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.

158s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2565 General Poster Session (Board #4F), Mon, 8:00 AM-12:00 PM<br />

IFN-� to improve immunotherapy for melanoma. Presenting Author: Marco<br />

Donia, Center for Cancer Immune Therapy, Department <strong>of</strong> Hematology,<br />

Copenhagen University Hospital Herlev, Herlev, Denmark<br />

Background: Adoptive cell therapy (ACT) with tumor infiltrating lymphocytes<br />

(TILs) holds the promise to change the long-term prognosis <strong>of</strong><br />

patients with metastatic melanoma. In this study we have explored<br />

strategies to improve the efficacy <strong>of</strong> cell products for infusion through<br />

tumor immune-sensitization. Methods: We examined whether objective<br />

responses in our pilot ACT trial (NCT937625) were associated with the<br />

number <strong>of</strong> tumor-reactive T cells infused. Subsequently, we assessed the<br />

ability <strong>of</strong> immune-sensitizing agents to modulate the constitutive response<br />

<strong>of</strong> 12 clinical grade TIL products to matched autologous short-term<br />

cultured melanoma cell lines. Results: Our data showed that a high absolute<br />

number <strong>of</strong> infused tumor-reactive T cells is critical to achieve an objective<br />

response. In addition, a defective population <strong>of</strong> tumor-specific CD8� and<br />

CD4� T cells unable to exert antitumor effector functions ex vivo was<br />

detected in most TIL products. However, an antitumor specific response<br />

could be restored by pretreatment <strong>of</strong> the autologous tumor cells with low<br />

dose IFN-�. Of note, IFN-� treatment was invariably associated with<br />

increased cancer immunogenicity as demonstrated by up-regulation <strong>of</strong><br />

MHC molecules. Conclusions: Combination strategies represent the next<br />

frontier <strong>of</strong> cancer immunotherapy. Although previous trials in melanoma<br />

showed that IFN-� does not mediate clinical benefit when used as<br />

monotherapy, our findings show that its ability to increase tumor cell<br />

immunogenicity may enhance the efficacy <strong>of</strong> current immune-based<br />

therapies such as ACT through improved activation <strong>of</strong> pre-existing but<br />

unresponsive tumor specific lymphocytes. Therefore, additional studies on<br />

the possibility <strong>of</strong> a sequential combination <strong>of</strong> low dose IFN-� and ACT are<br />

highly warranted.<br />

2567 General Poster Session (Board #4H), Mon, 8:00 AM-12:00 PM<br />

Human pharmacokinetic (PK) characterization <strong>of</strong> the novel dual-action<br />

anti-HER3/EGFR antibody MEHD7945A (MEHD) in patients with refractory/<br />

recurrent epithelial tumors. Presenting Author: Manuel Hidalgo, START-<br />

Madrid, Centro Integral Oncológico Clara Campal, Madrid, Spain<br />

Background: MEHD is a novel dual-action human IgG1 antibody that blocks<br />

ligand binding to HER3 and EGFR, and elicits antibody-dependent<br />

cell-mediated cytotoxicity (ADCC). MEHD demonstrates single-agent activity<br />

in a broad panel <strong>of</strong> tumor models, including models resistant to<br />

anti-HER3 or anti-EGFR treatment alone. The objective <strong>of</strong> this analysis was<br />

to characterize the PK <strong>of</strong> MEHD associated with body weight (BW)-based<br />

dosing used in a phase I study in patients with epithelial tumors and to<br />

evaluate the potential for using fixed dosing in future studies. Methods:<br />

Preliminary non-compartmental and population PK analyses were performed<br />

using patient data from the dose-escalation stage [1, 4, 10, 15, 22,<br />

and 30 mg/kg every two weeks (q2w)] and expansion stage (14 mg/kg q2w)<br />

<strong>of</strong> the phase I study. Patient demographic data and other relevant clinical<br />

covariates were evaluated in the population analysis. PK simulation <strong>of</strong><br />

