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.

200s Developmental Therapeutics—Experimental Therapeutics<br />

TPS3109 General Poster Session (Board #21C), Mon, 8:00 AM-12:00 PM<br />

Vorinostat to restore sensitivity to aromatase inhibitor therapy in metastatic<br />

breast cancer: A phase II clinical trial with ER imaging correlates.<br />

Presenting Author: Hannah M. Linden, University <strong>of</strong> Washington, Seattle,<br />

WA<br />

Background: Endocrine refractory,Indolent, s<strong>of</strong>t tissue, and bone dominant<br />

metastatic breast cancer is a clinical problem, for which alternatives to<br />

chemotherapy are needed, Histone deacetylace inhibitors (HDACi) have<br />

shown pre-clinical promise in estrogen receptor-modulation and restoring<br />

sensitivity to endocrine manipulation, suggesting potential clinical benefit.<br />

HDACi restore ER expression in ER-negative cells which are then sensitive<br />

to AI treatment in xenografts (Sabnis 2011) and are toxic to ER-positive<br />

cell lines (Huang 2000). Vorinostat is an FDA approved HDACi for CTCL,<br />

and could have a beneficial role in restoring ER-signaling in endocrineresistant<br />

tumors. We hypothesized that HDACi therapy could restore<br />

sensitivity to aromatase inhibitor (AI) therapy, in patients with prior<br />

progression on AI, and allow continued endocrine therapy. Recent data<br />

suggest clinical benefit <strong>of</strong> Tamoxifen and vorinostat given continuously<br />

(Munster 2011) as well as entinostat and an AI (Yardley 2011). Our prior<br />

work has shown the feasibility <strong>of</strong> monitoring regional ER expression and<br />

ER-estrogen binding in vivo using [F-18]fluoroestradiol (FES) PET imaging<br />

(Linden 2011) providing a biomarker to interrogate the molecular target.<br />

Methods: Patients with metastatic breast cancer with prior clinical benefit<br />

from endocrine manipulation who progressed on AI therapy are eligible.<br />

Patients must be <strong>of</strong>f ER blocking therapies, and functionally postmenopausal.<br />

Baseline FES and FDG PET are performed and patients receive<br />

investigational vorinostat treatment at 400 mg po BID for 2 weeks followed<br />

by serial FES and FDG PET to assess the impact <strong>of</strong> vorinostat on ER<br />

expression and tumor metabolism. Patients are retreated on their prior AI as<br />

a single agent for 6 weeks, followed by paired FES and FDG and clinical<br />

assessment. Patients with clinical benefit may continue on treatment: 2<br />

weeks <strong>of</strong> vorinostat followed by 6 weeks <strong>of</strong> AI in 8-week cycles until<br />

progressive disease or toxicity. We have enrolled 4 <strong>of</strong> a planned 20 patients.<br />

TPS3111 General Poster Session (Board #21E), Mon, 8:00 AM-12:00 PM<br />

A phase I and pharmacologic study <strong>of</strong> MM-111, a bispecific HER2/HER3<br />

antibody fusion protein, in combination with multiple treatment regimens<br />

in patients with advanced HER2-positive solid tumors. Presenting Author:<br />

Donald A. Richards, Texas Oncology-Tyler, Tyler, TX<br />

Background: The HER2/HER3 oncogenic heterodimer is a potent HER<br />

receptor pairing with respect to strength <strong>of</strong> interaction, receptor tyrosine<br />

phosphorylation, and downstream signaling through mitogen activated<br />

protein kinase and phosphoinositide-3 kinase pathways. MM-111 is a novel<br />

bispecific antibody fusion protein that inhibits ligand activated HER3<br />

signaling through abrogation <strong>of</strong> ligand binding specifically in HER2<br />

overexpressing tumors. In preclinical models <strong>of</strong> HER2� solid tumors,<br />

