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.

188s Developmental Therapeutics—Experimental Therapeutics<br />

3060 General Poster Session (Board #15B), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> EP-100, a luteinizing hormone releasing hormone<br />

(LHRH) ligand conjugated to a synthetic cytolytic peptide in patients with<br />

advanced refractory LHRH- receptor (R)-expressing tumors. Presenting<br />

Author: Ramesh K. Ramanathan, Virginia G. Piper Cancer Center at<br />

Scottsdale Healthcare/TGen, Scottsdale, AZ<br />

Background: EP-100 is a synthetic 28 amino acid LHRH peptide conjugated<br />

to an 18 aa cytolytic peptide. Selective targeting in LHRH-R<br />

expressing cells occurs via direct binding to the extracellular membrane<br />

receptor followed by disruption <strong>of</strong> the membrane by the cytolytic peptide<br />

portion. A first in human, phase I study was performed. Methods: Eligible<br />

pts had adequate organ function and LHRH-R tumor expression, as<br />

determined by IHC. EP-100 was given IV (30-60 min) weekly x3for<br />

cohorts 1- 6 (n�20 at 0.6 - 5.2 mg/m2 ) and then twice weekly x3for<br />

cohorts 7- 11 (n�18 at 7.8 - 40 mg/m2 ) with a week break. The<br />

pharmacokinetic pr<strong>of</strong>ile <strong>of</strong> EP-100 and antibody production against EP-<br />

100 was determined by a validated HPLC/MS and ELISA respectively.<br />

Potential activity <strong>of</strong> EP-100 on the pituitary gonadotropes was determined<br />

by plasma LH and FSH. Results: We screened 97 pts, archival tumor tissue<br />

obtained from 81 pts; 53 (65%) were positive for LHRH-R, <strong>of</strong> which 37<br />

were enrolled (female 76%) and treated. The LHRH-R expression rate in<br />

breast was 81% (N�21) and in ovarian cancer 56% (n�18). Most pts had<br />

breast (n� 16, 42%) ovary (n�7,18%), colon (n�4, 11%) or uterine<br />

(n�3, 8%) cancer. Pts were treated in a 3 � 3 standard dose escalation<br />

scheme. Doses were escalated 100% in the absence <strong>of</strong> a grade 2 drug<br />

related toxicity. MTD was not reached at the highest dose level <strong>of</strong> 40<br />

mg/m2 . Only one DLT occurred, a grade 2 increase in ALT/AST. The only<br />

other drug-related toxicity was a self limited infusion-related skin reaction<br />

(grade 1-2) in 10 pts. EP-100 was rapidly cleared from circulation, mean<br />

half life ranged from 7.1 � 3.8 min to 15.9 � 3.6 min. Best response was<br />

stable disease for �12 weeks in 5 pts. In 3 pts rapid, sustained decreases<br />

in LH/FSH levels were noted. Antibody production against EP-100 was<br />

absent in all pts. Conclusions: The study has completed accrual, the phase<br />

II dose is 30-40 mg/m2 twice a week x 3 weeks.EP-100is safe, well<br />

tolerated and not antigenic/immunogenic. Preclinical studies indicated<br />

synergy with paclitaxel and doxorubicin. A phase II study <strong>of</strong> EP-100 plus<br />

paclitaxel in ovarian cancer is planned.<br />

3062 General Poster Session (Board #15D), Mon, 8:00 AM-12:00 PM<br />

Open-label extension study <strong>of</strong> the RNAi therapeutic ALN-VSP02 in cancer<br />

patients responding to therapy. Presenting Author: Maria Alsina, Molecular<br />

Therapeutics Research Unit, Vall d’Hebron University Hospital, Barcelona,<br />

Spain<br />

Background: ALN-VSP02 is an RNA interference (RNAi) therapeutic comprised<br />

<strong>of</strong> lipid nanoparticle-formulated small interfering RNAs targeting<br />

vascular endothelial growth factor (VEGF)-A and kinesin spindle protein<br />

(KSP). In a phase 1 trial, ALN-VSP02 administered as an iv infusion q2 wks<br />

was well-tolerated and showed evidence <strong>of</strong> anti-VEGF pharmacology and<br />

antitumor activity. Methods: Patients treated on the phase I trial with stable<br />

disease (SD) or better after 4 months (8 doses) were eligible to continue on<br />

an extension study until disease progression. Main objectives included<br />

continued evaluation <strong>of</strong> safety/tolerability and assessment <strong>of</strong> disease<br />

response. Results: Seven <strong>of</strong> 37 patients (18.9%) evaluable for response<br />

went onto the extension study, including 1 <strong>of</strong> 7 (14.2%) at 0.4 mg/kg, 2 <strong>of</strong><br />

