Pivotal Experiment Fund2023-09-14T16:17:34-04:00
Banner Building a Biotech Venture

The Pivotal Experiment Fund (PEF) is a strategic and competitive program intended to advance translation of Medicine by Design’s team projects portfolio. The PEF aims to build a robust, preclinical pipeline of regenerative medicine-based therapies, enabling technologies and ventures that have strong potential for clinical, socio-economic and/or commercial impact. Medicine by Design will invest up to $3 million in the PEF.

On This Page

Round 1 Funded Projects

Guiding stem cell therapy with bright-contrast MRI

Lead Investigator:

Hai-Ling Margaret Cheng
Institute of Biomedical Engineering, University of Toronto

Co-Investigators:

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

Dr. Michael G. Fehlings
Krembil Research Institute, University Health Network

Stem cell therapy offers immense promise in promoting the repair of diseased or injured tissue and is currently used to treat patients with blood disorders. Several other organs, such as the heart and central nervous system(CNS), are at the threshold of clinical trials. However, broader clinical acceptance of stem cell therapy still faces significant hurdles. Questions related to the immediate success of cell targeting and grafting,the ongoing status of treatment, an improved understanding of graft rejection versus host disease, or long-term tumor-forming potential remain difficult to answer without a means to repeatedly see viable stem cells inside the body. Currently, no gold standard exists for in vivo, deep-tissue cellular imaging.

Integration of Ex Vivo Lung Perfusion (EVLP) with hemodialysis to to enable extended donor lung preservation for transplantation

Lead Investigator:

Dr. Shaf Keshavjee
Sprott Department of Surgery, University Health Network

Co-Investigator:

Dr. Mingyao Liu
Toronto General Hospital Research Institute, University Health Network

The demand for life-saving lung transplantation vastly exceeds the current available supply of donor organs. At University Health Network (UHN), we have led the world in transplant innovations that address this critical patient need. This includes our development of a novel technology called Ex Vivo Lung Perfusion (EVLP), where donor lungs are placed in a protective sterile dome and provided oxygen, nutrients and heat, allowing them to fully “breathe” on their own while outside of the body. Transplant surgeons use EVLP to monitor donor lung quality and make sure that only the best lungs are accepted for transplant. Traditionally, up to 80% of available donor lungs have some type of suspected injury and are usually declined for transplant. We can take advantage of the duration of EVLP—currently up to six hours—to apply novel therapies to repair the organ. In fact, at Medicine by Design we are working on many new gene, drug and stem cell therapies that use EVLP to repair more lungs for transplant. However, these advanced therapies need more time on EVLP—around 48 hours—to achieve their full effect. Achieving preservation over this extended period of time is difficult, mainly due to the buildup of excess by-products produced by the breathing lung. Our Medicine by Design –Pivotal Experiment Fund project will explore the integration of dialysis with EVLP, with the goal to use dialysis to safely remove harmful by-products and maintain lung preservation to 48 hours. We have recently achieved safe 36 hour preservation using this technique, suggesting our ambitious goal is now within reach. If successful, we will work closely with our industry partners to provide access to this technology to transplant surgeons everywhere, and make donor lungs available for all end-stage lung disease patients in desperate need.

Cellular reprogramming to promote neural repair in pre-clinical models of neurotrauma and neurodegeneration

Lead Investigator:

Cindi Morshead
Department of Surgery, University of Toronto

Co-Investigator:

Carol Schuurmans
Sunnybrook Research Institute

Maryam Faiz
Department of Surgery, University of Toronto

Collaborator:

