NIH HEAL Initiative Expertise and Resources

Contact: NCATSDPIHEALCollab@nih.gov

NCATS is offering opportunities to apply our state-of-the-art technologies and extensive experience in therapeutic development to ideas and expertise in pain, addiction and overdose through collaboration. The opportunities for collaboration fall into two general categories: novel human cell-based screening platforms, and pharmacological probe and preclinical drug development (described below).

Novel Human Cell-Based Testing Platforms

Traditionally, discovery of small molecule probes and drugs has begun with screens in cell-free systems or cell lines with heterologously expressed genes, often of non-human origin. While these platforms have value, they may identify compounds that do not reflect native human cellular physiology or disease, which have a high likelihood of failure at later stages of the translational process. The advent of induced pluripotent stem cell (iPSC) and 3-dimensional tissue printing technologies offers the opportunity to develop screening platforms that more accurately reflect human disease physiology and will allow the discovery of more robust and ultimately successful probe compounds and drugs for pain, addiction and overdose.

iPSC-Derived Cell-Based Model Development

Computed 3-D Tumor Sphere

Investigators with a desire to develop and characterize iPSC-derived cell types relevant to nociception (e.g., primary and associative pain pathways), addiction (e.g., reward pathways), and overdose should apply. Proposals in this space should focus on development, in-depth characterization and rigorous utilization of human iPSC-based assays related to modeling and reversing pain, addiction, and opioid overdose. The NCATS Stem Cell Translation Laboratory has specialized expertise in development of robust, reproducible and scalable automated iPSC differentiation protocols and comprehensive cell characterization. Among the lab’s capabilities are:

  • Advanced imaging technologies (e.g., high-content confocal, calcium imaging, optogenetics) and data analysis for functional cell characterization, including longitudinal tracking of cell behaviors with multiple measurements over days, weeks or months;
  • High-throughput electrophysiology methods (e.g., high-density multi-electrode arrays [26,400 electrodes/well]) to streamline monitoring of electrical activity at high spatiotemporal resolution;
  • Measurement of cell signaling pathways, metabolism and specific targets (e.g., cyclic AMP, PKA activity, CREB phosphorylation, energy metabolism);
  • Combined single-cell transcriptomic and proteomic analyses that provide information on drug response in individual cells; and
  • Access to large numbers of iPSC-derived sensory neurons (nociceptors), neuronal subtypes (e.g., GABAergic, glutamatergic, dopaminergic), and astrocytes in chemically defined conditions already extant in the lab.

Development of 3-Dimensional Biofabricated Tissue Models

Investigators with a desire to develop multicellular constructs that mimic the structure and function of tissues involved in pain (e.g., DRG, PAG, thalamus), addiction (e.g., ventral tegmentum, nucleus accumbens), or overdose (e.g., medulla) using relevant human primary or iPSC-derived cells should apply. The expertise of the NCATS 3-D Tissue Bioprinting Laboratory is the biofabrication of architecturally and physiologically accurate normal and disease-relevant tissue models in multi-well plate format, to increase the throughput of drug testing and to create models that are better predictors of human response to new drugs. This laboratory integrates advances in tissue engineering technologies, 3-D bioprinters, biocompatible polymers and hydrogels to validate the morphology and physiology of the biofabricated human tissues. The laboratory is also developing disease-relevant assay readouts in 3-D tissues that are compatible with the screening capabilities available at NCATS. Specifically, the 3-D Tissue Bioprinting Laboratory offers:

  • Collaborative biofabrication of 3-D tissues in multi-well plate format for drug screening;
  • Architectural, physiological and pharmacological validation of biofabricated 3-D tissue models;
  • Development of high-throughput quantitative endpoint measurements on 3-D tissue models;
  • Screening of drugs using the biofabricated 3-D tissue-in-a-dish relevant models, including stem cell-derived organoids;
  • Development and testing of compounds in 3-D blood-brain barrier models; and
  • Joint development of engineering and 3-D tissue technology platforms.

Pharmacological Probe and Preclinical Drug Development

The first step in qualifying a novel molecular target as potentially useful in therapeutic applications is the creation of a small molecule “probe” compound that can test the therapeutic hypothesis in cell-based or animal model systems. If those studies are promising, then development of a “drug” compound can begin, involving the multi-step recursive process to impart the properties required for submission of an Investigational New Drug (IND) application to the Food and Drug Administration (FDA) or other regulatory agency.

Pharmacological Probe Development

Investigators who have identified potential pain, addiction or overdose targets can collaborate with this program to generate an optimized probe that will enable the testing of a therapeutic hypothesis. The laboratory encompasses assay development; quantitative high-throughput screening (qHTS) to identify promising compounds to modulate novel targets; and optimization by medicinal chemists to optimize potency, selectivity and pharmacokinetic properties required of an in vitro/in vivo pharmacological probe of the novel target. Assays of target activity adaptable for HTS, secondary assays to guide medicinal chemistry optimization and biological validation, and animal efficacy models, when applicable, should be available in the applicant’s laboratory. Probes can also be tested using the iPSC or 3-D bioprinting platforms described above. Sharing probes with the scientific community will be a priority. Capabilities include:

  • Adaptation/miniaturization of assays for HTS and new assay development
  • Large diversity screening chemical libraries and specialty libraries for mechanistic dissection and drug repurposing
  • Counterscreening/confirmatory assays, including for common artifactual activities
  • Chemical informatics
  • Medicinal chemistry
  • In vitro ADMET and in vivo DMPK characterization
  • HEAL target and compound library on 1,536-well and 384-well plate formats

Preclinical Drug Development

Investigators/companies/nonprofit research institutions that have identified promising lead small molecule compounds or prototype biologic, gene or cell therapies for indications in pain, addiction or overdose can form joint project teams with the NCATS Therapeutic Development Branch (TDB) to develop IND-ready therapies for consideration by the FDA for clinical testing. Capabilities available through the TDB include:

  • Lead-to-clinical-candidate medicinal chemistry optimization
  • Repurposing of approved therapies
  • Formulation for optimal bioavailability
  • In vitro ADMET and in vivo DMPK characterization
  • Toxicological characterization, including in vitro and non-GMP and GMP toxicity studies
  • CMC studies
  • GMP scale-up and manufacturing of drug agent and drug product for clinical testing
  • Project management