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.

160s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2573^ General Poster Session (Board #5F), Mon, 8:00 AM-12:00 PM<br />

Messenger RNA vaccination and B-cell responses in NSCLC patients.<br />

Presenting Author: Martin Sebastian, Medizinische Klinik II Hämatologie/<br />

Onkologie, Rheumatologie, Infektiologie, HIV Klinikum der J.W. Goethe-<br />

Universität Frankfurt, Frankfurt am Main, Germany<br />

Background: Vaccination with mRNA is a novel technology in cancer<br />

immunotherapy. CV9201 consists <strong>of</strong> self-adjuvanted mRNA molecules<br />

(RNActive) coding for five non small cell lung cancer (NSCLC)-associated<br />

tumor antigens (MAGE-C1, MAGE-C2, NY-ESO-1, BIRC5, 5T4). We report<br />

final results <strong>of</strong> a phase I/IIa trial <strong>of</strong> CV9201 in patients (pts.) with NSCLC.<br />

Methods: Pts. with stage IIIB/IV NSCLC with a response or stable disease<br />

after first-line chemotherapy or chemoradiation were eligible. Cohorts <strong>of</strong> 3<br />

pts. were treated at three dose levels (400�g, 800�g and 1600�g<br />

CV9201) and observed for DLTs to select the highest tolerated dose for<br />

phase IIa. Primary endpoint was safety and tolerability; secondary endpoints<br />

were immune response, clinical efficacy and survival. Pts. received<br />

up to five vaccinations <strong>of</strong> CV9201 within 15 weeks. Antigen-specific<br />

immune responses against each <strong>of</strong> the 5 antigens were measured at<br />

baseline, and two weeks after the 3rd and 5th vaccination. Frequency <strong>of</strong><br />

lymphocyte subsets and expression <strong>of</strong> activation and maturation markers<br />

were measured and retrospectively correlated with immunological and<br />

clinical parameters. Results: 46 pts. were included (9 phase I; 37 phase<br />

IIa); No DLTs occurred, and the 1600 �g dose was investigated in phase<br />

IIa. The most frequent related adverse events were mild to moderate<br />

injection site reactions and flu-like symptoms. 3 patients (7%) had<br />

potentially related grade 3 AEs (fatigue, injection site granuloma, asthma<br />

attack) and no related SAEs were reported. There were no objective<br />

responses. Data on PFS and survival will be presented. Antigen specific<br />

immune responses against at least one antigen were induced in 65% <strong>of</strong> pts.<br />

(39% cellular and 49% humoral). Consistently, a significant �2 fold<br />

increase <strong>of</strong> pre germinal center B cells (pGCB) was observed in 61% <strong>of</strong> pts.<br />

This increase <strong>of</strong> pGCB correlated significantly (p�0.0028) with increase <strong>of</strong><br />

total CD4 effector T cells. Frequency <strong>of</strong> CD 4 T Reg cells did not increase<br />

during treatment. Conclusions: Vaccination with CV9201 has a favorable<br />

safety pr<strong>of</strong>ile and induces T and B cell responses against all included<br />

antigens. Vaccine-induced increase <strong>of</strong> pGCB is a new finding and might be<br />

used as a biomarker in cancer immunotherapy.<br />

2575 General Poster Session (Board #5H), Mon, 8:00 AM-12:00 PM<br />

Bispecific Fynomer-antibody fusion proteins targeting two epitopes on<br />

HER2. Presenting Author: Frederic Mourlane, Covagen AG, Zurich-<br />

Schlieren, Switzerland<br />

Background: Fynomers are small binding proteins (7 kDa) derived from the<br />

SH3 domain <strong>of</strong> Fyn kinase. They can be engineered to yield specific and<br />

high-affinity binding domains to target proteins <strong>of</strong> interest. In the past, we<br />

have isolated Fynomers with excellent physico-chemical properties binding<br />

to 16 different antigens. One important application <strong>of</strong> the Fynomer<br />

technology represents the genetic fusion <strong>of</strong> a Fynomer to an antibody to<br />

provide bispecific fusion proteins. Methods: Using phage display technology<br />

we have isolated Fynomers binding to tumor-associated antigens. After<br />

genetic fusion <strong>of</strong> these Fynomers to antibodies <strong>of</strong> interest the resulting<br />

bispecific proteins were evaluated in vitro and in vivo for their antitumoral<br />

activity. Results: The fusion <strong>of</strong> Fynomers to the antibodies <strong>of</strong> interest did not<br />

alter the favorable biophysical properties <strong>of</strong> the antibodies. First, the<br />

Fynomer-antibody fusion proteins could be purified with the same high<br />

yields from the supernatant <strong>of</strong> transiently transfected CHO cells as the<br />

unmodified antibodies (in the range <strong>of</strong> 100 mg/L). Second, the purified<br />

fusion proteins were monomeric and showed no signs <strong>of</strong> aggregation even<br />

after four months <strong>of</strong> storage at 4 °C or -20 °C in PBS as determined by size<br />

exclusion chromatography. Third, the Fc-mediated effector functions <strong>of</strong><br />

antibodies were not altered by their fusion with Fynomers. Antibodydependent<br />

cellular cytotoxicity (ADCC) was maintained, and the affinity to<br />

the neonatal Fc-receptor (FcRn) was similar as observed for the unmodified<br />

antibody. In addition, the bispecific Fynomer-antibody fusion proteins<br />

demonstrated excellent growth inhibition <strong>of</strong> tumor cells in vitro and high<br />

efficacy in vivo in tumor xenograft mouse models. Conclusions: These<br />

encouraging preclinical results indicate that the bispecific Fynomerantibody<br />

fusions have highly promising properties with a great potential for<br />

further preclinical and clinical development.<br />

2574 General Poster Session (Board #5G), Mon, 8:00 AM-12:00 PM<br />

Adoptive cell therapy for melanoma patients using tumor-infiltrating<br />

lymphocytes, lymphodepleting chemotherapy, and low-dose interleukin-2.<br />

Presenting Author: Eva Ellebaek, Department <strong>of</strong> Oncology and Center for<br />

