Spanning stem cell therapy, endogenous repair, gene therapy and the creation of new technology, Medicine by Design’s Cycle 2 (2019-2022) research program leverages the advantages of large‐scale, multi‐institution funding and combines advances in new technology with important biological questions to achieve translatable outcomes.

Medicine by Design is investing nearly $21 million in 12 Cycle 2 projects.

Investigating how age-related clonal hematopoiesis drives HSC stemness properties and how this leads to inflammatory diseases of the heart

heart icon

Lead Investigator:

John Dick
University Health Network

Co-Investigators:

Sagi Abelson, Philip Awadalla, Gary Bader, Phyllis Billia, Steven Chan, Slava Epelman, Mathieu Lupien

Project Overview: Age-related clonal hematopoiesis (ARCH) is a common feature of aging where blood stem cells acquire genetic mutations that cause more proliferation than is typical, leading their progeny to become overrepresented in bulk blood cells. ARCH has been observed in hematopoietic bone marrow stem cells and is highly correlated with the development of inflammatory conditions associated with aging such as cardiovascular disease (CVD).

This project aims to understand the cellular and molecular factors that drive ARCH. Findings will be used to develop a new class of blood-based biomarkers to better monitor CVD progression, which may be used to improve selection of recipients who are most suited for cell-based therapy, and to develop ways to prevent ARCH-mediated hematopoietic stem cell expansion as a potential approach to prevent CVD.

Development of novel cell and tissue therapies to treat liver failure

liver icon

Lead Investigator:

Gordon Keller
University Health Network

Co-Investigators:

Gary Bader, Christine Bear, Axel Guenther, Sonya MacParland, Ian McGilvray, Shinichiro Ogawa, Molly Shoichet

Project Overview: The development of an hPSC-derived cell/tissue therapy to treat liver failure depends on the generation of cells and/or engineered tissue that can efficiently engraft and restore liver function. Efforts to develop protocols to promote differentiation and development of hepatocytes have been hindered by the limited repopulation potential of this cell population. A major factor contributing to this limitation is the lack of a detailed understanding of the primary cell type(s) that mediate(s) hepatocyte engraftment.

Led by a world-leading team of researchers with expertise in stem cell and liver biology, tissue engineering, liver transplantation and bioinformatics, this project aims to understand the liver’s unique ability to regenerate in order to develop effective strategies for engineering liver tissue constructs for therapeutic applications targeting liver failure. Molecular profiling of primary liver tissue aims to identify a liver repopulating cell, which will be tested in mouse transplantation studies. Co-transplantation of supportive cells to test their ability to either promote hepatocyte engraftment and repopulation in the liver, or actually regenerate specific cell types in the liver, will be tested. This team will also work on developing an advanced hPSC-derived liver organoid with rudimentary bile ducts that can provide stable liver function following ectopic transplantation.

Targeting mesoderm differentiation bottlenecks for regenerative therapies

enabling technologies icon

Lead Investigator:

Shana Kelley
Leslie Dan Faculty of Pharmacy

Co-Investigators:

Stephane Angers, Ben Blencowe, Alison McGuigan, Jason Moffat, Keith Pardee, Sachdev Sidhu

Project Overview: Stem-cell based therapies have the potential to regenerate and repair diseased or damaged tissues in a variety of conditions, however a major hurdle to advancing these therapies is the high-cost of manufacturing the cell type of interest at the required yield and purity to achieve an efficacious and economically viable treatment.

This project is addressing this challenge by uncovering the roots of cellular heterogeneity during stem cell differentiation by combining genome-wide CRISPR and reporter-based screening approaches with high-throughput microfluidics-based profiling to guide the development of biologic reagents that enable robust on-cell screening platforms for the isolation of novel antibodies, and synthetic biology approaches for engineering therapeutic proteins.

Lungs by Design

lungs icon

Lead Investigator:

Shaf Keshavjee
University Health Network

Co-Investigators:

Cristina Amon, Aimy Bazylak, Marcelo Cypel, Tom Waddell

Project Overview: Lung transplantation remains the primary life-saving intervention for patients with end-stage lung disease. However, due to early graft failure related to primary graft dysfunction and late graft failure from chronic rejection and fibrosis the recipient survival rate at five years post-transplant remains approximately 50%.

This leading team of surgeon-scientists and engineers has developed an ex vivo lung perfusion (EVLP) system which maintains donor lung function and viability for up to 24 hours. They aim to develop new preservation solutions and hardware features to advance the EVLP technology from a short-term assessment tool to a longer-term repair platform that will include targeted hPSC-derived epithelial cell replacement therapy.

