Our recently completed Phase 1 animal studies demonstrated the ability of our novel second generation partial (p)MHC construct, DR?1-hMOG-35-55, to treat acute and chronic EAE with potency comparable to the parent RTL1000 molecule (pDR2-MOG-35-55) in matched DR2-Tg mice and in DR2 negative mismatched mice. Having met our Phase 1 milestones, our Phase II application will produce a fully humanized DR?1-hMOG-35-55 molecule and assemble preclinical data necessary to support a pre-IND meeting with FDA, at which critical input will be obtained in order to enable the filing of a Phase 1 safety and tolerability study in humans. These data will include characterization of the pharmacokinetics (PK), toxicokinetics (TK) and downstream effects of potential anti-drug antibody (ADA) production. Additionally, this study will assess as an in vivo biomarker the intrinsic activity of DR?1-hMOG-35-55 to inhibit peripheral blood mononuclear cell (PBMC) expression of CD74, the major cellular receptor for macrophage migration inhibitory factor (MIF) that has been implicated in MS disease progression both in the literature and in samples from MS subjects. Our proposed studies are patterned after the preclinical development plan that enabled RTL1000 human clinical trials and are designed to stage DR?1- hMOG-35-55 for cGMP manufacture and formal preclinical studies necessary for a Phase 1 clinical trial in human subjects. The clinical protocol to be supported by these preclinical studies will generally follow that used for RTL1000 (Double-Blind, Placebo-Controlled, Phase 1, Dose-Escalation Study of the Safety of a Single Dose of RTL1000 in Subjects with Relapsing Remitting or Secondary Progressive Multiple Sclerosis) which demonstrated that RTL1000 was safe and well tolerated at doses <60mg (1,000g/kg). Doses of DR?1-hMOG- 35-55 to be infused in the human Phase 1 trial will be guided by pre-IND discussions with FDA and will depend on the minimal effective dose (MED) as well as the maximum tolerated dose (MTD) to be determined for both sexes in this proposal. Data from our Phase 1 STTR has established that a higher dose of DR?1-hMOG-35-55 is required for inhibiting T cell activation in vitro and for treatment of chronic EAE in females vs. males, thus necessitating establishment of separate MEDs and overlapping efficacy curves to yield a common dose range. It is anticipated that these studies will facilitate filing of a successful IND for initiating therapeutic trials with DR?1- hMOG-35-55 in subjects with MS.

Public Health Relevance

This project will obtain preclinical data on DR?1-MOG-35-55, a novel MHC class II construct that has the potential to treat multiple sclerosis (MS). The aims will optimize the production, evaluate the toxicity and pharmacokinetics and determine the proper dosing of DR?1-MOG-35-55. Due to its universal expression, DR?1-MOG-35-55 would be a ?match? for all human recipients and would thus nicely complement RTL1000 and address the unmet need of treating HLA-DR2 negative patients.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
Small Business Technology Transfer (STTR) Grants - Phase II (R42)
Project #
5R42AI122574-03
Application #
9529493
Study Section
Special Emphasis Panel (ZRG1)
Program Officer
Minnicozzi, Michael
Project Start
2016-02-01
Project End
2019-06-30
Budget Start
2018-07-01
Budget End
2019-06-30
Support Year
3
Fiscal Year
2018
Total Cost
Indirect Cost
Name
Virogenomics Biodevelopment, Inc.
Department
Type
DUNS #
079809660
City
Tigard
State
OR
Country
United States
Zip Code
97223
Meza-Romero, Roberto; Benedek, Gil; Gerstner, Grant et al. (2018) Increased CD74 binding and EAE treatment efficacy of a modified DR?1 molecular construct. Metab Brain Dis :
Yang, Liu; Liu, Zhijia; Ren, Honglei et al. (2017) DR?1-MOG-35-55 treatment reduces lesion volumes and improves neurological deficits after traumatic brain injury. Metab Brain Dis 32:1395-1402
Benedek, Gil; Meza-Romero, Roberto; Jordan, Kelley et al. (2017) MIF and D-DT are potential disease severity modifiers in male MS subjects. Proc Natl Acad Sci U S A 114:E8421-E8429
Meza-Romero, Roberto; Benedek, Gil; Jordan, Kelley et al. (2016) Modeling of both shared and distinct interactions between MIF and its homologue D-DT with their common receptor CD74. Cytokine 88:62-70