5 (40%) at 0.7 mg/kg, and 4 <strong>of</strong> 11 (36.3%) at 1.0 mg/kg. All had<br />

progressed after one or more prior therapies. Tumor types included head<br />

and neck squamous cell carcinoma, angiosarcoma, endometrial cancer,<br />

renal cell carcinoma (RCC, N�2), and pancreatic neuroendocrine tumor<br />

(PNET, N�2). At the time <strong>of</strong> enrollment, 6 had SD and one (endometrial<br />

cancer with multiple liver metastases) had an unconfirmed partial response<br />

(PR). The average length <strong>of</strong> time on treatment (including phase I and<br />

extension studies) was 9.5 months (range 5-19). As <strong>of</strong> January 2012, 3<br />

patients remain on study, including the endometrial cancer patient with an<br />

ongoing PR who has had �80% tumor regression after 19 months <strong>of</strong><br />

treatment at 0.7 mg/kg and two patients with RCC and PNET with<br />

continued SD after nearly 1 year <strong>of</strong> treatment at 1.0 mg/kg. The other<br />

patients with RCC and PNET at 1.0 mg/kg with SD came <strong>of</strong>f after 8.5 and<br />

5.5 months, respectively, for adverse events that included fatigue or<br />

elevated alkaline phosphatase. A decrease in spleen volume, likely an<br />

on-target effect and not associated with any adverse events, occurred to a<br />

greater degree on the extension study than on the phase I trial and was most<br />

pronounced in patients receiving � 12 doses. Conclusions: ALN-VSP02 has<br />

preliminary activity against endometrial cancer, RCC and PNET and a<br />

favorable safety pr<strong>of</strong>ile that permits chronic dosing. Phase II trials are<br />

warranted in these and other VEGF-overexpressing tumors.<br />

3061 General Poster Session (Board #15C), Mon, 8:00 AM-12:00 PM<br />

Improved sorafenib activity on hepatocellular carcinoma in Notch3 silenced<br />

in vivo and in vitro models. Presenting Author: Michele Baglioni,<br />

Department <strong>of</strong> <strong>Clinical</strong> Medicine, University <strong>of</strong> Bologna, and Center for<br />

Applied Biomedical Research (CRBA), S. Orsola-Malpighi Hospital, Bologna,<br />

Italy<br />

Background: Sorafenib is the only approved drug for the treatment <strong>of</strong> the<br />

advanced stage <strong>of</strong> HCC. Although its efficacy has been proven with<br />

randomized clinical trials, the clinical benefit seen in overall survival for<br />

advanced HCC patients treated with sorafenib could be improved. New<br />

molecules or combination <strong>of</strong> novel targeted agents to improve sorafenib<br />

efficacy for advanced stage HCC patients are needed. Notch genes are a<br />

family <strong>of</strong> receptors involved in many cell fate regulations, their expression<br />

has been found altered in many tumors, including HCC. The aim <strong>of</strong> this<br />

study was to study the combination <strong>of</strong> sorafenib and Notch3 signaling<br />

inhibition to improve sorafenib’s therapeutic effect. Methods: HepG2 and<br />

Huh7 cellular models were used for Notch3 stable silencing by retroviral<br />

introduction <strong>of</strong> specific interfering RNAs Xenograft models in both Notch3<br />

stable shRNA cell lines have been developed using NOD/SCID mice.<br />

Animals bearing tumors were treated with 60 mg/kg <strong>of</strong> sorafenib for 21<br />

consecutive days. Notch3, p21 and pGSK3�Ser9 protein expression were<br />

also analyzed in 20 human HCCs. Results: Notch3 silencing (shN3) in<br />

HuH7 and HepG2 cell lines treated with sorafenib showed an increase in<br />

cell death (3.8 to 5 fold increase) when shN3 cell lines were compared with<br />

their relative controls (GL2). A difference in tumor growth was observed<br />

between GL2 negative control vs shN3 xenografts in both HepG2 and Huh7<br />

after 21 days <strong>of</strong> sorafenib treatment. Two tailed student’s t test (shN3 vs<br />