1000 subjects with a log-normal BW distribution was performed to<br />

compare the inter-individual variability <strong>of</strong> MEHD exposure following fixed<br />

or BW-based dosing. Results: As expected,MEHD exhibited nonlinear PK.<br />

In the noncompartmenal analysis, the apparent clearance (CL) decreased<br />

in a dose-dependent fashion (about 40 to 9.9 mL/day/kg from 1 to 30<br />

mg/kg) and approached linearity at doses �10 mg/kg (q2w). In the<br />

population analysis, the PK pr<strong>of</strong>ile <strong>of</strong> MEHD was well described by a two<br />

compartment model with linear and nonlinear clearance. The targetmediated<br />

clearance was consistent with that <strong>of</strong> anti-EGFR antibodies. The<br />

nonspecific CL and central volume <strong>of</strong> distribution (V1) values were<br />

approximately 6 mL/day/kg and 52.4 mL/kg, respectively. BW had a<br />

moderate effect on V1, but not on CL. PK simulations suggest that,<br />

compared with BW-based dosing, fixed dosing would result in less<br />

inter-individual variability in MEHD exposure. Both 1100 mg q2w or 1650<br />

mg q3w <strong>of</strong> MEHD achieve the targeted therapeutic exposure. Conclusions:<br />

The dual-action antibody MEHD demonstrated PK consistent with anti-<br />

EGFR antibodies. Fixed dosing <strong>of</strong> MEHD on an every 2 or 3 week schedule<br />

is supported.<br />

2566 General Poster Session (Board #4G), Mon, 8:00 AM-12:00 PM<br />

A first-in-human phase Ib study to evaluate the MEK inhibitor GDC-0973,<br />

combined with the pan-PI3K inhibitor GDC-0941, in patients with advanced<br />

solid tumors. Presenting Author: Patricia LoRusso, Karmanos<br />

Cancer Institute, Detroit, MI<br />

Background: Both RAS/RAF/MEK and PI3K/Akt signaling pathways are<br />

deregulated in many tumor types. Targeting both pathways may be more<br />

efficacious than targeting either pathway alone. In preclinical models,<br />

concurrent administration <strong>of</strong> GDC-0973, a potent, selective, MEK1/2<br />

inhibitor and GDC-0941, a potent class I PI3K inhibitor, shows improved<br />

efficacy compared to either agent alone dosed continuously or intermittently.<br />

Methods: A phase Ib dose-escalation study with 3�3 design was<br />

initiated in patients (pts) with advanced solid tumors to evaluate the safety<br />

and pharmacokinetics (PK) <strong>of</strong> oral dosing <strong>of</strong> GDC-0973 and GDC-0941.<br />

Pts received: concurrent GDC-0973 � GDC-0941 once daily (qd) on a 21<br />

day on/7 day <strong>of</strong>f (21/7) schedule; intermittent GDC-0973 on Days 1, 4, 8,<br />

11, 15, 18 <strong>of</strong> a 28 day cycle � GDC-0941 qd on a 21/7 schedule (MEK<br />

int); or GDC-0973 � GDC-0941 qd on a 7 day on /7 day <strong>of</strong>f schedule (7/7).<br />

Starting doses were 20 mg GDC-0973 � 80 mg GDC-0941 (21/7), 100 mg<br />

GDC-0973 � 130 mg GDC-0941 (MEK int); 40 mg GDC-0973 � 130 mg<br />

GDC-0941 (7/7). Serial plasma PK samples, FDG-PET, and CT scans were<br />

obtained. Results: 78 pts have enrolled. DLTs were G3 lipase (n�1), G4<br />

CPK elevation (n�1). Compared to the 21/7 MTD <strong>of</strong> 40 mg GDC-0973 �<br />

100 mg GDC-0941, higher doses <strong>of</strong> GDC-0973 � GDC-0941 were<br />

tolerated on the MEK int schedule. Overall, adverse events related to the<br />

study drug combination in � 20% pts were diarrhea, rash, nausea, fatigue,<br />

vomiting, decreased appetite, dysgeusia, and elevated CPK. Preliminary<br />

analysis indicated PK <strong>of</strong> GDC-0973 and GDC-0941 are not altered when<br />

dosed in combination. Of 46 evaluable pts, 26 had an FDG-PET partial<br />

metabolic response (� 20% decrease in mean SUVmax from baseline) at �1<br />

time points. <strong>Part</strong>ial responses were observed in 3 pts (mBRAF melanoma,<br />

mBRAF pancreatic ca, mKRAS endometrioid ca); 5 pts had stable disease<br />

� 5 months. Conclusions: Combination dosing <strong>of</strong> GDC-0973 and GDC-<br />