JoAnne McLaurin
Sunnybrook Research Institute

Brain disorders are among the most serious health problems facing modern society. As human lifespan has increased, so has the incidence of death and disability due to injury and disease. The increased incidence of neurological disease in our aging population has led to an enormous economic burden and decreased life quality for those affected. Our goal is to develop innovative new strategies to repair the injured or diseased central nervous system (CNS)using a novel strategy to promote neuro repair centered on the generation of new cells to replace those lost to injury or disease. To achieve this goal,we propose to manipulate resident neural cells to change their identity and/or function.Specifically, we have developed a novel gene product that can be delivered directly to the brain and target a population of brain cells called astrocytes. Astrocytes are an attractive cell source to target because of their abundance in the brain, their activation after injury,and their contribution to the pathology of neurodegenerative disease and neuro trauma. Our gene therapy product will ‘reprogram’ or convert astrocytes to neurons, which are thecells that are lost following injury (e.g., neuro trauma,such as stroke) and in neurodegenerative disease (e.g.,Amyotrophic Lateral Sclerosis, ALS; Alzheimer’s Disease, AD). Our pivotal experiments are designed specifically to assess the therapeutic efficacy of our novel gene product sin pre-clinical models of stroke and ALS, for which there are currently no known curative strategies; even small improvements will be paradigm shifting for these intractable conditions. Our outcome measures of success are functional behavioural assays –undoubtedly the most relevant when translating our findings to humans. While we propose to target ALS and stroke here in, we have important proof-of-concept data in an AD model supporting the idea that our approach is broadly applicable to other CNS injuries and neurodegenerative diseases.

Enabling allogenic pre-clinical application of hPSC-derived hepatocytes and liver tissue with immune evasion strategies

Lead Investigator:

Shinichiro Ogawa
McEwen Stem Cell Institute, University Health Network

Co-Investigator:

Gordon Keller
McEwen Stem Cell Institute, University Health Network

Sonya MacParland
Toronto General Hospital Research Institute, University Health Network

Dr. Ian McGilvray
Toronto General Hospital, University Health Network

Hepatocytes are the predominant cell type in liver and are responsible for over 500 functions, including detoxification, protein synthesis and production of chemicals that enable food digestion. Therefore, hepatocyte injury, caused by various etiologies, such as viral infections, metabolic disorders and drug toxicity, will result in either acute or chronic liver failure. Although acute liver failure is relatively uncommon, over 3 million Canadians suffer from chronic liver disease, which will inevitably lead to end-stage liver failure. Currently, the only treatment for end-stage liver disease is whole organ transplantation which is limited by a severe shortage of available donor livers. At least a quarter of the patients on the liver transplant waiting list die annually while waiting for transplantation. Given these statics, new liver regenerative therapies are desperately needed.Human pluripotent stem cell (hPSC) derived hepatocyte transplantation represents an exciting alternative approach to whole organ replacement for restoring liver function. Successful long-term engraftment of hPSC-derived hepatocytes will, however, require strategies to overcome immunological rejection of the graft by the recipient. Gene-editing tools and immune protective material engineering technologies offer the potential to protect implanted cells from an allogeneic immune response. Our team will take advantage of our expertise in stem cell biology, immunology and liver transplantation to focus on developing a safe clinical-grade product to treat the end-stage liver failure.

Round 2 Funded Projects

Targeting mesoderm differentiation bottlenecks for regenerative therapies

Lead Investigator:

Shana Kelley
Formerly Leslie Dan Faculty of Pharmacy, University of Toronto
(Now Northwestern University)

Co-Investigators:

Jason Moffat
Donnelly Centre for Cellular & Biomolecular Research, University of Toronto

Stephane Angers
Donnelly Centre for Cellular & Biomolecular Research, University of Toronto

Acute graft-versus-host disease (aGVHD) is a common complication of allogeneic hematopoietic stem cell transplantation, occurring in 30–50% of patients. One of the largest unmet needs in the field of transplantation is the discovery of targets and drug candidates to overcome graft rejection. Using a proprietary platform that enables high-throughput genomic screening on the basis of phenotypic outputs, we will provide a new pipeline of novel therapeutic targets and have already identified potential chemical scaffolds for drug development. In particular, our effort focused on cytokine secretion has produced a variety of targets that have been validated in vitro and represent promising leads for therapeutic development in the area of endogenous repair. This pivotal experiment fund (PEF) project will validate targets controlling cytokine secretion in vivo and identify lead compounds for therapeutic development. Project deliverables will also produce important datasets for the development of startup companies or IP for license. The product under development in this effort is a new drug for graft rejection discovered using a versatile platform for therapeutic target discovery. Further validation of the platform will provide the foundation for future partnerships and commercialization activity.