Cardiac regeneration using pluripotent stem cells

heart icon

Lead Investigator:

Dr. Michael Laflamme
McEwen Stem Cell Institute, University Health Network

Co-Investigators:

Hai-Ling Cheng, Dr. Slava Epelman, Mark Friedberg, Nilesh Ghugre, Anthony Gramolini, Scott Heximer, Gordon Keller

Project Overview: This team is capitalizing on the world-leading effort in Toronto to “re-muscularize” injured hearts with hPSC-derived cardiac muscle (CM) grafts, transforming the treatment of heart failure from one of disease management to one that is truly restorative. Bringing together researchers from the University Health Network, Sunnybrook Health Systems, SickKids Research Institute, and U of T, this team is poised to initiate a first-in-human clinical trial to test their hPSC-based cardiac cell therapy.

Medicine by Design is ensuring they stay at the forefront of this effort by leveraging funding from the Ted Rogers Centre for Heart Research and BlueRock Therapeutics to further invest in this work, with a focus on overcoming the critical remaining barriers to translation by developing “next-generation” hPSC-derived cardiac muscle cells to improve graft integration, survival, function and non-invasive monitoring. These hPSC-CM grafts will also be evaluated for a new application – to support the right ventricle under high pressure, which is the predominant heart failure mechanism in pulmonary hypertension and congenital heart disease, a major pediatric indication.

Stem cell-based approaches to endogenous repair of brain and skeletal muscle

brain icon

Lead Investigator:

Freda Miller
The Hospital for Sick Children

Co-Investigators:

Gary Bader, Penney Gilbert, Sid Goyal, David Kaplan, Yun Li, Alison McGuigan, Cindi Morshead, Peter Zandstra

Project Overview: There is keen interest in the environmental signals within stem cell tissue niches that promote tissue repair in settings where regeneration does not occur naturally. To date, studies to identify these signals have been conducted in an inefficient and non-robust manner, examining one signal at a time, hand-picked from large datasets, in expensive preclinical animal models.

In this project, candidate therapeutics (novel small molecules and/or re-purposed drugs) for endogenous stem cell-mediated repair of brain demyelinating disorders and muscular dystrophy will be identified using revolutionary molecular screening approaches that apply high-throughput single cell transcriptomics and computational modeling to predict the signals that exert responses of interest (e.g. repair-like activity) and then validating these combinations in rapid, experimentally amenable and accurate in vitro models of human brain and muscle stem cell tissue.

Gene therapies to promote neuroregeneration and enhance neuroplasticity

brain icon musculoskeletal icon

Lead Investigator:

Cindi Morshead
University of Toronto

Co-Investigators:

Isabelle Aubert, Maryam Faiz, Carol Schuurmans, Melanie Woodin

Project Overview: Building on combined expertise in cell and molecular biology, genetics, gene therapy, central nervous system regeneration, disease models, and behavior analyses, this team will develop broadly applicable and clinically relevant genetic reprogramming strategies to generate astrocytes and neurons as a treatment for central nervous system injuries and neurodegenerative disorders, for which there are currently no known cures.

Using a chemogenetics neuromodulation approach in pre-clinical stroke and Amyotrophic Lateral Sclerosis models, focused ultrasound will be used to activate designer receptors exclusively activated by designer drugs (DREADDs) in specific neurons to regulate excitation/inhibition activity as a strategy to restore damaged neural networks. The team will also explore cell reprogramming approaches, delivering transcription factors to astrocytes and neurons in their pre-clinical models to track effects on disease progression and neural repair. To support clinical translation efforts, hPSC-derived 3D cerebral organoids will be used to optimize and “humanize” the approach.

Cloaking to allow cellular cures

Lead Investigator:

Andras Nagy
Sinai Health System

Co-Investigators:

Maria Cristina Nostro, Sara Vasconcelos

Project Overview: The immune system remains one of the major barriers to effectively translate the advances in the generation of stem cell-derived tissues to treat/cure degenerative diseases. Successfully hiding cells from functional immune system will eliminate the need for immune suppression of patients of cell therapies. Combining with the safe cell platform strategy will abolish the inherent risk of cloaked cells to form tumours.

This project will develop advanced cellular and molecular approaches to treat or even cure a broad set disease. Our prototype disease is type1 diabetes (T1D). T1D is autoimmune in nature, where the diabetic’s own immune system attacks and kills insulin-producing β-cells. The attempts to replace the lost β-cells with transplanted cells are challenged by the ongoing autoimmunity, as well as by inflammation, stress, cell death, and alloreaction in the case of transplanted cells that are not host-derived. Our immune cloaked and safe cells will give a solution to the challenge and hold promise to treat T1D.