0941 is generally well tolerated, with toxicities similar to those observed in<br />

single agent GDC-0973 and GDC-0941 phase 1 trials. There are early signs<br />

<strong>of</strong> anti-tumor activity. Dose escalation on MEK int and 7/7 schedules<br />

continues and updated data will be presented.<br />

2568 General Poster Session (Board #5A), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> MEHD7945A (MEHD), a first-in-class HER3/EGFR<br />

dual-action antibody, in patients (pts) with refractory/recurrent epithelial<br />

tumors: Expansion cohorts. Presenting Author: Andres Cervantes-Ruiperez,<br />

Department <strong>of</strong> Hematology and Medical Oncology, INCLIVA, University <strong>of</strong><br />

Valencia, Valencia, Spain<br />

Background: Dysregulated human epidermal growth factor receptor tyrosine<br />

kinase (HER RTK) signaling is an important driver <strong>of</strong> tumor growth,<br />

metastasis, and survival. Extensive co-expression and heterodimerization<br />

suggest that simultaneous blockade <strong>of</strong> multiple HER RTKs may be more<br />

effective than blockade <strong>of</strong> a single RTK. MEHD is a novel dual-action<br />

human IgG1 antibody. Each antigen-binding fragment blocks ligand<br />

binding to HER3 or EGFR, and intended to inhibit signaling from all major<br />

ligand-dependent HER dimers. MEHD has single-agent activity in multiple<br />

tumor models including models resistant to anti-HER3 or anti-EGFR.<br />

Methods: This Phase I study evaluated safety, tolerability, pharmacokinetics<br />

(PK), and pharmacodynamic (PD) activity <strong>of</strong> intravenous MEHD every 2<br />

weeks (q2w). Tumor PD assessments were pre- and post-dose FDG-PET<br />

and IHC for pS6, pPRAS40 or pERK in tumor biopsies. Tumor CT<br />

assessments were performed q8w. No dose-limiting toxicity (DLT) was<br />

observed in six 3�3 cohorts from 1-30 mg/kg (n�30). We report results<br />

from 4 tumor-specific cohorts receiving 14 mg/kg MEHD (recommended<br />

Phase II dose). Results: As <strong>of</strong> 21 Nov 2011, 36 pts (CRC�12, NSCLC�9,<br />

SCCHN�10, pancreatic�5), median age 62.5 (35–87), all PS 0-1,<br />

median # prior regimens 3.5 (1-8), received a median <strong>of</strong> 4 doses (1-9) <strong>of</strong><br />

MEHD; 18 pts remain on study. PK data are consistent with human<br />

anti-EGFR antibodies. No related Grade (G) �3 adverse events (AEs) have<br />

been observed. Common related G1/2 AEs included rash/dermatitis (53%),<br />

diarrhea (36%), fatigue (22%), paronychia (19%), dry skin, nausea, and<br />

decreased appetite (all 17%), asthenia and stomatitis/oral pain (all 14%).<br />

Related AEs � 24 h after first infusion included G1/2 headache (42%),<br />

fever (33%), and chills (17%), and decreased in intensity and frequency<br />

with later infusions. Early PD data indicate target inhibition in 8/36 pts<br />

(SCCHN�2, NSCLC�2, CRC�4), and best response by RECIST includes 2<br />

PR (both SCCHN), 6 pts with SD � 8 weeks (NSCLC�2, CRC�4), 7/8<br />

previously treated with EGFR inhibitors. Conclusions: MEHD at 14 mg/kg<br />

q2w has an encouraging safety pr<strong>of</strong>ile and evidence <strong>of</strong> anti-tumor activity.<br />

Phase II studies are planned.<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!