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.

10084 General Poster Session (Board #52E), Sun, 8:00 AM-12:00 PM<br />

Activity <strong>of</strong> MK1775, a selective Wee1 inhibitor, alone or in combination<br />

with gemcitabine, in sarcomas. Presenting Author: Jenny Kreahling, H. Lee<br />

M<strong>of</strong>fitt Cancer Center & Research Institute, Tampa, FL<br />

Background: Sarcomas consist <strong>of</strong> more than 50 subtypes <strong>of</strong> mesenchymal<br />

tumors. Doxorubicin alone or in combination has been the primary therapy<br />

for treatment <strong>of</strong> sarcomas; however, the response rates are suboptimal in<br />

many <strong>of</strong> the more common adult subtypes <strong>of</strong> s<strong>of</strong>t tissue sarcoma.<br />

Accordingly, new agents are needed for the treatment <strong>of</strong> this heterogeneous<br />

group <strong>of</strong> diseases. Wee1 is a critical component <strong>of</strong> the G2/M cell cycle<br />

checkpoint control and mediates cell cycle arrest by regulating the<br />

phosphorylation <strong>of</strong> CDC2. Methods: MK1775 treatment <strong>of</strong> multiple sarcoma<br />

preclinical models at clinically relevant concentrations leads to<br />

unscheduled entry into mitosis and initiation <strong>of</strong> apoptotic cell death. In our<br />

current study we have investigated the therapeutic efficacy <strong>of</strong> MK1775 in<br />

sarcoma cell lines, patient-derived tumor explants ex vivo and in vivo in a<br />

xenograft model <strong>of</strong> osteosarcoma both alone and in combination with<br />

gemcitabine. Results: In patient-derived bone and s<strong>of</strong>t tissue sarcoma<br />

samples ex vivo treatments show MK1775 in combination with gemcitabine<br />

causes significant apoptotic cell death suggesting that this treatment<br />

may represent a novel approach in the treatment <strong>of</strong> sarcomas. The cytotoxic<br />

effect <strong>of</strong> Wee1 inhibition on sarcoma cells appears to be independent <strong>of</strong><br />

p53 mutational status. Furthermore, in a patient-derived osteosarcoma<br />

xenograft mouse model we show the therapeutic efficacy <strong>of</strong> MK1775 in vivo<br />

by utilizing magnetic resonance imaging (MRI) and diffusion MRI methods.<br />

Our data shows MK1775 in combination with gemcitabine dramatically<br />

slows tumor growth, increases apoptotic cell death and increases CDC2<br />

activity. Cell viability, a clinically established prognostic indicator <strong>of</strong><br />

survival, was lowest with the combination and very low in animals treated<br />

with MK1775 alone. This was mainly due to increased mineralization <strong>of</strong> the<br />

tumors. Caspase-3 was increased in MK1775 treated animals by immunohistochemistry<br />

as well. Conclusions: These results together with the<br />

promising safety pr<strong>of</strong>ile <strong>of</strong> MK1775 strongly suggest that this drug can be<br />

used as a potential therapeutic agent alone or in combination with<br />

gemcitabine in the treatment <strong>of</strong> both adult as well as pediatric sarcoma<br />

patients.<br />

10086 General Poster Session (Board #52G), Sun, 8:00 AM-12:00 PM<br />

Early assessment <strong>of</strong> MCV to predict clinical outcome in patients with<br />

advanced gastrointestinal stromal tumors (GIST) receiving imatinib. Presenting<br />

Author: Anastasia Constantinidou, The Royal Marsden Hospital NHS<br />

Foundation Trust and the Institute <strong>of</strong> Cancer Research, London, United<br />

Kingdom<br />

Background: In patients with advanced GIST, imatinib trough levels have<br />

been associated with a lower rate <strong>of</strong> clinical benefit. Anecdotal observations<br />

have suggested that commencing treatment with imatinib may be associated<br />

with an increase in red blood cell size as measured by MCV and MCH,<br />

indicating the effect <strong>of</strong> imatinib on KIT signalling in the bone marrow.<br />

