SBIR-STTR Award

Development of an inhibitor of VEGF expression
Award last edited on: 7/23/07

Sponsored Program
SBIR
Awarding Agency
NIH : NCI
Total Award Amount
$1,647,757
Award Phase
2
Solicitation Topic Code
-----

Principal Investigator
Joseph M Colacino

Company Information

PTC Therapeutics Inc

100 Corporate Court
South Plainfield, NJ 07080
   (908) 222-7000
   info@ptcbio.com
   www.ptcbio.com
Location: Single
Congr. District: 06
County: Middlesx

Phase I

Contract Number: 1R43CA108330-01
Start Date: 00/00/00    Completed: 00/00/00
Phase I year
2004
Phase I Amount
$188,172
Up-regulation of vascular endothelial growth factor (VEGF), a key factor for angiogenesis, is an important contributor to the pathogenesis of cancers, diabetic retinopathy, and exudative macular degeneration. The abundance of VEGF is in large part controlled at the post-transcriptional level by sequences in both the 5'- and 3'-untranslated regions (UTRs) of its mRNA. The 5'-UTR contains an internal ribosomal entry site (IRES) that mediates a unique, cap-independent mode of translation initiation. Under hypoxic conditions, cap-dependent translation is dramatically impaired and the translation of the VEGF mRNA occurs through its cap-independent IRES. Thus, even under severe hypoxic conditions, cells are capable of producing large amounts of VEGF resulting in angiogenesis to support further tumor growth or aberrant neovascularization as occurs in ocular diseases. In addition, the 3'-UTR harbors multiple AU-rich stability determinants that have been previously shown to regulate VEGF mRNA turnover rates. Using PTC's proprietary technology platforms, we have successfully identified a collection of compounds that inhibit the expression of VEGF post transcriptionally. The initial results from medicinal chemistry efforts are very encouraging, since all three lead series identified contain compounds with low nanomolar activity for the inhibition of VEGF expression. Preliminary selectivity studies demonstrated that there is a subset of compounds that specifically inhibit VEGF production, while a second set of compounds inhibits VEGF expression as well as other angiogenesis factors such as FGF-2 expression. Based on these results, the aims of this proposal are to further characterize these compounds identified from HTS and preliminary SAR studies in a battery of selectivity assays and cytotoxicity assays; initiate structure-activity studies around the lead compounds to enhance potency, efficacy and selectivity; and develop end-point assays to measure the therapeutic efficacies of compounds for future studies in animals.

Thesaurus Terms:
angiogenesis inhibitor, chemical structure function, drug screening /evaluation, pharmacokinetics, vascular endothelial growth factor biomarker, fibroblast growth factor, hypoxia, messenger RNA, neoplasm /cancer blood supply, neoplastic growth, nucleic acid sequence, posttranscriptional RNA processing, retinopathy of prematurity, ribosomal RNA athymic mouse, cell line, enzyme linked immunosorbent assay, genetically modified animal, human tissue, immunocytochemistry, laboratory mouse

Phase II

Contract Number: 2R44CA108330-02
Start Date: 00/00/00    Completed: 00/00/00
Phase II year
2005
(last award dollars: 2006)
Phase II Amount
$1,459,585

Up-regulation of vascular endothelial growth factor (VEGF), a key factor for angiogenesis, is an important contributor to the pathogenesis of cancer. The abundance of VEGF is in large part controlled at the posttranscriptional levels by sequences in both the 5'- and 3'-untranslated regions (UTRs) of its mRNA. The 5'- UTR contains an internal ribosomal entry site (IRES) that mediates a unique, cap-independent mode of translation initiation. Under hypoxic conditions, cap-dependent translation is dramatically impaired and the translation of the VEGF mRNA occurs through its cap-independent IRES. Thus, even under severe hypoxic conditions, cells are capable of producing large amounts of VEGF resulting in angiogenesis to support further tumor growth. Using PTC's proprietary technology platforms, we have successfully identified a compound that selectively and potently inhibits the expression of VEGF post transcriptionally. In the Phase I of this grant, we proposed to (1) characterize the UTR-selectivity of these compounds, (2) perform medicinal chemistry lead optimization to improve potency, selectivity, and pharmaceutical properties, and (3) establish a pharmacodynamic assay to measure in vivo efficacy. We exceeded these goals, and have identified a compound that is potent, seletive, and orally active in vivo. Here, in Phase II, we propose the following: (1) Conduct efficacy studies in a number of tumor models to establish the spectra of tumor types, optimal dosing regimens, and combination therapies utilizing this agent. Efficacy and combination studies will focus on the following five indications: soft tissue fibrosarcom; glioblastoma; hormone sensitive breast cancer; gastric cancer; and ovarian cancer. (2) Conduct ADME studies and toxicology of the lead compound to enable and design the clinical trials. These studies will include both non-GLP as well as IND-enabling GLP studies. (3) CMC, (4) Further elucidate the mechanism of action, and (5) Preparationion and submission of IND to initiate Phase I trials establishing safety and proof-of-concept in humans