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.

610s Pediatric Oncology<br />

9516 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Relationship <strong>of</strong> divergent ancestral genetic variation on chromosome 6p22<br />

and racial disparities in survival in neuroblastoma. Presenting Author:<br />

Navin R. Pinto, The University <strong>of</strong> Chicago, Chicago, IL<br />

Background: An increased prevalence <strong>of</strong> high-risk disease and worse<br />

outcome are observed in children with neuroblastoma who self-report as<br />

black versus white. We sought to determine whether genetic variation<br />

would explain this racial disparity. Methods: After quality control, we<br />

analyzed 511,836 germline genetic variants in 2,709 ethnically diverse<br />

children with neuroblastoma enrolled on Children’s Oncology Group study<br />

ANBL00B1 from 2001 to 2009. Genetic variation was summarized by<br />

conducting principal components analysis. The first principal component<br />

(PC1) separated patients with African ancestry from all others. PC1 was<br />

used as a continuous variable for ordinal regression with risk group and a<br />

Cox proportional hazard model <strong>of</strong> EFS. To identify genetic mechanisms for<br />

the observed disparities, we developed a method using genome-wide<br />

variation data applied to high-risk versus non-high risk samples. We<br />

identified a comprehensive list <strong>of</strong> loci with significant divergence between<br />

the ancestral populations. We then tested each such locus for association<br />

with high-risk phenotype using logistic regression with the proportion <strong>of</strong><br />

African ancestry estimated by ADMIXTURE as covariate. Finally, top SNPs<br />

were added to multivariate models <strong>of</strong> both risk and event-free survival to<br />

determine if any top associations could abrogate observed disparities.<br />

Results: PC1 was associated with both risk (p � 0.007) and EFS (p �<br />

0.037). We identified 72 population-divergent SNPs nominally associated<br />

with high-risk disease (p � 0.001). The risk allele for one <strong>of</strong> the top SNPs:<br />

rs9295536 (p � 9.2x10-8 ) was more common in the African ancestral<br />

population, was associated with high-risk phenotype and poor outcome in<br />

all patients and validated in a Caucasian-only subanalysis. In multivariate<br />

testing, this SNP abrogated the PC1 association with EFS (p � 0.18).<br />

Conclusions: A SNP with high divergence between ancestral populations on<br />

chromosome 6p22 accounts for the observed racial disparity in survival and<br />

is also a common genetic variant associated with survival in patients<br />

derived from either European or African ancestry (Bonferroni adjusted p �<br />

0.05). Studies to elucidate the function <strong>of</strong> this SNP are underway.<br />

9518 <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

Use <strong>of</strong> whole genome sequencing to identify novel mutations in distinct<br />

subgroups <strong>of</strong> medulloblastoma. Presenting Author: Giles W. Robinson, St.<br />

Jude Children’s Research Hospital, Memphis, TN<br />

Background: Medulloblastoma is a malignant childhood brain tumor comprising<br />

four discrete subgroups (SHH-subgroup, WNT-subgroup, subgroup-3<br />

and subgroup-4). The genetic alterations that drive these subgroups<br />

and that might serve as treatment targets are largely unknown. Methods: We<br />

sequenced entire genomes <strong>of</strong> 37 tumors and matched normal blood. 136<br />

somatically mutated genes identified in this discovery cohort were sequenced<br />

in an additional 56 medulloblastomas. All tumors were classified<br />

into the 4 subgroups by expression pr<strong>of</strong>iling and immunohistochemistry. All<br />

mutations were validated by custom capture, 454, or Sanger sequencing.<br />

Results: Recurrent mutations were detected in 49 genes: 41 are not yet<br />

implicated in medulloblastoma. Several target distinct components <strong>of</strong> the<br />

epigenetic machinery in different disease subgroups, e.g., regulators <strong>of</strong><br />

H3K27 and H3K4 trimethylation in subgroup-3 and 4 (e.g., KDM6A and<br />

ZMYM3), and CTNNB1-associated chromatin remodellers in WNTsubgroup<br />

tumors (e.g., SMARCA4 and CREBBP). Modelling <strong>of</strong> mutations in<br />

mouse lower rhombic lip progenitors that generate WNT-subgroup tumours,<br />

identified genes that maintain this cell lineage (DDX3X) as well as mutated<br />

genes that initiate (CDH1) or cooperate (PIK3CA) in tumourigenesis.<br />

Conclusions: We have identified several new recurrent somatic mutations<br />

that are enriched in specific subgroups <strong>of</strong> medulloblastoma. Alterations<br />

affecting subgroup-3 and 4 tumors appear to disrupt chromatin marking,<br />

most notably H3K27me3 , potentially preserving a stem cell-like state in<br />

tumor cells. Mutations in WNT subgroup tumors affect binding partners <strong>of</strong><br />

CTNNB1 that regulate WNT-response gene transcription. These data<br />

provide important new insights into the pathogenesis <strong>of</strong> medulloblastoma<br />

subgroups and highlight targets for therapeutic development.<br />

9517 Oral Abstract Session, Mon, 8:00 AM-11:00 AM<br />

Phase II study <strong>of</strong> temozolomide in combination with topotecan (TOTEM) in<br />

relapsed or refractory neuroblastoma and other pediatric solid malignancies:<br />

A European ITCC study. Presenting Author: Angela Di Giannatale,<br />

Institute Gustave Roussy, Villejuif, France<br />

Background: Temozolomide and topotecan have shown activity in several<br />

pediatric cancers, including neuroblastoma. Resistance to alkylating agents<br />

due to MGMT expression, MMR deficiency or microsatellite instability may<br />

be overcome through the combination with topoisomerase I inhibitors. The<br />

combination <strong>of</strong> temozolomide and topotecan (TOTEM) was well tolerated<br />

and showed preliminary activity in children with neuroblastoma and glioma<br />

(Rubie et al, 2010). Methods: This multicenter, non-randomized, multicohort<br />