MM-111 inhibits ligand-induced HER3 phosphorylation, cell cycle progression,<br />

and tumor growth. HER2 is a well-established target <strong>of</strong> anticancer<br />

agents such as trastuzumab and lapatinib, specifically in breast and gastric<br />

tumors. Preclinical data demonstrate that MM-111 potentiates the antitumor<br />

activity <strong>of</strong> both trastuzumab and lapatinib and this phase I study seeks<br />

to determine if MM-111 can be safely used in combination with various<br />

standard <strong>of</strong> care HER2 targeting regimens, namely; (1) trastuzumab,<br />

cisplatin and, capecitabine, (2) trastuzumab and lapatinib, and (3)<br />

trastuzumab and paclitaxel. Methods: This is a multi-arm phase I, open<br />

label, multicenter, dose-escalation study <strong>of</strong> MM-111 in an add-on design<br />

that evaluates the safety, pharmacokinetics (PK), and anti-tumor activity <strong>of</strong><br />

MM-111 in combination. Each arm is designed to run as a separate phase I<br />

study to address safety and tolerability <strong>of</strong> each combination. For eligibility,<br />

patients are required to have documented advanced HER2-positive cancer,<br />

with adequate bone marrow, hepato-renal and cardiac function. The dose<br />

escalation for each arm utilizes a standard “3 � 3” design with MM-111<br />

dosed initially at a moderate dose <strong>of</strong> 10 mg/kg and escalated up to 20<br />

mg/kg to identify a phase II dose in combination with each regimen. Once<br />

the maximum tolerated dose or phase II dose is identified, expansion<br />

cohorts will accrue to further evaluate these combinations. The flexibility <strong>of</strong><br />

the design allows additional combinations arms to be added. Key exploratory<br />

analyses will include an evaluation <strong>of</strong> safety, PK, and efficacy as<br />

evidenced by best overall response and duration <strong>of</strong> response.<br />

TPS3110 General Poster Session (Board #21D), Mon, 8:00 AM-12:00 PM<br />

Design <strong>of</strong> a phase I clinical trial with PNT2258, a novel DNA interference<br />

oligonucleotide (DNAi), in patients with advanced solid tumors. Presenting<br />

Author: Drew Warren Rasco, South Texas Accelerated Research Therapeutics,<br />

LLC, San Antonio, TX<br />

Background: With the knowledge that Bcl-2 facilitates drug resistance and<br />

cell survival, a DNA interference (DNAi) strategy was applied to silence<br />

Bcl-2 in cancer cells and promote apoptosis. DNAi differs from cytoplasmic<br />

mRNA targeting (antisense, RNAi, and miRNA targets) as it targets<br />

genomic DNA, blocking transcription. PNT100, a first in class DNAi, is a<br />

novel single-stranded 24-base unmodified DNA designed to bind to an<br />

upstream region <strong>of</strong> the Bcl-2 promoter. The drug product (PNT2258) is<br />

PNT100 encapsulated in a specialized pH tunable liposome and is being<br />

assessed for safety and tolerability in a phase I trial. PNT2258 avoids the<br />

toxicities associated with modified oligonucleotides and double-stranded<br />

RNAs; since the liposome formulation is anionic and contains no surface<br />

spacers, vehicle toxicities are minimal. Xenograft experiments demonstrated<br />

marked single agent activity in a diffuse large cell lymphoma, and<br />

therapy potentiation when combined with either rituximab in Daudi-<br />

Burkitt’s Lymphoma or docetaxel in A375 melanoma. Methods: An openlabel,<br />

single-arm, first-in-man phase I dose-escalation study <strong>of</strong> PNT2258<br />

in patients with advanced solid tumors was designed to evaluate safety,<br />

tolerability, dose-limiting toxicities, pharmacokinetics, and pharmacodynamics<br />

<strong>of</strong> PNT2258 to recommend a dose for phase II studies. In this<br />

phase I study, pharmacodynamic effects <strong>of</strong> PNT2258 will be evaluated<br />

through analyses <strong>of</strong> soluble serum and plasma markers and peripheral<br />

blood mononuclear cells. Patients will receive PNT2258 as an intravenous<br />

infusion over 2 hours once daily for 5 consecutive days (days 1-5) <strong>of</strong> each<br />