The endothelial-macrophage niche: A novel concept in the regulation of macrophage abundance and phenotype in tissue homeostasis, injury and regeneration

Lead Investigator:

Clint Robbins
University Health Network

Co-Investigators:

Myron Cybulsky, Jason Fish

Project Overview: Endogenous repair of the vasculature in the setting of chronic disease is impaired by the presence of systemic cardiovascular risk factors (e.g. smoking, diabetes). The lack of regeneration of the aorta, the largest blood vessel in the human body, is particularly problematic, with degeneration leading to Abdominal Aortic Aneurysm. Understanding the molecular mechanisms that control “anti-regenerative” vascular micro-environments are needed if new strategies to regenerate the aorta and reverse aneurysm progression are to be developed.

In this project, the role of local communication between endothelial cells (ECs) and macrophages (Mφs) during arterial homeostasis, injury during disease and the healing process will be studied.  Specifically, the role of EC-derived factors in maintaining the EC-Mφ niche in a model of aortic aneurysm formation will be evaluated with a view to identifying mechanisms of cell-cell communication between healthy versus dysfunctional ECs and Mφs. The insights gained from these studies will inform therapeutic strategies to promote a pro-regenerative micro-environment to stimulate repair with relevance to other vascular beds including the heart, lung, pancreas and liver.

Paving the way for endogenous repair with exogenous delivery: Investigating material exchange, synaptic connectivity and paracrine signalling in neuron transplantation

brain icon

Lead Investigator:

Molly Shoichet
University of Toronto

Co-Investigators:

Julie Lefebvre, Derek van der Kooy, Valerie Wallace

Project Overview: The goal of this project is to tailor and improve transplant outcomes from cell-based retinal therapy to achieve photoreceptor preservation, replacement and vision repair. By leveraging innovations in stem cell and transplantation biology, and novel approaches to synaptic connectivity and biomolecule delivery, this team will investigate the therapeutic benefit of cytoplasmic material exchange from transplanted human photoreceptors, develop tools to detect and promote human photoreceptor synaptic connectivity and integration for vision restoration, and deliver exogenous paracrine factors to the retina to enhance host photoreceptor survival.

Pre-clinical models of human neurological diseases

brain icon

Lead Investigator:

Jeff Wrana
Sinai Health System

Co-Investigators:

Liliana Attisano, Ben Blencowe, Sid Goyal, Laurence Pelletier

Project Overview: The establishment of human cerebral organoids (hCOs) from human pluripotent stem cells (hPSCs) has revealed morphogenetic processes that recapitulate unique features of human brain development. Organoids can thus facilitate the dissection of mechanisms underlying tissue dynamics in development and disease and bridge the gap between traditional 2D cell culture and in vivo genetic animal models.

This project will build upon this team’s previous work to characterize vascularized hCOs using a novel microfluidic platform combined with single cell profiling and tissue imaging. The establishment of robust, vascularized hCOs will be leveraged to develop more physiologically relevant pre-clinical human models of neurological diseases and disorders, including the identification of neurotoxicity and drug penetration through the blood-brain barrier. These studies will lead to a deep understanding of the development of human neural systems and their response to injury, provide new pre-clinical tools that will drive more efficient and safer drug development, and illuminate potential therapeutic strategies to treat stroke.

ImmunoEngineering program

Lead Investigator:

J.C. Zúñiga-Pflücker
Sunnybrook Health Sciences Centre

Co-Investigators:

Sarah Crome, Naoto Hirano, Tracy McGaha, Andras Nagy, Cristina Nostro, Sara Vasconcelos

Project Overview: The project will investigate strategies to prevent the immune system from rejecting transplanted tissues derived from stem cells — a key challenge for regenerative medicine. Their team is using as its model stem cell-derived beta cells — pancreatic cells that produce insulin — which are being widely studied as a possible regenerative medicine therapy for diabetes. ​

​The team is investigating two approaches to prevent immune rejection of the transplanted beta cells. The first strategy is to develop tissue-specific regulatory T cells (Tregs) that can dampen the immune system’s response specifically to the transplanted beta cells without inducing systemic immunosuppression. A key part of this strategy includes harnessing tissue-resident innate cells — immune cells such as macrophages that recognize general features of pathogens rather than targeting specific invaders — to promote immune tolerance, beta-cell engraftment, and Treg function. The second approach involves “cloaking” the precursor cells from the immune system to prevent rejection. Cloaking will also be used to develop a single, safe cell line, which will serve as a “universal” source of therapeutic cells, reducing the need for genetically matched donor tissue.​