Phase II study included children with neuroblastoma according to a<br />

2-stage Simon design, and patients with central nervous system (CNS) and<br />

extra-cranial solid tumors in a descriptive design. Temozolomide was<br />

administered orally at 150 mg/m2 followed by topotecan at 0.75 mg/m2<br />

intravenously for 5 consecutive days every 28 days. The main endpoint was<br />

objective response (OR), i.e., Complete or <strong>Part</strong>ial Response (CR�PR),<br />

evaluated after 2 cycles according to WHO criteria, or INRC criteria for<br />

neuroblastoma patients with mIBG-positive lesions, by an independent<br />

radiological review. Independent review <strong>of</strong> mIBG imaging is pending.<br />

Results: 103 patients, median age 9.4 years (range 1-21), were treated<br />

between June 2009 and May 2011 in 18 centers: 38 neuroblastoma, 33<br />

CNS tumors and 32 other solid tumors. Overall 420 cycles were administered<br />

(median 3 per patient; range 1-12). Grade 3 or 4 neutropenia was<br />

frequent (55% courses), though only 6% <strong>of</strong> patients developed febrile<br />

neutropenia. In the neuroblastoma cohort, 1 CR and 7 PR were observed,<br />

leading to an estimated OR rate <strong>of</strong> 21% (95%CI, 10-37%). Additionally 22<br />

patients had disease stabilization (SD), leading to an overall tumor control<br />

(CR�PR�SD) <strong>of</strong> 79% (63-90%), and a 12-month progression-free survival<br />

rate <strong>of</strong> 47% (31-64%). Overall, 17/102 evaluable patients achieved<br />

an OR (17%, 10-25%), with 1 CR and 3 PR in 9 medulloblastoma (44%,<br />

14-79%), 2 PR in 4 PNET, 1 PR in 12 malignant glioma, and 2 PR in 9<br />

RMS. Conclusions: Temozolomide-topotecan combination results in significant<br />

tumor control in children with neuroblastoma and medulloblastoma/<br />

PNET with favorable toxicity pr<strong>of</strong>ile.<br />

9519 <strong>Clinical</strong> Science Symposium, Mon, 11:30 AM-1:00 PM<br />

A phase I/II study <strong>of</strong> LDE225, a smoothened (Smo) antagonist, in pediatric<br />

patients with recurrent medulloblastoma (MB) or other solid tumors.<br />

Presenting Author: Birgit Geoerger, Institut Gustave Roussy, Villejuif,<br />

France<br />

Background: Hedgehog (Hh) signaling is crucial in the development and<br />

homeostasis <strong>of</strong> many human organs and tissues. Aberrant Hh signaling is<br />

involved in tumorigenesis through promotion <strong>of</strong> cell proliferation, survival,<br />

and differentiation in wide range <strong>of</strong> human cancers, including approximately<br />

30% <strong>of</strong> MBs. LDE225 is a potent and selective inhibitor <strong>of</strong> Smo, a<br />

key positive regulator <strong>of</strong> Hh signaling. The phase I is exploring the safety<br />

and pharmacokinetics <strong>of</strong> LDE225 in pediatric patients with advanced solid<br />

tumors that are potentially dependent on Hh signaling. Preliminary data<br />

from the ongoing phase I are presented. Methods: Dose-escalation was<br />

performed according to a Bayesian design starting at 372 mg/m2 <strong>of</strong><br />

continuous once daily oral LDE225. Safety and preliminary efficacy <strong>of</strong><br />

LDE225 at the maximum tolerated dose will be assessed in pediatric and<br />

adult patients with recurrent MB in a phase II expansion part. Pharmacokinetic<br />

pr<strong>of</strong>iles were performed at Day 1 and 21. Tumor samples were<br />

analyzed for Hh pathway activation status using a 5-gene Hh signature<br />

assay. Results: Thirty-three patients (24 MB, 3 rhabdomyoscarcoma [RMS],<br />

3 osteosarcoma, and 1 each <strong>of</strong> neuroblastoma, gliomatosis and oligoastrocytoma)<br />

with a median age <strong>of</strong> 13 years (range, 4–17 y) have enrolled.<br />

Dose-limiting toxicity <strong>of</strong> Grade 4 creatine phosphokinase elevation occurred<br />

in 1 RMS patient out <strong>of</strong> 7 patients treated at 372 mg/m2 .No<br />

dose-limiting toxicity was observed at 233 and 425 mg /m2 . LDE225 at<br />

233 and 372 mg/m2 was absorbed with a median Tmax <strong>of</strong> 2 h (range, 1–24<br />

h). Systemic exposures were comparable with adults. Two MB patients<br />

achieved a confirmed complete response (CR) at doses <strong>of</strong> 372 and 425<br />

mg/m2 . Analysis <strong>of</strong> 14 available MB tumor samples using the 5-gene Hh<br />

signature assay showed that the 2 CR patients have Hh-activated tumor.<br />

The remaining 12 tumor samples were from patients who did not achieve<br />

response and were determined to be Hh pathway non-activated. Conclusions:<br />

LDE225 is well tolerated in pediatric patients with advanced malignancies.<br />

Preliminary data show promising efficacy in medulloblastoma patients and<br />

support the use <strong>of</strong> the 5-gene Hh signature assay as a pre-selection tool in<br />

future trials.<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!