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.

3105 General Poster Session (Board #20G), Mon, 8:00 AM-12:00 PM<br />

A phase Ib trial <strong>of</strong> FOLFIRINOX plus saridegib, an oral hedgehog (Hh)<br />

inhibitor, in pts with advanced pancreatic cancer (PDAC). Presenting<br />

Author: Andrew H. Ko, University <strong>of</strong> California, San Francisco Comprehensive<br />

Cancer Center, San Francisco, CA<br />

Background: FOLFIRINOX has emerged as the optimal 1st-line treatment<br />

option for pts with advanced PDAC and good performance status; whether it<br />

can serve as the backbone upon which to add targeted agents in clinical<br />

trial design remains uncertain. The goal <strong>of</strong> this multicenter phase Ib study<br />

is to evaluate FOLFIRINOX in combination with saridegib, a novel oral<br />

agent that inhibits the Hh signaling pathway. In preclinical models <strong>of</strong><br />

PDAC, saridegib increases chemotherapy delivery by depleting peritumoral<br />

stroma and increasing vascularity. Methods: Pts with previously untreated<br />

metastatic or locally advanced PDAC and ECOG PS 0-1 were eligible.<br />

Treatment consists <strong>of</strong> once-daily saridegib with concurrent administration<br />

<strong>of</strong> biweekly FOLFIRINOX (omitting the 5-FU bolus). A 3�3 dose escalation<br />

design was used (see dose levels below). Prophylactic WBC growth factor<br />

support is mandated. DLT definitions include ALT/AST �10x ULN, grade 4<br />

plts or ANC �5 d, or grade 3-4 nonheme toxicity. CT scans are obtained<br />

every 4 cycles. Limited PK analyses are performed. Results: Seven pts have<br />

been enrolled at the first 2 dose levels. Grade 1-2 AEs include GI (N/V/D),<br />

dehydration, fatigue, and LFT abnormalities. There was one DLT (grade 3<br />

ALT elevation) at DL2. Other serious toxicities seen include grade 3 nausea<br />

(DL1) and grade 3 diarrhea (DL2). Tumor shrinkage has been observed in<br />

all 4 pts at DL1, ranging from 17-54%, with 2 unconfirmed PRs. Final MTD<br />

determination and updated safety and efficacy data will be presented at the<br />

meeting. Conclusions: A modified FOLFIRINOX regimen can be safely<br />

administered in combination with novel agents in clinical trials <strong>of</strong> PDAC.<br />

While saridegib was not beneficial when added to gemcitabine in a separate<br />

randomized phase II study, early evidence <strong>of</strong> significant responses on the<br />

current trial suggests that a more intensive chemotherapy platform may<br />

represent a preferable strategy in PDAC trial design.<br />

Dose level<br />

5-FU bolus<br />

(mg/m2)<br />

5-FU infusion<br />

(mg/m2 x 46 hrs)<br />

LV<br />

(mg/m2) Oxaliplatin Irinotecan<br />

(mg/m2) (mg/m2)<br />

IPI-926<br />

(mg/day)<br />

-1 -- 1600 400 50 120 130<br />

1* -- 1920 400 65 150 130<br />

2 -- 2400 400 85 180 130<br />

3 -- 2400 400 85 180 160<br />

*Starting dose level.<br />

3107^ General Poster Session (Board #21A), Mon, 8:00 AM-12:00 PM<br />

First-in-human phase I dose-escalation study <strong>of</strong> the oral selective C-met<br />

inhibitor EMD 1204831 in patients with advanced solid tumors. Presenting<br />

Author: Hesham M. Amin, Department <strong>of</strong> Hematopathology, University<br />

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

Background: The cell surface receptor tyrosine kinase c-Met and its ligand,<br />

the hepatocyte growth factor, are implicated in tumor cell migration,<br />

invasion, survival, and proliferation. EMD 1204831 is a novel potent and<br />

highly selective reversible, ATP-competitive small molecule c-Met inhibitor.<br />

