SBIR-STTR Award

Determination of Optimal Antisense Oligonucleotide Chemistry for Efficient and Safe Splicing Modulation in T Cells
Award last edited on: 1/15/2024

Sponsored Program
STTR
Awarding Agency
NIH : NIAID
Total Award Amount
$283,667
Award Phase
1
Solicitation Topic Code
855
Principal Investigator
Gaddiel Galarza-Munoz

Company Information

Autoimmunity Biologic Solutions Inc

3433 Cove View Boulevard Unit 2504
Galveston, TX 77554
   (787) 420-7613
   N/A
   www.abstherapeutics.com

Research Institution

University of Texas - Galveston

Phase I

Contract Number: 1R41AI149920-01
Start Date: 5/5/2020    Completed: 4/30/2021
Phase I year
2020
Phase I Amount
$283,667
Multiple Sclerosis (MS) is the most common neurological disease of early adulthood and is mediated by autoimmune mechanisms that lead to demyelination and neuronal damage in the central nervous system, resulting in progressive neurological dysfunction. There is no cure for the disease and current treatments focus on preventing future immunological attacks, mainly by suppressing the immune system. This leads to adverse side effects that are often severe or fatal. Accordingly, there is a clear unmet need for the development of effective and well-tolerated therapies to arrest MS development. This has been challenging because MS has numerous etiologies and the molecular mechanisms underlying these etiologies are not well understood. We uncovered the molecular underpinnings of an MS etiology and hope this knowledge will translate into a targeted therapy for MS. This specific etiology results from up-regulation of the soluble form of the Interleukin- 7 Receptor (sIL7R), which has been shown to aggravate the progression and severity of the disease in the Experimental Autoimmune Encephalomyelitis (EAE) mouse model of MS, and to be elevated in patients of several autoimmune diseases including MS, Type I diabetes, Rheumatoid arthritis and Systemic lupus erythematosus. Given that sIL7R is produced by abnormal exclusion of exon 6 from IL7R pre-mRNAs, we developed a novel biologic drug, a splicing-modulating antisense oligonucleotide (SM-ASO; IL7R-005) that corrects this abnormal splicing and restores normal expression of IL7R protein isoforms. IL7R-005 represents a major improvement over current MS therapies in that by correcting IL7R splicing, it diminishes expression of the pathogenic sIL7R isoform, without affecting the function of the membrane-bound IL7R (mIL7R), which is vital for proper immune function, thereby avoiding the adverse immunosuppressive effects of current drugs. T cells are the major producers of sIL7R in humans, and thus to reduce sIL7R levels, IL7R-005 needs to be delivered into T cells in vivo. Although SM-ASOs have been shown to modulate RNA splicing decisions in many tissues in vivo (e.g. FDA-approved Spinraza), the delivery and functionality of SM-ASOs in T cells have not been thoroughly examined, and represents the major hurdle to expand the use of SM-ASOs for treatment of immunological disorders and the development of novel immunotherapies. Here, we address this obstacle by conducting an in-depth, side-by-side analysis of the influence of diverse chemical modifications on the efficiency of SM-ASOs in primary T cells and their potential toxic effects in relevant cell models. This is critical as the chemical modifications of the SM-ASOs could influence their pharmacological properties (e.g., cellular uptake) differentially across cell-types, and thus define the potency of SM-ASOs in a cell-type specific matter. The chemical modifications of the SM-ASOs also dictate potential harmful effects, such as hepatic or renal toxicities. Therefore, this in-depth analysis will enable selection of the chemistry with the optimal therapeutic index (i.e., high potency, low toxicity) for efficient splicing modulation in T cells.

Public Health Relevance Statement:
PROJECT NARRATIVE Antisense oligonucleotides (ASOs), in particular splicing-modulating ASOs (SM-ASOs), are a promising and powerful tool for the treatment of genetic diseases, and such promise has started to come to fruition by recent FDA-approvals of several ASOs for previously intractable diseases. Albeit their recent success in the clinics, an expanded application for treatment of immunological disorders or generation of novel immunotherapies has been somewhat limited by difficulties with delivery of ASOs into immune cells. The key to overcome this limitation lies in the chemical modifications of the ASOs, and thus this proposal aims to overcome this limitation by determining the optimal ASO chemistry for efficient and safe modulation of splicing in T cells.

Project Terms:
Address; Affect; Animal Model; Antisense Oligonucleotides; Autoimmune Diseases; Autoimmune Process; Award; base; Biological; Biological Assay; Callithrix; Cell model; cell type; Cells; Chemicals; Chemistry; Clinic; Clinical Research; Cognitive; Complex; Cultured Cells; Demyelinations; Development; Diagnosis; Disease; emerging adult; Emotional; Etiology; Exclusion; Exhibits; Exons; Experimental Autoimmune Encephalomyelitis; FDA approved; Fruit; Funding; Future; Generations; Genetic; Genetic Diseases; Goals; Grant; Hepatic; Hepatotoxicity; Human; IL7R gene; Immune; immune function; Immune system; Immune System Diseases; Immunologics; Immunomodulators; Immunosuppression; Immunotherapy; improved; in vivo; in vivo evaluation; Insulin-Dependent Diabetes Mellitus; Interleukin 7 Receptor; Investigation; Kidney; Knowledge; Lead; Measures; Mediating; Medical; Membrane; Modeling; Modification; Molecular; motor impairment; mouse model; mRNA Precursor; Multiple Sclerosis; nephrotoxicity; nervous system disorder; Neuraxis; Neurologic Dysfunctions; neuron loss; Neurons; novel; Pathogenicity; Patients; personalized medicine; Pharmaceutical Preparations; Pharmacology; Phase; preclinical study; prevent; Property; Protein Isoforms; Research; Rheumatoid Arthritis; RNA Splicing; Safety; Sensory; Severity of illness; Side; side effect; Small Business Innovation Research Grant; Small Business Technology Transfer Research; success; Systemic Lupus Erythematosus; T-Lymphocyte; targeted treatment; Testing; Therapeutic; Therapeutic Index; Therapeutic Intervention; Tissues; tool; Toxic effect; Toxicology; Translating; Up-Regulation; uptake; Work

Phase II

Contract Number: ----------
Start Date: 00/00/00    Completed: 00/00/00
Phase II year
----
Phase II Amount
----