21-day treatment cycle (3 weeks). The starting dose <strong>of</strong> 1 mg/m2 with<br />

PNT2258 administered to one patient per cohort and dose-escalation will<br />

proceed by dose-doubling in each successive cohort until a dose level <strong>of</strong> 64<br />

mg/m2 is attained, provided no dose-limiting toxicities are observed in<br />

cycle 1. Thereafter, dose escalations shall proceed at 33% increments <strong>of</strong><br />

the previous cohort dose-level to 85, 113, and 150 mg/m2 with expansions<br />

<strong>of</strong> up to six patients per cohort as needed. The ten planned dose cohorts<br />

have been completed with all patients enrolled.<br />

TPS3112 General Poster Session (Board #21F), Mon, 8:00 AM-12:00 PM<br />

A phase I trial <strong>of</strong> riluzole and sorafenib in patients with advanced solid<br />

tumors and melanoma. Presenting Author: Janice M. Mehnert, Cancer<br />

Institute <strong>of</strong> New Jersey, New Brunswick, NJ<br />

Background: Metabotropic glutamate receptor 1 (GRM1) has been identified<br />

as a potential therapeutic target in melanoma. Over 60% <strong>of</strong> human<br />

melanomas express this cell surface glutamate receptor and excitation <strong>of</strong><br />

GRM1 results in the activation <strong>of</strong> MAPK and PI3K/AKT pathways. Riluzole,<br />

an oral GRM1 blocking agent, results in growth arrest <strong>of</strong> melanoma cells in<br />

vitro and in vivo. We previously reported that administration <strong>of</strong> riluzole to<br />

melanoma patients suppressed activity <strong>of</strong> the PI3K/AKT and MAPK<br />

pathways in paired tumor samples (Yip Clin Cancer Res 2009). In<br />

preclinical studies, the efficacy <strong>of</strong> riluzole was attenuated in melanoma<br />

cells harboring BRAFV600E mutations, but sorafenib, a RAF kinase<br />

inhibitor, enhanced the effect <strong>of</strong> riluzole on these cells. The combination <strong>of</strong><br />

riluzole and sorafenib was additive or synergistic in both BRAF mutant and<br />

BRAF wildtype melanoma cells in vitro and in BRAF wildtype cells in a<br />

xenograft model (Lee HJ Clin Cancer Res 2011). We thus designed a phase<br />

I trial to test the combination <strong>of</strong> riluzole with sorafenib in patients with solid<br />

tumors and advanced melanoma. Methods: The primary objective <strong>of</strong> this<br />

trial is identification <strong>of</strong> the maximum tolerated dose (MTD). An expansion<br />

cohort at the MTD is planned for patients with advanced melanoma to<br />

examine the correlation <strong>of</strong> clinical or radiographic response with signaling<br />

through the MAPK and PI3K/AKT pathways and with GRM1 receptor status<br />

<strong>of</strong> individual tumors. Eligible patients must have advanced solid tumors<br />

(phase I) or stage III unresectable/stage IV melanoma with biopsiable tumor<br />

(expansion cohort) and ECOG PS � 2. Riluzole will be administered at 100<br />

mg twice daily combined with sorafenib beginning at 200 mg daily and<br />

escalating in subsequent cohorts at 200 mg increments. Correlative<br />

studies: Tumors will be assessed for BRAF and NRAS mutational status.<br />

Pretreatment tumor blocks will be examined for GRM1 receptor status by<br />

immunohistochemistry. Pre and post treatment levels <strong>of</strong> pERK and pAKT<br />

will be measured in paired tumor samples to assess effects <strong>of</strong> treatment on<br />

MAPK and PI3K signaling. Limited sampling pharmacokinetic studies will<br />

be performed. Progress: Accrual to three cohorts is complete without DLT.<br />

Accrual to the final planned cohort is in progress.<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!