Methods: This is a phase I, first-in-human clinical trial with escalating<br />

doses <strong>of</strong> EMD 1204831 (NCT01110083). The primary objective was to<br />

determine the maximum tolerated dose (MTD). Secondary objectives<br />

included evaluation <strong>of</strong> safety, pharmacokinetics (PK), pharmacodynamics<br />

(Pd), and preliminary anti-tumor activity. Eligible patients had advanced<br />

solid tumors not amenable to standard therapies. Following a classical 3�3<br />

dose-escalation scheme, successive cohorts <strong>of</strong> patients were treated with<br />

twice daily (BID) oral EMD 1204831 in 21-day cycles. Pd markers were<br />

evaluated in paired tumor biopsies (phospho-c-Met). Results: Until 31<br />

December 2011, 30 patients were enrolled and treated. The dose was<br />

escalated in successive cohorts starting from 50 mg BID up to 1400 mg<br />

BID. After first (single) administration, median Cmax and AUC0–12 values<br />

increased with dose. At higher doses, a decrease in exposure <strong>of</strong> EMD<br />

1204831 was noted after multiple dosing, potentially caused by autoinduction<br />

<strong>of</strong> the compound’s metabolism. Further dose escalation was discontinued,<br />

and no further patients were enrolled. One dose-limiting toxicity (DLT)<br />

<strong>of</strong> grade (G) 3 pancreatitis, considered as a serious adverse event (AE), was<br />

observed at 400 mg BID. No other DLTs or treatment-related serious AEs<br />

were observed. The remaining treatment-related AEs <strong>of</strong> G2 or higher<br />

included G3 and G2 lipase elevation (n�1 for each grade), G2 upper<br />

abdominal pain (n�2), G2 gastroesophageal reflux disease (n�2), and G2<br />

constipation (n�1). Twenty-five patients (83%) had no drug-related<br />

toxicity greater than G1. Of 29 patients evaluable for anti-tumor activity, 3<br />

had stable disease lasting for at least 4 months. Conclusions: Due to<br />

potential autoinduction <strong>of</strong> the compound’s metabolism, dose escalation<br />

was discontinued before an MTD was reached. Final safety, PK, and clinical<br />

tumor response results will be presented.<br />

Developmental Therapeutics—Experimental Therapeutics<br />

199s<br />

3106 General Poster Session (Board #20H), Mon, 8:00 AM-12:00 PM<br />

NRAS mutations in patients with advanced cancers treated with targetbased<br />

therapies in early-phase clinical trials. Presenting Author: Vinu<br />

Madhusudanannair, Department <strong>of</strong> Investigational Cancer Therapeutics<br />

(Phase I Program), University <strong>of</strong> Texas M. D. Anderson Cancer Center,<br />

Houston, TX<br />

Background: NRAS mutations in cancers increase MAPK signaling. It is<br />

plausible that NRAS mutations may be associated with sensitivity to drugs<br />

targeting the MAPK pathway. Methods: Tumors from 689 patients with<br />

advanced cancers referred to the <strong>Clinical</strong> Center for Targeted Therapy<br />

(phase I program) were screened in a CLIA-certified laboratory for NRAS<br />

mutations and if feasible, for PIK3CA, KRAS, BRAF mutations and PTEN<br />

aberrations. Whenever possible, patients with NRAS mutations were<br />

treated with agents targeting the RAF-MEK pathway. Results: Of the 689<br />

patients, 58 (8%) had NRAS mutations. NRAS mutations were most<br />

prevalent in melanoma (26/170, 25%), thyroid (5/26, 19%), endometrial<br />

(2/27, 7%), non-small cell lung (1/31, 3%), ovarian (1/60, 2%) and<br />

colorectal cancers (1/74, 1%). NRAS mutations were not seen in any <strong>of</strong> the<br />

tested head and neck squamous cell cancers (n � 44); sarcomas (n � 41)<br />

and breast cancers (n � 25). None (0/62, 0%) <strong>of</strong> the patients tested for<br />

