This Strategic Alliance seeks funding to develop an immunotherapeutic intervention for prescription opioid abuse that combines a lead oxycodone vaccine with immunomodulators formulated through a novel thermosensitive gelling polymer technology to enhance the post-vaccination antibody (Ab) response. Vaccines for substance use disorders (SUD) have the potential to be a long-lasting, safe, cheap, and effective intervention. Vaccines for SUD stimulate the patient's own immune system to produce anti-drug Ab that prevent drug distribution to the brain and drug-induced behavior. First-generation SUD vaccines did not meet expectations, yet demonstrated proof-of-principle in the subset of immunized subjects that achieved high levels of anti-drug serum Ab. Generation of Ab results from T cell-dependent B cell differentiation in germinal centers (GC) in secondary lymphoid organs. GC-dependent antigen-specific B and T cell differentiation is regulated by specific signaling pathways controlled by cytokines (e.g., interleukins), co- stimulatory molecules, and immune checkpoints. A wealth of pre-clinical studies have tested whether the efficacy of SUD vaccines can be improved by hapten and bioconjugation chemistry, adjuvant, peptides and protein carriers, liposome-protein nanoparticle, or polymer-based nanoparticles. In contrast, no studies have tested the broad range of available immunomodulators, which have revolutionized immunotherapy for cancer and autoimmune diseases. Using the model vaccine 6OXY-KLH, our group has identified immunological mechanisms underlying post-vaccination Ab and vaccine efficacy, and we are currently exploiting these targets for developing more effective SUD vaccines. In a screening study to identify potential lead immunomodulators, we found that co-administration of 6OXY-KLH with IL-4, or a monoclonal Ab (mAb) that blocks IL-2 signaling, increased efficacy against oxycodone. To effectively combine the 6OXY-KLH with IL-4 and/or other immunomodulators in a suitable injectable formulation, we have partnered with i-novion, a start-up company specialized in polymer technology, which has exclusive ownership of novel pentablock co-polymers (PBC).
AIM1 will test whether combination of 6OXY-KLH and selected immunomodulators improves post-vaccination anti-oxycodone Ab, 6OXY-specific B cells, and vaccine efficacy in mice.
AIM2 will test whether PBC-assisted delivery improves efficacy of 6OXY-KLH and leads from AIM1.
AIM3 will test the efficacy of leads from AIM1 and AIM2, in blocking oxycodone-induced striatal dopamine release and conditioned place preference in mice, and oxycodone i.v. self-administration in rats. Completion of these aims will generate an immunotherapeutic intervention for prescription opioid abuse, ready for cGMP production and IND enabling studies. Completion of this project will provide data supporting a novel mechanism-based strategy for enhancing post-vaccination Ab responses, which could be applied to vaccines for other SUD or challenging diseases such as HIV, malaria, or cancer.

Public Health Relevance

~22 million Americans suffer from substance use disorders (SUD), costing the public an excess of $700 billion/yr in crime, lost work productivity, and health care. Among other drugs, the abuse of prescription opiate analgesics has significantly increased over the last decade, and ~2.1 million Americans are dependent on prescription opioids. The prescription opioids oxycodone and hydrocodone also act as gateway drugs to heroin use. Compared to existing medications, therapeutic vaccines for opiates have the potential to provide a low-cost, long-lasting, safe, and effective intervention that lacks CNS-related side effects, abuse liability, and diversion. Due to their selectivity for the targeted opioid and different mechanism of action, vaccines may also be used in combination with methadone, buprenorphine and naltrexone. This Strategic Alliance seeks funding to develop a next-generation vaccine for prescription opioid abuse by a two-pronged approach: 1) combining a lead oxycodone-based immunogen with immunomodulators to enhance the quantity and quality of the post- vaccination anti-drug antibody response, and 2) the best immunotherapeutic cocktail will be formulated in novel gelling polymers to increase vaccine efficacy by focusing delivery of vaccine components to the injection site, prevent systemic effects of interleukins and immunomodulators, and create a depot for sustained release.

Agency
National Institute of Health (NIH)
Institute
National Institute on Drug Abuse (NIDA)
Type
Research Project (R01)
Project #
5R01DA041730-02
Application #
9355154
Study Section
Special Emphasis Panel (ZDA1-JXR-D (09)S)
Program Officer
Rapaka, Rao
Project Start
2016-09-30
Project End
2019-05-31
Budget Start
2017-06-01
Budget End
2018-05-31
Support Year
2
Fiscal Year
2017
Total Cost
$581,016
Indirect Cost
$53,507
Name
Minneapolis Medical Research Fdn, Inc.
Department
Type
Research Institutes
DUNS #
068195064
City
Minneapolis
State
MN
Country
United States
Zip Code
55415
Laudenbach, Megan; Baruffaldi, Federico; Robinson, Christine et al. (2018) Blocking interleukin-4 enhances efficacy of vaccines for treatment of opioid abuse and prevention of opioid overdose. Sci Rep 8:5508
Baruffaldi, Federico; Kelcher, April Huseby; Laudenbach, Megan et al. (2018) Preclinical Efficacy and Characterization of Candidate Vaccines for Treatment of Opioid Use Disorders Using Clinically Viable Carrier Proteins. Mol Pharm 15:4947-4962