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.

146s Developmental Therapeutics—<strong>Clinical</strong> Pharmacology and Immunotherapy<br />

2517 Poster Discussion Session (Board #5), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

A phase I safety study <strong>of</strong> NPC-1C: A novel, therapeutic antibody to treat<br />

pancreas and colorectal cancers. Presenting Author: Philip M. Arlen,<br />

Neogenix Oncology, Rockville, MD<br />

Background: NPC-1C (NEO-101; Ensituximab) is a chimeric monoclonal<br />

antibody being developed as a novel biological treatment for pancreatic and<br />

colorectal cancers. This antibody was selected from a panel <strong>of</strong> hybridomas<br />

generated from mice immunized with semi-purified membrane-associated<br />

proteins derived from biologically screened, pooled human allogeneic colon<br />

cancer tissues. The NPC-1C target appears to be a variant <strong>of</strong> MUC5AC that<br />

is expressed specifically by human colon and pancreatic tumors with<br />

minimal cross-reactivity to normal GI tract tissues. Methods: A phase I open<br />

label, multi-center dose escalation clinical trial with NPC-1C has completed<br />

accrual <strong>of</strong> subjects with advanced pancreatic and colorectal cancer<br />

refractory to standard therapy. The primary objectives are to determine<br />

safety and tolerability <strong>of</strong> escalating doses <strong>of</strong> NPC-1C and to assess<br />

pharmacokinetics (PK) and select immune responses to the antibody at<br />

each dose level. Secondary objectives are to evaluate evidence <strong>of</strong> clinical<br />

benefit and to explore immunologic correlates Results: Three cohorts <strong>of</strong><br />

subjects have been treated with NPC-1C at escalating dose levels <strong>of</strong> 1, 1.5,<br />

and 2 mg/kg IV every two weeks. All 15 subjects (10 colorectal, 5<br />

pancreatic) have enrolled on study (5/15 non-evaluable). No DLT was noted<br />

at the 1mg/kg dose level with 2 <strong>of</strong> 3 patients showing stable disease at<br />

completion <strong>of</strong> Course 1. At the 2 mg/kg dose level, one patient demonstrated<br />

stable disease after course 1. At escalating rates <strong>of</strong> infusion, 3<br />

subjects experienced mild to moderate hemolysis that resolved without<br />

intervention. All 6 patients who have received 1.5 mg/kg at a constant rate<br />

<strong>of</strong> infusion have been evaluated, with no dose limiting toxicity. One <strong>of</strong> 5<br />

patients re-staged thus far in the 1.5 mg/kg cohort showed stable disease<br />

on scans after completing the first course <strong>of</strong> treatment (4 doses), and<br />

continued to receive additional doses. Preliminary data show no drug<br />

induced antibody responses in treated subjects. PK, HAMA and cytokine<br />

evaluation is ongoing. Conclusions: NPC-1C is tolerated well at the current<br />

dose level <strong>of</strong> 1.5 mg/kg every 2 weeks without producing any DLT We plan<br />

to continue accrual in the Phase 2A to evaluate clinical responses and<br />

additional safety data.<br />

2519 Poster Discussion Session (Board #7), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Assessment <strong>of</strong> a novel immunological biomarker SNP panel from a phase<br />

III trial <strong>of</strong> Bacillus Calmette-Guérin (BCG) adjuvant treatment in stage III<br />

melanoma patients. Presenting Author: Connie Ging Ting Chiu, Department<br />

<strong>of</strong> Molecular Oncology and Division <strong>of</strong> Surgical Oncology, John Wayne<br />