KRAS had simultaneous NRAS and KRAS mutations, and only 2/42 (5%)<br />

tested for BRAF had simultaneous NRAS and BRAF mutations. Of 58<br />

patients with NRAS mutations, 14 (24%) were enrolled in trials that<br />

included MEK axis inhibitors. Of these 14 patients, 13 had melanoma and<br />

1 had ovarian cancer and 4/14 (29%) demonstrated tumor shrinkage<br />

�10% from baseline. In comparison, only 22/187 (12%) patients with<br />

wild-type/unknown NRAS, KRAS and BRAF treated on the same trials<br />

demonstrated tumor shrinkage �10% (p�0.086). Conclusions: NRAS<br />

mutations are diagnosed in variety <strong>of</strong> advanced cancers. Patients with<br />

NRAS mutations demonstrated a trend to better antitumor activity with<br />

MEK axis inhibitors compared to patients without NRAS, KRAS or BRAF<br />

mutations. Further studies are warranted to delineate the role <strong>of</strong> NRAS<br />

mutations in patients treated with MEK axis inhibitors.<br />

3108 General Poster Session (Board #21B), Mon, 8:00 AM-12:00 PM<br />

Phase I trials <strong>of</strong> PS-341 (bortezomib, B) in combination with topotecan (T)<br />

in advanced solid tumors: A potential pharmacokinetic (PK) interaction.<br />

Presenting Author: Robert Morgan, City <strong>of</strong> Hope, Duarte, CA<br />

Background: Preclinical data has demonstrated that B when combined with<br />

camptothecins enhances apoptotic effects which may be independent <strong>of</strong><br />

NF-�B status and dependent on the sequence <strong>of</strong> exposure to the proteasome<br />

inhibitor. Other data suggests PK interactions <strong>of</strong> B with some<br />

chemotherapeutic agents. It is hypothesized that B will act synergistically<br />

with T and enhance the apoptotic effects <strong>of</strong> T. Conflicting data exists<br />

regarding schedule dependent synergistic cytotoxicity <strong>of</strong> the combination.<br />

This study examined both sequences <strong>of</strong> administration and determined the<br />

PK <strong>of</strong> each sequence. Methods: A standard 3 � 3 schedule was used in both<br />

sequences (S). The dose <strong>of</strong> B was fixed with escalating doses <strong>of</strong> T. S1: B 1.3<br />

mg/m2 on days 1, 4, 8, and 11.T2to3.5mg/m2 on days 1 and 8 was given<br />

6 h before B. S2: B 1.3 mg/m2 on days 1, 4, 8, 11 followed by T2to3<br />

mg/m2 given 6 h after B. Cycle length was q28 days. PK sampling for both<br />

sequences was performed. Results: 54 pts (S1: 27, S2: 27) received a<br />

median <strong>of</strong> 2 cycles (range 1-7) <strong>of</strong> therapy. Pts were heavily pre-treated. For<br />

S1, the maximum tolerated dose (MTD) <strong>of</strong> B and T were 1.3 and 3 mg/m2 respectively. For S2, the MTD <strong>of</strong> B and T were 1.3 and 2.5 mg/m2 respectively. Dose-limiting toxicities (2 pt in each sequence) were gr 3<br />

thrombocytopenia in S1, and gr 4 thrombocytopenia in S2. Stable disease<br />

was seen in 6/27 pts in S1, and 6/27 in S2. PK results for S2 show no<br />

significant difference between the day 1 and day 8 PKs. However, the mean<br />

AUC (454�150 mg/Lxhr) and Cmax (123�51 mg/L) at the MTD <strong>of</strong> 2.5<br />

mg/m2 reached on this trial is similar to those reported with single agent<br />

doses <strong>of</strong> 4 mg/m2 given over 30 minutes weekly x 3. As a result, the<br />

estimated topotecan clearance (11.3�5 L/hr) when given following bortezomib<br />

is approximately one half <strong>of</strong> the reported clearance for the single<br />

agent. (Curtis et al, J Clin Pharmacol, 50:268, 2010). Conclusions: The<br />

tolerability and toxicity <strong>of</strong> both sequences was similar. MTD differs with S2<br />

having a lower tolerated dose <strong>of</strong> T. PK results suggest a possible drug-drug<br />

interaction with decreased clearance <strong>of</strong> T with S2. PK results for S1 are<br />

pending. Supported by CCSG CA62505.<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!