SBIR-STTR Award

Novel Inhibitor of Pde4 for the Treatment of Opioid Use Disorder
Award last edited on: 8/27/2021

Sponsored Program
SBIR
Awarding Agency
NIH : NIDA
Total Award Amount
$349,867
Award Phase
1
Solicitation Topic Code
-----

Principal Investigator
Mai Ngoc-Anh Tran

Company Information

Fannin Partners LLC

3900 Essex Lane Suite 575
Houston, TX 77027
   (713) 966-5844
   innovate@fannininnovation.com
   www.fannininnovation.com
Location: Single
Congr. District: 07
County: Harris

Phase I

Contract Number: 1R43DA053124-01
Start Date: 9/30/2020    Completed: 8/31/2021
Phase I year
2020
Phase I Amount
$349,867
Currently, opioid-use disorders are mitigated by replacing illicit opioids with prescription opioids, like methadone and buprenorphine. While this may work for some patients, few patients successfully reach opioid-abstinence. Thus, new agents targeting opioid-use disorders via non- opioid approaches are overdue. One-way opioids induce such tolerance and dependence is through a secondary messenger, cAMP. This signaling molecule reinforces the use of opioids on a biochemical level. This pathway can be altered by an increase of cAMP, by preventing the breakdown of cAMP by phosphodiesterases, PDE. Neuroinflammation is also attenuated by the inhibition of PDE and increased cAMP. On a biological level, chronic opioid-use causes neuroinflammation. Morphine causes immune cells in the brain to become pro-inflammatory, while elevated cAMP induces an anti-inflammatory response. Previous phosphodiesterase-4 inhibitors, PDE4 inhibitors, have shown promise in treating substance abuse disorders, like opioid- use disorders, by reducing this neuroinflammation. However, current inhibitors inhibit multiple PDE4s, including the PDE4D subtype that induces significant nausea and vomiting, limiting these inhibitorsÂ’ therapeutic utility. Inhibition of the PDE4B subtype non-toxically interferes with the classic feedback loop in substance use disorders. Thus, this proposal aims to advance a lead compound from a selective PDE4B series. The lead compound has shown significant selectivity for PDE4B over PDE4D. When tested in vitro, the series showed potent anti-inflammatory activity. Additionally, the lead compound has a satisfactory pharmacokinetic profile with decent brain penetration, half-life, bioavailability, etc. When tested against other neurological targets, the lead compound had slight activity against 2 receptors, which may mitigate substance abuse disorders and reduce nausea and vomiting. When tested in a self-administration substance abuse model, the lead compound potently reduced the drug seeking behavior, like prior PDE4 inhibitors. This proposal will focus on confirming lack of toxicity and efficacy. For aim 1, the lead will be tested for off-target liabilities, which may be early indicators of toxicities. In aim 2, toxicity within animal models will be assessed, and induction of nausea and vomiting to the leading competitor. Finally, aim 3 will test efficacy in animal models of opioid self-administration and recurrence. The ultimate goal of this proposal will advance a compound with superior therapeutic use for the treatment of opioid-use disorders.

Public Health Relevance Statement:
Project Narrative Treatment of opioid-use disorders with pharmaceuticals centers around prescribing other opioid agonists. An alternative strategy is to inhibit phosphodiesterase-4B (PDE4B), an enzyme that activates a central mechanism of addiction and induces neuroinflammation. PDE4B inhibitors, however, are non-selective and also inhibit family member PDE4D, resulting in nausea and vomiting, which limits these inhibitorsÂ’ therapeutic use. We propose to develop a promising PDE4B inhibitor that is substantially more specific to PDE4B than any other inhibitor in clinical development or on the market, offering a unique combination of increased potency and reduced gastrointestinal toxicity for the treatment of opioid-use disorders.

Project Terms:
Abstinence; activating transcription factor; addiction; adherence rate; Affect; androgenic; Animal Model; Anti-Inflammatory Agents; Attenuated; Binding; Bioavailable; Biochemical; Biological; Biological Availability; Brain; Brain region; Buprenorphine; Categories; Cell Nucleus; Cells; Cessation of life; Chemicals; Chronic; Clinic; Clinical; clinical candidate; clinical development; craving; CREB1 gene; Cyclic AMP; Cyclic AMP-Dependent Protein Kinases; Cyclic GMP; Data; Dependence; Development; Disease; Distress; Dopamine Receptor; dosage; Dose; Dose-Limiting; Drug Kinetics; drug seeking behavior; effective therapy; Effectiveness; efficacy testing; Enzymes; Exhibits; Family member; Feedback; gastrointestinal; Genes; Genetic Transcription; Goals; Gold; Half-Life; high throughput screening; Illicit Drugs; illicit opioid; Immune; In Vitro; in vitro Assay; in vitro testing; in vivo; in vivo Model; Inflammatory; Inflammatory Response; inhibitor/antagonist; Lead; lead series; Link; Maximum Tolerated Dose; Methadone; Modeling; Morphine; Naloxone; Narcan; Nausea; Nausea and Vomiting; neurobehavioral; neuroinflammation; Neurologic; New Agents; non-opioid analgesic; novel; opiate tolerance; Opioid; Opioid agonist; opioid overdose; opioid use; opioid use disorder; opioid withdrawal; Oral; Outcome; Overdose; Pathway interactions; Patients; PDE4B; Penetration; Pharmaceutical Preparations; Pharmacologic Substance; Pharmacology; Phase; Phosphodiesterase Inhibitors; phosphodiesterase IV; phosphoric diester hydrolase; Plasma; pre-clinical; preclinical development; prescription opioid; prevent; Property; Rattus; receptor; Recurrence; Relapse; Rolipram; Safety; scale up; Self Administration; Series; Shrews; Signal Transduction; Signaling Molecule; Small Business Innovation Research Grant; Specificity; Substance abuse problem; Substance Use Disorder; Synaptic Transmission; targeted agent; Testing; Therapeutic; Therapeutic Uses; Toxic effect; Toxicology; Vomiting; Withdrawal; Work

Phase II

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