Cancer Institute, Santa Monica, CA<br />

Background: Melanoma is an immunogenic cancer, whereby immunotherapy<br />

in stage III patients has shown some success. However, there are no<br />

effective immune biomarker tests to identify patients most likely to benefit<br />

from immunotherapy. BCG (Bacillus Calmette-Guérin) therapy is a form <strong>of</strong><br />

immunomodulation. The objective was to assess the predictive clinical<br />

effect <strong>of</strong> a single nucleotide polymorphism(SNP) immune biomarker panel<br />

in stage III resected melanoma patients treated with BCG. Methods:<br />

MMAIT-III was a phase III prospective randomized international multicenter<br />

trial <strong>of</strong> BCG�melanoma vaccine vs BCG�placebo after complete<br />

resection <strong>of</strong> stage III melanoma patients (NIH #NCT00052130). 120/292<br />

patients with palpable disease treated with resection and BCG�placebo<br />

had lymphocytes(PBL) from USA sites available for analysis. Endpoints<br />

were overall survival(OS) and disease-free survival(DFS), with a 10-yr<br />

follow-up. PBL DNA was assessed by MassARRAY MALDI-TOF for 28 SNPs<br />

associated with macrophage/monocyte-related immune response pathways<br />

to BCG/tuberculosis. A pilot study(n�34) from phase II BCG trial confirmed<br />

presence <strong>of</strong> the SNPs. A logistic regression determined a SNP score,<br />

and a cut<strong>of</strong>f was identified by ROC and used as a predictor in a Cox<br />

proportional hazard model in a verification study(n�120). Results: 9 SNPs<br />

in 6 genes had prognostic value: NRAMP1 and CD14, 18, 195, 209, 282.<br />

The 9-SNP panel distinguished patients in 2 survival groups(OS median<br />

4.9-yrs vs 1.5-yrs, p�0.0008), AUC�0.77. SNP biomarker positivity<br />

demonstrates significant association with 10-yr OS (59.7% vs 15.7%; HR<br />

1.97, CI 1.11-3.50, p�0.018) and DFS (47.2% vs 12.3%; HR 2.35, CI<br />

1.43-3.92, p�0.0007). In Cox model, the SNP panel was a significant<br />

predictor <strong>of</strong> OS(HR 2.69, CI 1.56-9.00, p�0.0052) and DFS(HR 2.33, CI<br />

1.49-5.16, p�0.0003) independent <strong>of</strong> known melanoma prognostic<br />

factors. Conclusions: The 9-SNP panel identified patients with an exceptionally<br />

favourable disease outcome, and may represent a stratifying predictive<br />

SNP panel for identifying patients that are inherently responsive to BCG<br />

therapy and potentially other immunomodulating agents in melanoma.<br />

2518 Poster Discussion Session (Board #6), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Myeloid-derived suppressor cell quantity prior to treatment with ipilimumab<br />

at 10mg/kg to predict for overall survival in patients with metastatic<br />

melanoma. Presenting Author: Shigehisa Kitano, Memorial Sloan-<br />

Kettering Cancer Center, New York, NY<br />

Background: Ipilimumab, an antibody that blocks the function <strong>of</strong> the<br />

immune inhibitory molecule cytotoxic T lymphocyte antigen 4 (CTLA-4),<br />

significantly prolongs survival in patients with metastatic melanoma.<br />

Approximately 30% <strong>of</strong> patients derive clinical benefit from therapy.<br />

Defining biomarkers <strong>of</strong> response to ipilimumab therapy would enable<br />

selection <strong>of</strong> patients more likely to respond and is relevant for both<br />

practicing clinicians and for clinical trial design. We performed a pilot<br />

correlative study evaluating myeloid derived suppressor cells (MDSC), a<br />

population <strong>of</strong> immune suppressive monocytic cells, as a biomarker <strong>of</strong><br />

clinical outcome. Methods: Peripheral blood from 26 patients with stage IV<br />

melanoma treated with ipilimumab 10mg/kg every 3 weeks for 4 doses at<br />

our center, as part <strong>of</strong> an expanded access program (BMS CA184-045) was<br />

assessed for MDSC quantity (%CD14� ,HLA-DRlow/- cells) pre-treatment, at<br />

week 7, week 12, and week 24 by flow cytometry. MDSC ability to inhibit T<br />

cell proliferation was tested using an in vitro suppression assay. Results: We<br />

found that lower MDSC quantity pre-treatment predicted for improved<br />

overall survival (Hazard ratio 1.07 (1.03, 1.11) p�0.002) and trended<br />

toward associating with clinical benefit measured at week 24 imaging<br />

(p�0.09). This effect was independent <strong>of</strong> pre-treatment or week 7 absolute<br />

