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.

118s Central Nervous System Tumors<br />

2011 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Modulation <strong>of</strong> antiangiogenic resistance: Targeting the JAK/STAT3 pathway<br />

in glioblastoma. Presenting Author: John Frederick De Groot, University<br />

<strong>of</strong> Texas M. D. Anderson Cancer Center, Houston, TX<br />

Background: Determining the mechanism <strong>of</strong> treatment failure <strong>of</strong> antiangiogenic<br />

therapies would provide insight into either potential combination<br />

approaches or salvage therapies. Methods: Tumor and monocyte expression<br />

<strong>of</strong> phosphorylated-signal transducer and activator <strong>of</strong> transcription 3 (p-<br />

STAT3) was compared between recurrent glioblastoma patients treated<br />

with either conventional chemotherapy or bevacizumab (BEV), and secondarily<br />

validated in murine model systems <strong>of</strong> intracerebral glioma treated with<br />

either BEV or cediranib. The JAK/STAT3 inhibitor AZD1480 alone and in<br />

combination with cediranib was evaluated in immunocompetent mice<br />

bearing intracerebral GL261 xenografts to evaluate therapeutic synergy.<br />

Results: Human glioblastoma patients failing BEV have an increase in<br />

intratumoral p-STAT3- expression with a mean number <strong>of</strong> p-STAT3<br />

expressing cells <strong>of</strong> 36.5 � 8.6% (n�7) compared to a mean <strong>of</strong> 20.2 � 4%<br />

(n�5) in patients never receiving antiangiogenic therapy. In a separate<br />

cohort <strong>of</strong> recurrent glioblastoma patients, the up-regulation <strong>of</strong> p-STAT3<br />

was detected in monocytes within 24 hours after initial BEV administration.<br />

In intracranial xenograft models, both BEV and cediranib extended<br />

median animal survival time, but during treatment failure BEV increased<br />

the mean percentage <strong>of</strong> p-STAT3-expressing cells to 24.6 � 6.4%<br />

(P�0.0011), and cediranib increased this to 16.2 � 1.8% (P�0.0125)<br />

relative to 9.0 � 4.4% in controls. Administration <strong>of</strong> the JAK/STAT3<br />

inhibitor AZD1480 alone and in combination with cediranib reduced tumor<br />

hypoxia and infiltration <strong>of</strong> VEGF inhibitor-induced p-STAT3 macrophages.<br />

The combination reduced tumor volume by 80% compared to untreated<br />

controls, and by 65% compared to cediranib or AZD1480 treatment alone.<br />

Microvascular density and tumor de-differentiation were reduced, indicating<br />

that up-regulation <strong>of</strong> the STAT3 pathway can mediate resistance to<br />

antiangiogenic therapy. Conclusions: Cumulatively, these data suggest that<br />

a prominent mechanism <strong>of</strong> failure <strong>of</strong> anti-VEGF therapy in malignant<br />

glioma patients involves up-regulation <strong>of</strong> the STAT3 pathway, and resistance<br />

to antiangiogenic therapy can be modulated with inhibitors <strong>of</strong> the<br />

JAK/STAT pathway.<br />

2013 Poster Discussion Session (Board #1), Fri, 1:00 PM-5:00 PM and<br />

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

Phase I safety and pharmacokinetic (PK) study <strong>of</strong> veliparib in combination<br />

with whole brain radiation therapy (WBRT) in patients (pts) with brain<br />

metastases. Presenting Author: Minesh P. Mehta, Northwestern University,<br />

Chicago, IL<br />

Background: Veliparib is an oral PARP-1 and -2 inhibitor that enhances the<br />

antitumor activity <strong>of</strong> DNA damaging agents including radiation therapy in<br />

vivo. In pre-clinical models, veliparib crosses the blood-brain barrier. This<br />

ongoing phase I dose-escalation study evaluates the safety, PK, and<br />

provides preliminary antitumor activity <strong>of</strong> veliparib in combination with<br />

WBRT in pts with brain metastases. Methods: Pts with brain metastases<br />

from non-CNS primary solid malignancy, adequate organ function, RPA<br />

Class 2, and KPS �70 were treated with WBRT (37.5 Gy in 15 fractions or<br />

30 Gy in 10 fractions) QD with veliparib BID with every fraction <strong>of</strong> WBRT in<br />

escalating doses <strong>of</strong> 10, 20, 30, 50, 100, 150, and 200 mg; the final WBRT<br />

fraction was followed by 1 extra day <strong>of</strong> veliparib. Safety, PK, and tumor<br />

response by RECIST were assessed. Results: At the time <strong>of</strong> reporting 59 pts<br />

