Drug toxicity often goes undetected until clinical trials, the most costly and dangerous phase of drug development. Both the cultures of human cells and animal studies have limitations that cannot be overcome by incremental improvements in the drug testing protocols. A new generation of preclinical models - in form of integrated human tissue platforms, would be transformative to drug screening and predictive modeling of disease. This multiphase (UH2/UH3) proposal integrates in a novel and synergistic way some recent breakthroughs from four leading laboratories, towards developing a vascular/liver/cardiac microtissue system predictive of integrated human physiology. In the UH2 phase, we aim to establish iPS-based vascular, liver and cardiac microtissues providing (i) hierarchical tissue-specific architectures, (ii) functional representation of human biology of health, injury and disease, (iii) real-time biological readouts, and (iv) compatibility with high-throughput/high-content multi-tissue platforms for studies of drug toxicity and over long periods of time (e4 weeks). The work will be done using iPS-derived cells (to provide a large diversity of normal and disease genotypes) and using nondestructive monitoring of the tissue architecture and function (to provide real-time insights into the progression of cell and tissue responses). Our overall hypothesis is that tissue specific structure-function relationship can be achieved through control and biomimetic application of microenvironemental cues in vitro. Such microtissues will then be high-fidelity models for drug testing and toxicology. In the UH3 phase we aim to deploy an integrated cardiac/hepatic/vascular platform and demonstrate its utility for a broad range of genotypes and drugs. Towards these goals, we will pursue a set of coordinated aims with constant feedback, evaluation, and monitoring of the milestones.
Aim 1 is to develop cell-type specific labeling and sensing systems for on-line assessment of tissue architecture and cell function.
Aim 2 is to develop a perfusable branching vascular network serving as a model of the vascular bed and for assembling vascularized liver and cardiac tissues.
Aim 3 is to develop a liver module by assembling hepatic microtissues in hydrogel around the vascular tree.
Aim 4 is to develop a cardiac module by assembling matured cardiac microtissues in hydrogel around the vascular tree.
Aim 5 is to conduct studies of disease susceptibility, for the individual tissue modules and in the multi-tissue platform.
Aim 6 is to investigate human physiology and disease in multi-tissue platforms. This way, we aim to develop a radically new technology for studies of drugs in human tissue models. If successful, the proposed approach would radically enhance the translation of drug discovery into human applications, by enabling accurate preclinical data for better, faster and cheaper clinical trials.

Public Health Relevance

Broader Impact We propose to engineer three different microtissues highly resembling the structure and normal function of human capillary network, liver lobule and heart muscle, and to use these microtissues for screening of drugs and study of disease under conditions representative of whole bodyphysiology. To enable personalized approach to evaluation of drug regimens and study of disease, we will use adult stem cells derived from small samples of the patient's skin. This technology could greatly accelerate translation of discovery into new therapeutic modalities for the patients in need.

Agency
National Institute of Health (NIH)
Institute
National Institute of Biomedical Imaging and Bioengineering (NIBIB)
Type
Exploratory/Developmental Cooperative Agreement Phase II (UH3)
Project #
3UH3EB017103-05S2
Application #
9266151
Study Section
Special Emphasis Panel (ZRG1 (50)R)
Program Officer
Hunziker, Rosemarie
Project Start
2012-09-30
Project End
2017-06-30
Budget Start
2016-08-15
Budget End
2017-06-30
Support Year
5
Fiscal Year
2016
Total Cost
$158,240
Indirect Cost
$58,240
Name
Columbia University (N.Y.)
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
621889815
City
New York
State
NY
Country
United States
Zip Code
10032
Ronaldson-Bouchard, Kacey; Ma, Stephen P; Yeager, Keith et al. (2018) Advanced maturation of human cardiac tissue grown from pluripotent stem cells. Nature 556:239-243
Marturano-Kruik, Alessandro; Nava, Michele Maria; Yeager, Keith et al. (2018) Human bone perivascular niche-on-a-chip for studying metastatic colonization. Proc Natl Acad Sci U S A 115:1256-1261
Chen, Timothy; Vunjak-Novakovic, Gordana (2018) In vitro Models of Ischemia-Reperfusion Injury. Regen Eng Transl Med 4:142-153
Liu, Bohao; Lee, Benjamin W; Nakanishi, Koki et al. (2018) Cardiac recovery via extended cell-free delivery of extracellular vesicles secreted by cardiomyocytes derived from induced pluripotent stem cells. Nat Biomed Eng 2:293-303
Genet, Nafiisha; Bhatt, Neha; Bourdieu, Antonin et al. (2018) Multifaceted Roles of Connexin 43 in Stem Cell Niches. Curr Stem Cell Rep 4:1-12
Ronaldson-Bouchard, Kacey; Vunjak-Novakovic, Gordana (2018) Organs-on-a-Chip: A Fast Track for Engineered Human Tissues in Drug Development. Cell Stem Cell 22:310-324
Marturano-Kruik, A; Villasante, A; Yaeger, K et al. (2018) Biomechanical regulation of drug sensitivity in an engineered model of human tumor. Biomaterials 150:150-161
Polacheck, William J; Kutys, Matthew L; Yang, Jinling et al. (2017) A non-canonical Notch complex regulates adherens junctions and vascular barrier function. Nature 552:258-262
Villasante, A; Sakaguchi, K; Kim, J et al. (2017) Vascularized Tissue-Engineered Model for Studying Drug Resistance in Neuroblastoma. Theranostics 7:4099-4117
Eyckmans, Jeroen; Chen, Christopher S (2017) 3D culture models of tissues under tension. J Cell Sci 130:63-70

Showing the most recent 10 out of 61 publications