lymphocyte counts (ALC) and pre-treatment LDH when evaluated in a<br />

multivariate model with ALC and MDSC quantity HR 1.10; 95% CI 1.04,<br />

1.17 p�0.0006 and LDH and MDSC quantity HR 1.06; 95% CI 1.01,<br />

1.11 p � 0.013. Furthermore, a general trend <strong>of</strong> increasing MDSC number<br />

by week 24 from the pre-treatment baseline was associated with patients<br />

that did not achieve clinical benefit. MDSC suppressed peripheral blood T<br />

cell proliferation as measured by CFSE dilution in response to anti-CD3<br />

antibody stimulation. Conclusions: Pre-treatment MDSC quantity may<br />

predict clinical response following ipilimumab therapy. Further studies<br />

evaluating MDSC as a biomarker <strong>of</strong> ipilimumab therapy are warranted both<br />

retrospectively and prospectively in a broader group <strong>of</strong> patients.<br />

2520^ Poster Discussion Session (Board #8), Sat, 8:00 AM-12:00 PM and<br />

12:00 PM-1:00 PM<br />

Investigating genes and pathways that play a role in immune responses<br />

mediated by anti-CTLA-4 therapy. Presenting Author: Jianjun Gao, University<br />

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

Background: Blockade <strong>of</strong> the inhibitory T cell molecule CTLA-4 with a<br />

monoclonal antibody has led to enhanced anti-tumor immune responses<br />

and clinical benefit. Ipilimumab, an anti-CTLA-4 antibody (BMS), was<br />

recently FDA-approved for the treatment <strong>of</strong> metastatic melanoma. Only a<br />

subset <strong>of</strong> patients benefit from anti-CTLA-4. In order to identify genes and<br />

pathways that are induced by anti-CTLA-4, which may be used in future<br />

studies for potential correlation with clinical outcomes or provide additional<br />

targets for therapy, we purified and analyzed CD4 T cells from<br />

patients treated with anti-CTLA-4 for changes in gene expression pr<strong>of</strong>ile.<br />

We also obtained tumor tissues from treated patients for similar studies.<br />

Methods: On an IRB-approved Phase Ia pre-surgical clinical trial, 6 patients<br />

with localized bladder cancer were treated with two doses <strong>of</strong> Ipilimumab at<br />

10 mg/kg at weeks 1 and 4. Blood was collected pre-therapy and<br />

post-therapy (3 weeks after each dose). CD4 T cells were enriched from<br />

peripheral blood by using the CD4 T cell isolation kit from Miltenyi Biotec<br />

(Auburn, CA). Total RNA was isolated from purified CD4 T cells using<br />

Qiagen RNeasy kit for Affymetrix microarray analyses. Microarray data were<br />

then analyzed using Ingenuity iReport (Redwood City, CA). RT-PCR, RPPA<br />

and Western blot were used to confirm significant changes in genes or<br />

pathways identified in microarray analyses. Results: Ipilimumab treatment<br />

resulted in modulation <strong>of</strong> differentially expressed genes (DEGs). After dose<br />

#1, Ipilimumab only modulated 16 DEGs �2-fold (p�0.05) in CD4 T cells.<br />

After two doses <strong>of</strong> treatment, Ipilimumab significantly changed expression<br />

<strong>of</strong> a total <strong>of</strong> 100 DEGs. Further pathway analyses indicated that Ipilimumab<br />

induced a variety <strong>of</strong> pathways involved in cell cycle control, cell proliferation,<br />

apoptosis, and immune modulation. Specifically, these pathways<br />

include PI3K/AKT, MAP/ERK, IFN/JAK-STAT, granzyme, and protein<br />

ubiquination. Conclusions: Ipilimumab treatment results in modulation <strong>of</strong><br />

multiple genes and pathways, which likely play important roles in antitumor<br />

immune responses and need to be considered for future optimization<br />

<strong>of</strong> anti-CTLA-4 therapy and design <strong>of</strong> combination immunotherapy strategies.<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!