(M/F, 21/38; median age 57 y) had been treated. Baseline KPS was 70,<br />

80, 90, and 100 in 6.8, 32.2, 40.7, and 20.3% pts, respectively; primary<br />

tumor types were breast (n�20), NSCLC (n�20), melanoma (n�9),<br />

colorectal (n�2), and others (n�8); 71.2% pts had multiple lesions; and<br />

18.6% had prior brain SRS. Grade 3/4 treatment-emergent adverse events<br />

(TEAEs; �5%) were fatigue (6.8%), anemia (5.1%), hyponatraemia<br />

(5.1%), and thrombocytopenia (5.1%); other TEAEs (�20%) were fatigue<br />

(57.6%), headache (42.4%), nausea (40.7%), alopecia (28.8%), vomiting<br />

(22%), radiation skin reactions (22%), and decreased appetite (22%). PK<br />

<strong>of</strong> veliparib were approximately dose-proportional, with oral clearance <strong>of</strong><br />

21.6 � 14.2 L/h (mean � SD, n�45), minimal drug accumulation at day<br />

15, and no significant effect <strong>of</strong> food on bioavailability. Tumor response was<br />

evaluable in 48 pts. Best tumor response and median survival were 37.5%<br />

and 10 months (m) for NSCLC, and 52.9% and 12.5 m for breast cancer<br />

(excluding pts with leptomeningeal disease). Conclusions: Addition <strong>of</strong><br />

veliparib up to 200 mg BID was well tolerated with concurrent WBRT and<br />

dose escalation ongoing. The PK <strong>of</strong> veliparib was dose proportional with no<br />

food effect. Preliminary antitumor activity is encouraging and informative<br />

for the design <strong>of</strong> more definitive trials.<br />

2012 <strong>Clinical</strong> Science Symposium, Sat, 1:15 PM-2:45 PM<br />

Effects <strong>of</strong> cediranib (VEGF signaling inhibitor) on edema in newly diagnosed<br />

glioblastoma patients during initial chemoradiation. Presenting<br />

Author: Pavlina Polaskova, Martinos Center for Biomedical Imaging,<br />

Massachusetts General Hospital, Charlestown, MA<br />

Background: A significant benefit <strong>of</strong> antiangiogenic therapy is control <strong>of</strong><br />

brain edema. We evaluated the impact <strong>of</strong> adding cediranib to standard<br />

chemoradiation (CRT) on peritumoral edema in patients with newly<br />

diagnosed glioblastoma(GBM) during the initial 6 weeks <strong>of</strong> CRT. Methods:<br />

Two cohorts <strong>of</strong> patients were enrolled in two clinical trials. The control<br />

group (N�13) received radiation for 6 weeks plus temozolomide. The<br />

cediranib (CED) group received standard CRT plus daily cediranib (N�34).<br />

MRIs were performed at baseline and weekly during CRT. Volumes <strong>of</strong><br />

interest (VOIs) were drawn outlining the enhancing tumor on T1-weighted<br />

post contrast images and the abnormal FLAIR hyperintensity. ADC (apparent<br />

diffusion coefficient) maps were calculated from diffusion-weighted<br />

images and histograms <strong>of</strong> the distribution <strong>of</strong> ADC values created for each<br />

visit using the baseline FLAIR VOI to characterize the peritumoraledema.<br />

Patients were on stable or decreasing doses <strong>of</strong> steroids. Results: In the CED<br />

group, T1 and FLAIR VOI decreased during CRT vs controls where T1 VOI<br />

did not change and FLAIR VOI increased. By the end <strong>of</strong> CRT, the mode <strong>of</strong><br />

the ADC histogram in the CED group shifted to the left while the mode <strong>of</strong><br />

the controls shifted to the right. The skewness, a measure <strong>of</strong> asymmetry <strong>of</strong><br />