Paving the way for endogenous repair with exogenous delivery

Lead Investigator:

Molly Shoichet
Donnelly Centre for Cellular & Biomolecular Research, University of Toronto

Co-Investigators:

Dr. Rob Devenyi
Krembil Research Institute, University Health Network

Valerie Wallace
Krembil Research Institute, University Health Network

The number one cause of blindness in the western world is due to degeneration of the retina at the back of the eye, and specifically loss of photoreceptor cells, which are essential for vision. These cells degenerate in age-related macular degeneration (AMD), which impacts 200 million people worldwide and retinitis pigmentosa (RP), which impacts 2.5 million people. With only one product concept available to 10% of the AMD patients (i.e.,those with wet-AMD), and nothing for the other 90% of dry-AMD patients, and only one product that targets one of the over 100known genetic mutations(less than1.5% of the for RP population), there is a critical need for new treatments. This team proposes a product that will save the photoreceptors from dying thereby rescuing vision. The product builds on our proven track records in local delivery of slow release hydrogels, animal models of blindness, clinical translation and entrepreneurship. In their pivotal experiment, the team will bring two patented technologies together and build on significant in vivo data, to advance the product concept for vision rescue in rodent models of blindness, with the goal of ultimately rescuing vision.

Round 3 Funded Projects

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

Lead Investigator:

Dr. Filio (Phyllis) Billia
Toronto General Hospital Research Institute, University Health Network

Co-Investigators:

John E. Dick
Princess Margaret Cancer Centre, University Health Network

Sagi Abelson
Ontario Institute for Cancer Research

Transplantation is a treatment, not a cure, as median survival is ~10 years post-solid organ transplantation (SOT). The average cost for SOT in the USA as of 2020 is >US$400,000/organ. When being evaluated, patients are not only subjected to extensive testing pre-transplant, but also require intense surveillance post-transplant to: (1) mitigate complications of immunosuppression and (2) increase survival and quality of life. However, developing targeted strategies to improve outcomes in SOT recipients is limited by the constant low grade level of inflammation that is inherently present post-transplant. To date, a lot of effort by pharma has focused on the development of new non-invasive tools to identify rejection in SOT recipients. More critically important is the need to identify patients that require more rigorous surveillance, in contrast to those that don’t; all in an effort to reduce the morbidity associated with the invasiveness and burden of surveillance testing.The acquisition of specific somatic mutations in hematopoietic stem cells leading to clonalexpansion is referred to as clonal hematopoiesis (CH). In the literature, CH has been associated with inflammatory health conditions such as cardiovascular disease, stroke, and death. The association of CH in SOT has not been previously explored. Our preliminary data in heart transplant (HT) recipients is very encouraging showing an association between CH and an increased incidence of complications post- transplant, including death. The results of this study are already transformative as there are no other non-invasive tools able to stratify the risk of adverse outcomes before a transplant is done or in the post-transplant surveillance setting.We propose to develop a diagnostic platform to monitor SOT recipients throughout their time with their transplanted organ. With machine learning, we will develop prognostic algorithms to predict adverse outcomes in SOT recipients with CH and extend our early observations to other organs (including liver, lung, kidney and heart). The pivotal proof-of-concept experiments described in this PEF will be the first steps towards demonstrating the therapeutic potential and technological utility of this product. The increasing number of transplants performed every year and the potential to reduce post-SOT adverse outcomes makes this platform an attractive product for risk stratification in all SOT recipients.

Applications of tolerogenic innate lymphoid cells (ILCs) for cell therapies

Lead Investigator:

Sarah Crome
Toronto General Hospital Research Institute, University Health Network

Co-Investigators:

JC Zúñiga-Pflücker,
Sunnybrook Research Institute

This team is developing a first-in-class immune-cell therapy strategy involving populations of innate lymphocytes that produce IL-10for the treatment of a range of indications through the controlling of unwanted immune responses. They developed methods to isolate and expand IL-10 producing ILC2s in a manner that maintains expression of signature cytokines. A single infusion of expanded human IL-10 producing ILC2s protects from development of graft-versus host disease and transplant rejection in humanized mouse models. IL-10 producing ILC2s directly inhibit CD4+and CD8+ T cells proliferation, differentiation into pro-inflammatory cells and tissue trafficking. In Medicine by Design Pivotal Experiment Fund studies, the team aims to perform pre-clinical studies assessing their potency compared to the industry leading Treg cells, investigate whether they would have applications in combined cell therapies, as well develop strategies to induce their development from hematopoietic stem cells. If successful, this will provide the foundation to a first-in-class cell therapy technology with the potential to significantly impact patient’s lives.