GL2) P�0,04 and P�0,01 for Huh7 and HepG2 respectively. Molecular<br />

investigations have shown the involvement <strong>of</strong> p21 and GSK3� in the<br />

enhanced response to sorafenib in Notch 3 silenced models. A significant<br />

inverse correlation between Notch3 and pGSK3�Ser9 proteins accumulation<br />

(Spearman �� -0.666) (p � 0.01) and a direct correlation between<br />

Notch3 and p21 proteins expression (Spearman �� 0.681) (p � 0.01) was<br />

found in HCC samples obtained from patients. Conclusion: Our preclincal<br />

findings outline the effect <strong>of</strong> combined Notch3 inhibition and sorafenib.<br />

The molecular mechanisms responsible for sorafenib induced cell death<br />

associated with Notch3 silencing relays on inhibition <strong>of</strong> p21/CDKN1A and<br />

GSK3�Ser9 . This study is supported by a grant from Bayer Healthcare, Italy.<br />

3063 General Poster Session (Board #15E), Mon, 8:00 AM-12:00 PM<br />

Evaluation <strong>of</strong> toxicities <strong>of</strong> target therapy phase I and II trials on glioblastoma<br />

multiforme patients treated by curative radiotherapy: A metaanalysis.<br />

Presenting Author: Marcos Antonio Santos, Institut Gustave<br />

Roussy, Villejuif, France<br />

Background: An important limitation <strong>of</strong> the combination <strong>of</strong> target therapies<br />

(TT) to radiation (RT) is its potentially high level <strong>of</strong> toxicity. The objective <strong>of</strong><br />

this study was to describe and quantify the drug related acute adverse<br />

events presented by patients taking part in phase I and/or phase II trials on<br />

TT combined to curative RT for the treatment <strong>of</strong> glioblastoma multiforme<br />

(GBM). Methods: A meta-analysis <strong>of</strong> summary data including all phase I, I/II<br />

and II trials published between 2000 and 2011 on newly diagnosed GBM<br />

patients treated by TT with RT was performed. Pooled incidence rates (IR)<br />

<strong>of</strong> all and specific toxicities were estimated using a generalized linear<br />

mixed model with fixed and random study effects. The pooled median <strong>of</strong><br />

progression-free (PFS) and overall survivals (OS) were also considered.<br />

These results were compared to those obtained by patients receiving<br />

standard treatment (ST). Results: Twenty-one trials (8 phases I, 3 phases<br />

I/II and 10 phases II), testing 9 different drugs, have been selected,<br />

including 915 patients. The median follow-up varied from 1.4 to 34.0<br />

months. The estimated pooled IR <strong>of</strong> all toxicities was 68.6 [53.5-88.0] per<br />

1000 person-months <strong>of</strong> follow-up, compared to 20.7 [17.6-24.1] on ST<br />

(p�0.001). The pooled IRs <strong>of</strong> thrombocytopenia, neutropenia or leucopenia,<br />

fatigue, gastrointestinal events and treatment-related deaths are 11.2<br />

[7.5-16.9], 9.0 [6.1-13.4], 6.4 [4.1-10.0], 5.5 [2.9-10.2] and 3.2<br />

[1.6-6.4], compared to 4.1 [2.8-5.8], 4.9 [3.5-6.6], 4.6 [3.2-6.4], 0.8<br />

[0.3-1.6] and 0.1 [0.0-0.6], respectively, on ST. Median survival times are<br />

7.7 [6.1-9.8] and 13.8 months [11.9-16.1] on TT studies, and 6.9<br />

[5.8-8.2] and 14.6 [13.2-16.8], on ST, for PFS and OS, respectively,<br />

without statistically significant difference (p�0.39 and 0.55). The heterogeneity<br />

among specific toxicities and survival endpoints may be partially<br />

explained by the study design and/or the intake or not <strong>of</strong> temozolomide.<br />

Conclusions: The use <strong>of</strong> TT combined to RT represented a significative<br />

increase in severe adverse events in patients with GBM compared to ST,<br />

whereas no significant difference was observed in survival endpoints.<br />

Grants from French Cancer League.<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!