Methods: In this retrospective review we examined a possible connection<br />

between changes in MCV and MCH after the first 3 months (mo) <strong>of</strong><br />

treatment with imatinib and progression-free survival (PFS). Data for<br />

patients with inoperable advanced or metastatic GIST treated between<br />

2000 and 2011 were available for analysis. Patients were categorised in<br />

quartiles according to the distribution <strong>of</strong> the percentage <strong>of</strong> change <strong>of</strong> MCV<br />

and MCH between start <strong>of</strong> imatinib (baseline) and 3 (�/- 0.5) mo (ratio<br />

%�MCV change at 3 mo/MCV at baseline). PFS curves were plotted using<br />

the Kaplan-Meier method and compared using the log rank test. Results:<br />

One hundred and thirty patients were eligible for analysis and the<br />

demographics were: male:female 84:46, median age at presentation: 60.5<br />

(23.5-86.5) years, commonest primary tumour sites: stomach (72/130)<br />

and small bowel (44/130), commonest metastatic site: liver (76/130). In<br />

the patients with a 10% or over increase in MCV (p75-p100) at the 3 mo<br />

(�/-0.5) time point a significantly longer PFS was observed compared to<br />

those with a smaller magnitude <strong>of</strong> change (PFS <strong>of</strong> 37.1 mo vs 24.3 mo,<br />

p�0.032). A similar trend although not statistically significant was<br />

observed in patients with MCH <strong>of</strong> 15% or over (p75-p100) with PFS <strong>of</strong><br />

34.0 mo vs 25.8 (p�0.125). The two parameters (MCV and MCH) showed<br />

a high correlation <strong>of</strong> percentage change (r2�0.85). The patients with a<br />

10% MCV increase and /or 15% MCH increase (total�40) showed a<br />

significantly longer PFS (34.0 mo) compared to those with lower percentage<br />

change (PFS� 24.3) p�0.035. Conclusions: Simple laboratory parameters<br />

may be indicators <strong>of</strong> imatinib pharmacokinetics and may therefore be<br />

used as surrogate markers <strong>of</strong> clinical outcome. Larger prospective studies<br />

are required to confirm the results <strong>of</strong> this study.<br />

Sarcoma<br />

651s<br />

10085 General Poster Session (Board #52F), Sun, 8:00 AM-12:00 PM<br />

Pharmacokinetics <strong>of</strong> escalated dose <strong>of</strong> imatinib in patients with advanced<br />

gastrointestinal stromal tumors. Presenting Author: Changhoon Yoo, Department<br />

<strong>of</strong> Oncology, Asan Medical Center, University <strong>of</strong> Ulsan College <strong>of</strong><br />

Medicine, Seoul, South Korea<br />

Background: Although escalated dose (ED, 800 mg daily) <strong>of</strong> imatinib (IM) is<br />

one <strong>of</strong> important options in resistant gastrointestinal stromal tumors (GIST)<br />

on standard dose (SD, 400mg daily), there has been lack <strong>of</strong> pharmacokinetic<br />

data at ED. We therefore explore the potential relationship between<br />

IM plasma trough level (Cmin), and clinical outcomes or toxicities in<br />

patients treated with ED. Methods: Between 2008 and 2011, steady-state<br />

IM Cmin was measured in 66 patients with GIST who received ED <strong>of</strong> IM<br />

using liquid chromatography-tandem mass spectrometry. Percent change<br />

from IM Cmin at SD was calculated in 43 patients that both SD and ED IM<br />

Cmin were measured. The association with efficacy and toxicity was<br />

analyzed with grouping patients into quartiles according to IM Cmin at ED<br />

and percent change. Results: Median age was 59 years (range, 36–77) and<br />

40 patients were male. KIT exon 11 and 9 mutation was detected in 32 and<br />

18 patients, respectively. At ED, partial response was achieved in 4<br />

patients (6%) and stable disease was in 32 patients (48%). The median<br />

progression-free survival was 4.2 months (95% CI, 1.8-6.6) and overall<br />

survival was 38.5 months (95% CI, 7.6-69.3). The mean (� standard<br />

deviation) IM Cmin at ED was 3552 (� 1540) ng/mL. IM Cmin was<br />

significantly increased after dose escalation (p �0.001), and mean percent<br />

change from IM Cmin at SD was 162% (� 100). Body surface area<br />

(p�0.01), hemoglobin (p�0.001), and neutrophil count (p�0.006) were<br />

significant covariates for IM Cmin at ED in multivariate analysis. The group<br />

