

KPS was 90% (60-100), 36 patients underwent a subtotal resection and<br />

10 underwent biopsy. 26/30 patients taking corticosteroids were able to<br />

taper corticosteroids during chemoradiation. Off study reasons included<br />

toxicity (14), disease progression (18), and patient preference (2). Five<br />

patients remain on study without disease progression and 20 patients have<br />

died. Median duration on study was 158 days. Median progression free<br />

survival was 288 days (95%CI 240,�) and median overall survival was 786<br />

days (95%CI 411 ,�). Best radiographic response in patients who completed<br />

chemoradiation was CR in 2 patients, PR in 20 patients, and SD in<br />

15 patients. Patients with increased tumor perfusion during chemoradiation<br />

survived nearly 1 year longer (mean OS�611 days) than patients with<br />

decreased perfusion (mean OS�269 days). Conclusions: Cediranib was<br />

well tolerated and led to improved PFS and OS compared to historical<br />

controls, particularly in those with improved perfusion. This combination is<br />

being evaluated in an ongoing randomized trial (RTOG 0837).<br />

Central Nervous System Tumors<br />

117s<br />

2008b Oral Abstract Session, Sun, 8:00 AM-11:00 AM<br />

Chemotherapy plus radiotherapy (CT-RT) versus RT alone for patients with<br />

anaplastic oligodendroglioma: Long-term results <strong>of</strong> the RTOG 9402 phase<br />

III study. Presenting Author: J. Gregory Cairncross, University <strong>of</strong> Calgary,<br />

Calgary, AB, Canada<br />

Background: Anaplastic oligodendrogliomas, pure (AO) and mixed (AOA),<br />

are chemosensitive tumors, especially if co-deleted for chromosomes 1p<br />

and 19q, but whether addition <strong>of</strong> CT to RT prolongs overall survival (OS), is<br />

unknown. Methods: In the RTOG 9402 Phase III trial, patients (pts) with<br />

AO/AOA were randomly assigned to PCV [procarbazine, CCNU (lomustine)<br />

and vincristine] followed by immediate RT vs. immediate RT alone. Early<br />

analysis showed no OS benefit for the PCV�RT group but combined therapy<br />

was associated with a longer progression-free survival (PFS). It also showed<br />

that the finding <strong>of</strong> 1p/19q co-deletion was associated with a longer OS<br />

independent <strong>of</strong> treatment. The current analysis has a median follow up <strong>of</strong><br />

11.3 years (yrs). Results: Two hundred ninety-one patients were randomized,<br />

148 to PCV�RT and 143 to RT. PCV�RT was associated with longer<br />

PFS [2.5 vs. 1.7 yrs, hazard ratio (HR) 0.68, 95% confidence interval (CI)<br />

(0.53, 0.88), P � 0.003] and the 1p/19q co-deletion with a longer Median<br />

Survival Time (MST) [8.7 vs. 2.7 yrs, HR 0.41, 95% CI (0.30, 0.55), P �<br />

0.001]. For the entire cohort, there was no difference in MST by treatment<br />

[4.6 yrs for PCV�RT vs. 4.7 yrs for RT, HR 0.79, 95% CI (0.60, 1.04), P �<br />

0.1]. However, patients with 1p/19q co-deleted tumors lived much longer<br />

after PCV�RT (n � 59) than after RT (n � 67) [14.7 vs. 7.3 yrs, HR 0.59,<br />

95% CI (0.37, 0.95), P � 0.03]. There was no difference in MST by<br />

treatment in pts without the 1p/19q co-deletion [n�137; 2.6 vs. 2.7 yrs,<br />

HR 0.85, 95% CI (0.58, 1.23), P � 0.39]. Re-operation rates upon<br />

progression were similar between treatment arms in co-deleted pts (43%,<br />

PCV�RT vs. 54%, RT) but salvage CT rates were higher in the RT arm [57%<br />

vs. 81% (P � 0.04)]. Conclusions: PCV followed by immediate RT was a<br />

highly effective therapy for patients with 1p/19q co-deleted AO/AOA. In<br />

this setting, 1p/19q co-deletion was both prognostic and predictive, and<br />

the early PFS benefit in co-deleted cases was a harbinger <strong>of</strong> their longer OS.<br />

[This work was supported by RTOG grants U10 CA21661 and U10<br />

CA32115, NCCTG grant U10 CA25224, ECOG grants CA17145 and<br />

CA21115, SWOG grant CA32102, and CCOP grant U10 CA37422 from<br />

the National Cancer Institute (NCI)]<br />

2010 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Effect <strong>of</strong> antiangiogenic therapy on tumor-associated macrophages in<br />

recurrent glioblastoma. Presenting Author: Christine Lu-Emerson, Massachusetts<br />

General Hospital, Boston, MA<br />

Background: Antiangiogenic therapy is associated with increased radiographic<br />

responses in glioblastoma (GBM), but tumors invariably recur.<br />

Tumor associated macrophages (TAMs) have been proposed as a mechanism<br />

<strong>of</strong> resistance to anti-angiogenic therapy in preclinical models. To<br />

examine the role <strong>of</strong> TAMs in recurrent GBM, we analyzed autopsy<br />

specimens from patients with or without history <strong>of</strong> antiangiogenic therapy.<br />

Methods: We compared autopsy brain specimens from 17 recurrent GBM<br />

patients who received anti-angiogenic treatment and chemoradiation<br />

(AAT�) to 7 patients who received chemotherapy and/or radiotherapy<br />

without anti-angiogenic therapy, or no treatment (AAT-). TAMs were<br />

morphologically and phenotypically identified with flow cytometry and<br />

immunohistochemistry (IHC) with CD68, CD11b, CD14, and CD163<br />

markers. All specimens were obtained from the Department <strong>of</strong> Pathology at<br />

Massachusetts General Hospital and clinical information gained through<br />

review <strong>of</strong> the patients’ records. Results: Using flow cytometry, we observed<br />

an increase in CD11b�CD14� cells in the AAT� patients compared to<br />

AAT- patients. Using IHC analysis, we observed a significant increase in<br />

CD68� macrophages in the tumor bulk (p�0.01) and infiltrative areas<br />

(p�0.05) in AAT� versus AAT- patients. We also observed a significant<br />

increase in CD11b� myeloid cells in the tumor bulk (p�0.01) and a<br />

significant increase in CD163� cells in the infiltrative areas (p�0.05) in<br />

the AAT� group. Finally, we noted a trend toward an increase in CD163�<br />

cells in the tumor bulk (p�0.087) in the AAT� versus the AAT- patients.<br />

Conclusions: Patients with recurrent GBM after antiangiogenic therapy<br />

showed a significant increase in CD68� TAMs and in CD11b� cells in the<br />

tumor bulk. Additionally, antiangiogenic treatment induced an increase in<br />

CD68� and CD163� TAMs in the infiltrative region. These data indicate<br />

that TAMs may participate in escape from antiangiogenic therapy and may<br />

represent a future therapeutic target in recurrent GBM.<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!