the distribution, increased in the CED group and decreased in controls.<br />

Conclusions: In patients with newly diagnosed GBM treated with CRT and<br />

cediranib, tumor volume decreased on T1 and FLAIR images whereas the<br />

FLAIR volume significantly increased in the control group suggesting<br />

increased edema. The shift <strong>of</strong> mode to the right and decreasing skewness in<br />

controls (indicating an increase in the proportion <strong>of</strong> very high ADC values)<br />

suggests that adding cediranibprevented the development <strong>of</strong> edema and<br />

contributed to the resolution <strong>of</strong> existing edema. Preventing the edema by<br />

adding anti-VEGF treatment may improve the tolerability <strong>of</strong> CRT for GBM<br />

patients.<br />

T1 VOI<br />

(ml)<br />

FLAIR VOI<br />

(ml)<br />

ADC histogram mode<br />

(x10 -4 �m 2 /ms)<br />

ADC histogram<br />

skewness<br />

Baseline 6 wks Baseline 6 wks Baseline 6 wks Baseline 6 wks<br />

CED 18.0 11.5 36.0 32.9 10.8 10.1 2.64 2.97<br />

Control 14.8 14.5 25.9 57.1* 9.5 11.8* 2.62 2.42<br />

* Significant difference between CED and controls (p � .05).<br />

2014 Poster Discussion Session (Board #2), Fri, 1:00 PM-5:00 PM and<br />

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

FIP200 and Rb1 expression in CNS metastasis from breast cancer (CNS<br />

met): Potential predictors <strong>of</strong> patient outcome. Presenting Author: Nooshin<br />

Hashemi Sadraei, Cleveland Clinic Foundation, Cleveland, OH<br />

Background: Patients with CNS met have a poor outcome with significant<br />

variation in overall survival. No marker to predict survival exists. Rb1 is a<br />

negative regulator <strong>of</strong> the cell cycle, and FiP200 is an upstream signaling<br />

node that is distributed between the nucleus and cytoplasm. Nuclear<br />

FIP200 inhibits cell proliferation by promoting expression <strong>of</strong> Rb1, whereas<br />

cytoplasmic FIP200 promotes cell survival through autophagy. Previously<br />

FIP200 deletion/mutation has been reported in 20% <strong>of</strong> primary invasive<br />

breast cancer [BR ca] patients. FIP200 cellular localization and genetic<br />

alterations have not been examined in CNS met. Methods: A retrospective<br />

analysis <strong>of</strong> BR ca and CNS met was performed based on tissue availability<br />

from 2 institutions. FIP200 and Rb1 expression and localization was<br />

evaluated by immunohistochemistry. Genetic alterations were evaluated by<br />

DNA array analysis. Results: 80 patients (42 BR ca and 38 CNS met) were<br />

identified. Overall CNS met samples had significantly higher levels <strong>of</strong><br />

cytoplasmic FIP200 as compared to BR ca (52% vs 24% respectively,<br />

p�0.0002) and increased expression <strong>of</strong> FIP200 around areas <strong>of</strong> hypoxia/<br />

necrosis, consistent with increased autophagy. There was also a significant<br />

increase in expression <strong>of</strong> nuclear FIP200 and Rb1 in the CNS met<br />

compared to BR ca (p�0.0007 and p�0.0055 respectively). Median<br />

survival from development <strong>of</strong> CNS met was longer in the group with high<br />

levels <strong>of</strong> nuclear Rb1; 14 (3-27) vs 20 (12-33) months (low expression �<br />

20% vs high � 20%, p�0.1). The same finding was observed with FIP200,<br />

longer survival was observed with high nuclear expression; 16 (7-33) vs 19<br />

(0.4-27) months (low expression �20% vs high � 20%, p�0.2). Our DNA<br />

analysis for copy number variation and LOH in CNS met revealed no<br />

deletion in Rb1 or FIP200. Conclusions: The pattern <strong>of</strong> expression <strong>of</strong> Rb1<br />

and FIP200 in CNS met is different from BR ca. Notably, cytoplasmic<br />

FIP200 which promotes cell survival is highly expressed in CNS mets vs BR<br />

ca. This suggests metastatic breast cancer cells utilize autophagy as a<br />

survival mechanism. In addition, there is a trend to better survival <strong>of</strong> those<br />

patients with CNS met who express high nuclear FIP200 and Rb1, both <strong>of</strong><br />

which have anti-proliferative roles in the nucleus.<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!