<strong>of</strong> quartiles <strong>of</strong> IM Cmin at ED and percent change was not associated with<br />

response and survival outcomes. However, grade 3-4 hematologic or grade<br />

2-4 non-hematologic toxicity was observed in 9% (1/11) <strong>of</strong> patients in the<br />

lowest percent change quartile (Q1, �90%) compared with 56% (18/32)<br />

in Q2 to Q4 (p�0.012), although absolute values were not associated with<br />

toxicity. Conclusions: In GIST patients treated with ED <strong>of</strong> IM, IM Cmin was<br />

not associated with response and survival. <strong>Clinical</strong>ly significant toxicity<br />

following dose escalation was correlated with percent change <strong>of</strong> IM Cmin, rather than absolute values. Monitoring <strong>of</strong> IM Cmin might help to predict or<br />

manage ED <strong>of</strong> IM induced toxicity.<br />

10087 General Poster Session (Board #52H), Sun, 8:00 AM-12:00 PM<br />

SDHA and SDHB mutations in KIT/PDGFRA WT gastrointestinal stromal<br />

tumors. Presenting Author: Margherita Nannini, Seragnoli Department and<br />

GIST Study Group, University <strong>of</strong> Bologna, Bologna, Italy<br />

Background: KIT/PDGFRA wild-type (WT) GISTs harbour mutations on<br />

SDHB and SDHC and, more recently, we described mutations on SDHA<br />

using massively parallel sequencing approach. We sequenced SDHA and<br />

SDHB genes in a larger series in order to validate the data. Methods: SDHA<br />

gene (1-15 exons) and SDHB gene (1-8 exons) (even not all exons in all<br />

samples) were sequenced on tumor (T) and/or peripheral blood (PB) <strong>of</strong> WT<br />

GIST patients by Sanger Sequencing method. DNA was extracted from<br />

tumor specimens by the QIAmp DNA Mini kit (Qiagen, Milan, Italy) and<br />

amplified with specific primer pairs designed to amplify exons but not<br />

SDHA pseudo-genes located on chromosomes 3 and 5. Then, PCR products<br />

were purified with the Qiaquick PCR purification kit (Qiagen, Milan, Italy)<br />

and sequenced on both strands using the Big Dye Terminator v1.1 Cycle<br />

Sequencing kit (Applied Biosystems). Sanger sequencing was performed<br />

on ABI 3730 Genetic Analyzer (Applied Biosystems). Results: SDHA gene<br />

exons were sequenced on a total <strong>of</strong> 27 WT GIST patients, in particular on T,<br />

PB and both from 12, 6 and 9 patients respectively. SDHB gene exons were<br />

sequenced on a total <strong>of</strong> 18 out <strong>of</strong> 27 patients, in particular on T, PB and<br />

both from 7, 8 and 3 patients respectively. 8 SDHA mutations were found<br />

in 5 samples (18.5%). Besides those previously identified, 5 new SDHA<br />

mutations were found in other 3 samples: one sample harboured R171C<br />

and R589Q heterozygous missense mutation in exons 5 and 13 respectively.<br />

The other one harboured G419R and E564K heterozygous missense<br />

mutations in exons 9 and 13 respectively. The third sample harboured a<br />

delCAG immediately upstream <strong>of</strong> exon 5, in heterozygosis on PB and in<br />

homozygosis on T. A SDHB heterozygous mutation (301delT) in exon 4 was<br />

found on 1 PB sample. Conclusions: the presence <strong>of</strong> SDHA mutations has<br />

been confirmed in a subgroup <strong>of</strong> WT GIST patients. All subunits <strong>of</strong> SDH<br />

complex should be sequenced on WT GIST patients in order to explore the<br />

frequency and any linkage between each other and the pathogenetic and<br />

clinical significance.<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!