SBIR-STTR Award

Small Molecule MYC Degraders as Novel Cancer Therapeutic Agents
Award last edited on: 2/4/2024

Sponsored Program
SBIR
Awarding Agency
NIH : NCI
Total Award Amount
$1,339,826
Award Phase
2
Solicitation Topic Code
395
Principal Investigator
Dennis Liang Fei

Company Information

StemSynergy Therapeutics Inc

1951 Northwest 7th Avenue Suite 300
Miami, FL 33136
   (305) 753-0217
   capobianco@stemsynergy.com
   www.stemsynergy.com
Location: Single
Congr. District: 24
County: Miami-Dade

Phase I

Contract Number: 1R43CA268456-01A1
Start Date: 4/5/2022    Completed: 3/31/2023
Phase I year
2022
Phase I Amount
$399,999
The MYC family proteins are comprised of three paralogs termed Myc (c-myc), N-myc, and L-myc. The MYCproteins play a fundamental role in cell proliferation and oncogenesis by regulating cellular processes such asgene transcription, protein translation, cell cycle progression, and cell death. High levels of N-myc protein (genename: MYCN) are often found in tumors of neuroendocrine origins, where it has been shown to drive tumorgrowth. Amplification of the MYCN locus occurs in approximately 50% of high-risk neuroblastoma, which is themost common extracranial solid malignancy of childhood. N-myc protein levels are highly regulated by Aurorakinase A: N-myc binds to Aurora kinase A to "escape" proteasomal degradation. The tool small molecule Aurorakinase A inhibitor, CD532, effectively dissociates N-myc from Aurora kinase A, resulting in N-myc proteindestabilization and regression of MYCN-amplified neuroblastomas. Although CD532 is an excellent proof-of-concept molecule, this compound has poor solubility, limited permeability, and poor metabolic stability, makingit a poor drug candidate. To overcome these liabilities, we have developed distinct, novel small molecules, thateffectively dissociate N-myc from Aurora A and destabilize N-myc and that are more bioavailable than CD532.For simplicity, these compounds are referred to as "N-myc degraders".The primary goal of our Phase I proposal is to improve the potency, selectivity, drug-like properties, and in vivoefficacy of our lead N-myc degrader, SSTA-152. We propose two specific aims:Specific Aim 1. Increase the potency and selectivity of SSTA-152.Specific Aim 2. Improve drug-like properties and in vivo efficacy of SSTA-152.The overall goal is to develop a clinical N-myc degrader for treating N-myc-driven cancers, which fulfills asignificant unmet need in patients.

Public Health Relevance Statement:
PROJECT NARRATIVE This proposal seeks to develop novel N-myc degraders to treat N-myc-driven cancers such as MYCN-amplified neuroblastomas. Neuroblastoma is the most common extracranial solid malignancy of childhood, and amplification of the MYCN locus is well-established as a biomarker for high-risk chemotherapy-refractory neuroblastoma (30% - 40% long-term survival rate). Our preliminary results demonstrate that our N-myc degraders are effective in inhibiting the growth of MYCN-amplified human neuroblastoma cells.

Project Terms:
<21+ years old><20S Catalytic Proteasome><20S Core Proteasome><20S Proteasome><20S Proteosome>

Phase II

Contract Number: 2R44CA268456-02
Start Date: 4/5/2022    Completed: 8/31/2025
Phase II year
2023
Phase II Amount
$939,827
The MYC family proteins are comprised of three paralogs termed c-myc, N-myc, and L-myc. The MYC proteinsplay a fundamental role in cell proliferation and oncogenesis by regulating cellular processes such as genetranscription, protein translation, cell cycle progression, and cell death. While N-myc and L-myc driveoncogenesis in a small number of cancer types, the requirement of c-myc is widespread in a broad range ofhuman cancers. MYC protein levels are highly regulated by Aurora A: both c-myc and N-myc bind to Aurorakinase A to "escape" proteasomal degradation. With the support of SBIR Phase I, we have successfully identifiednovel small molecules that 1). directly target the MYC:Aurora A binding interface, 2). potently degrade bothendogenous N-myc and c-myc (henceforth "MYC degraders"), 3). are metabolically stable and orallybioavailable, and 4). are efficacious in inhibiting the growth of tumors dependent on either N-myc or c-myc.For the SBIR Phase II period, we plan to advance pre-clinical development of our MYC degraders with anemphasis on treating c-myc-dependent cancers. We propose two specific aims:Specific Aim 1: Test the efficacy of SSTA-315 across c-myc dependent cancers, while in parallel developderivative c-myc degraders with improved potency and efficacy.Specific Aim 2: Evaluate the safety profile of SSTA-315 (or an alternative lead MYC degrader) in IND-enablingGLP toxicity studies.Successful completion of the proposed studies will complete IND-enabling safety studies for our lead MYCdegrader, preparing for the IND registration with the FDA as the immediate next step. Given that MYC proteinsare deregulated in most human cancers, our MYC degraders have potential to impact the lives of millions ofcancer patients in U.S. and represent a significant market opportunity.

Public Health Relevance Statement:
PROJECT NARRATIVE This proposal seeks to continue the pre-clinical development of novel MYC degraders to treat c-myc-dependent non-small cell line cancers (NSCLCs) and colorectal cancers (CRCs). We show that our MYC degraders harbor excellent drug-like properties and are effective in inhibiting the growth of c-myc-dependent NSCLC and CRC in pre-clinical models. Our MYC degraders have potential to impact the lives of millions of cancer patients in U.S. and represent a significant market opportunity.

Project Terms:
<21+ years old><20S Catalytic Proteasome><20S Core Proteasome><20S Proteasome><20S Proteosome>
© Copyright 1983-2024  |  Innovation Development Institute, LLC   |  Swampscott, MA  |  All Rights Reserved.