Cancer ImmuneTherapy, Department <strong>of</strong> Hematology, Copenhagen University<br />

Hospital Herlev, Herlev, Denmark<br />

Background: Adoptive T cell therapy is based on isolation <strong>of</strong> tumor<br />

infiltrating lymphocytes (TIL) from autologous tumor, in vitro activation and<br />

expansion, and the reinfusion <strong>of</strong> these cells into a lymphodepleted patient.<br />

Together with high-dose interleukin (IL)-2 this treatment has in a few other<br />

countries successively been given to patients with advanced malignant<br />

melanoma and an impressive 50% response rate has been observed. Here<br />

we report the experience from a Danish Translational Research Center<br />

using low-dose subcutaneous IL-2 injections. Methods: A pilot trial including<br />

patients with metastatic melanoma, PS �1, age �70, measurable and<br />

progressive disease, and no CNS involvement. Six patients were treated<br />

with lymphodepleting chemotherapy, TIL infusion, and 14 days <strong>of</strong> low-dose<br />

IL-2, 2 MIU/day. Results: Low-dose IL-2 considerably decreased the<br />

toxicity <strong>of</strong> the treatment with no grade 3-4 events related to this drug. Two<br />

<strong>of</strong> the 6 treated patients have an ongoing complete response (30� and 7�<br />

months), 2 patients had stable disease (4.5 and 5 months) and 2 patients<br />

progressed shortly after treatment. Five patients went through surgery but<br />

were not treated because <strong>of</strong> rapid disease progression (2), development <strong>of</strong><br />

brain metastases (2) or the inability to grow cells (1). Tumor-reactivity <strong>of</strong><br />

the infused cells and peripheral monocytes before and after therapy were<br />

analyzed. High tumor responses in the infusion products were correlated to<br />

clinical response and also an induction <strong>of</strong> tumor reactive T cells were seen<br />

in the peripheral blood for 1 patient in complete response. Conclusions:<br />

Complete and durable responses are induced after treatment with adoptive<br />

cell transfer in combination with low-dose IL-2 questioning the need for<br />

high and toxic doses <strong>of</strong> IL-2.<br />

2576 General Poster Session (Board #6A), Mon, 8:00 AM-12:00 PM<br />

A phase I study <strong>of</strong> the safety and pharmacodynamic effects <strong>of</strong> everolimus in<br />

combination with lenalidomide in patients with advanced solid malignancies.<br />

Presenting Author: Ta<strong>of</strong>eek Kunle Owonikoko, Winship Cancer Institute,<br />

Emory University, Atlanta, GA<br />

Background: Everolimus (E), an mTOR inhibitor, and Lenalidomide (L) are<br />

both potent anti cancer agents with immunomodulatory effects. Preclinical<br />

evidence <strong>of</strong> enhanced cytotoxicity <strong>of</strong> the combination <strong>of</strong> an mTOR inhibitor<br />

and lenalidomide provided the rationale for this phase I study. Methods:<br />

Patients with advanced solid malignancies, ECOG performance status (PS)<br />

0-2 and adequate organ function were eligible. Using standard 3�3 dose<br />

escalation schema, patients were treated in cohorts <strong>of</strong> three with increasing<br />

doses <strong>of</strong> E (5mg, 10mg) and L (10, 15, 20, 25mg) on day 1-28 <strong>of</strong> a 28-day<br />

cycle. Treatment cycles were repeated until disease progression by RECIST<br />

criteria or intolerable toxicity. Pharmacodynamic (PD) effects <strong>of</strong> treatment<br />

on circulating B and T lymphocytes subsets were assessed by multiparameter<br />

flow cytometry at baseline and after 2 cycles. Results: We enrolled 21<br />

patients (thyroid-5, salivary gland-5, colon-4, sarcoma-2, and others-5);<br />

median age-58 (29-74); male-13; ECOG PS <strong>of</strong> 0, 1, 2 (3/17/1). Salient<br />

grade 3/4 toxicities included rash (67%), anemia (19%) and vomiting (5%)<br />

without dose limiting toxicities. The median number <strong>of</strong> completed cycles<br />

was3(1-�15) and best response in 14 <strong>of</strong> 18 evaluable patients was<br />

stable disease (range 2- �14 months); median PFS was 116 days (14-<br />

�498). The RP2D <strong>of</strong> E and L was defined as 10mg and 25mg once daily,<br />

respectively. PD endpoint analysis after 2 cycles <strong>of</strong> treatment showed a<br />

significant increase in activated cytotoxic T cell subset (CD8�ICOS1�) in<br />

patients with salivary or thyroid cancers compared to other tumor types<br />

(�2051.0 vs. �394 vs. -1687.4 respectively; p�0.0303) and a trend for<br />

an increase in patients with non-progressing tumors (�661.7 vs. -384.1;<br />

p�0.0988). Other significant correlations were: Changes in total B-cells<br />

(CD3-CD19�) with PFS; NK cells with age (p�0.0211) and activated T<br />

cells (CD3�CD69�; CD3�CD62L-; p�0.01) with gender. Conclusions:<br />

The combination <strong>of</strong> E and L is well tolerated at the recommended single<br />

agent doses and showed promising efficacy in neuroendocrine and salivary<br />

adenoidcystic carcinoma. Modulation <strong>of</strong> activated T cell subsets<br />

(CD8�ICOS1�) correlates with efficacy <strong>of</strong> these agents.<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!