Distribution, elimination and gold standard benchmarking of the multi-motif dendron RNA nanoparticle

Lead Investigator:

Omar F. Khan
Institute of Biomedical Engineering & Department of Immunology, University of Toronto

This PEF supports the development of a new platform technology, the multi-motif dendron RNA delivery molecule. This PEF has three segments: an ADME study and two parallel studies designed to support the development specific products (vaccines and intracellular enzyme replacement therapies). The ADME studyis the core, foundational activity that will support the platform. The first parallel study is to benchmark our prophylactic work where we compare the performance of vaccines made with our delivery molecule to those made with the delivery molecules found in marketed mRNA vaccines.If successful, this platform can be deployed to create vaccines against many different infectious diseases. The total market for mRNA infectious disease vaccines (excluding COVID-19) is $41.4B.The second parallel study is to build an mRNA nanoparticle to treat ornithine transcarbamylase deficiency (OTCD). This therapeutic is benchmarked against RAVICTI, the marketed OTCD treatment drug.If successful, patients will require fewer doses and can enjoy a higher quality of life. The market for OTCD is expected to reach $856Mby 2027.The ADME and vaccine data will support our upcoming industry project where we will use our fully characterized lead molecule to create a next-generation vaccine. Similarly, the ADME and OTCD data will support our negotiations with a second industry partner for a project where we take our well-characterized mRNA therapeutic and further develop it into a product that does not require the cold chain for storage and distribution, thereby filling a major unmet need.

Directed evolution of growth factor mimics for low-cost scaling of regenerative medicine

Lead Investigator:

Keith Pardee
Leslie Dan Faculty of Pharmacy, University of Toronto

Regenerative and cell-based immunotherapies are on track to revolutionize the treatment of many diseases, especially those which have been recalcitrant to treatment such as tissue loss due to disease or injury. While exciting, the emerging field of regenerative medicine faces the critical challenge of how to scale the production of cells therapies so that they can be brought to market at a price that enables broad access. The culture of human cells requires growth factor to promote the healthy expansion of cells and ensure that cells remain in a clinically relevant state (e.g. pluripotent). These growth factors can be incredibly expensive, often over a million dollars a gram, and their natural instability can pose additional expense for biomanufacturing. Here, we propose to use cell-free directed evolution to develop antibodies that mimic the binding of growth factors to cell receptors. This proposal builds on proof-of-concept work by others1, which showed that antibodies could phenocopy growth factors and even provide greater receptor selectivity over natural growth factors. Moreover, antibodies exhibit considerable natural stability, adding to the appeal of this approach. We have been pioneering cell-free technologies for years, including the cell-free expression of complex antibodies that mimic growth factors (Wnt). This positions our group well to develop new antibody mimics for four growth factors that are key to the culture of clinically relevant stem cells and immune cells. In addition to generating potentially high-value growth factor mimics, the project will also establish a cell-free, directed evolution discovery platform that can be reprogrammed. With the platform in-hand, we anticipate the production of other antibodies for cell therapy, high-value reagents for biomanufacturing and, in the longer-term,therapeutics.Conversations with industry have also highlighted a need for antibodies in cell purification and bead-based activation, suggesting there is a deep well of potential application. Seeking to make a meaningful impact on human health, our goal will be to ensure that antibodies generated by this project make it to market, contributing to the health and economy of Canadians.

About the Pivotal Experiment Fund

The PEF will bridge a critical gap within the innovation ecosystem by supporting early-stage regenerative medicine research discoveries to a point where follow-on investment from third parties is attractive. A pivotal experiment is one that enables a “go/no go” decision on the merits of a product concept based on outcomes that drive a value inflection in the development plan. The PEF is a strategic and competitive program intended to advance translation of select Medicine by Design team (Cycle 2) projects.

The PEF will

  • Provide project funding of up to $250,000 over 6 to 12 months. In exceptional cases, consideration may be given to a larger investment, subject to discussions with Medicine by Design during the proposal development process.
  • Support the generation of pivotal proof-of-concept and/or validation data, enabling the first steps towards demonstrating therapeutic potential and/or technological utility.
  • Support projects that are defined by clear, milestone-driven (clear go/no go) decision points over a 6 to 12 months period.
  • Encourage projects that leverage co-investment from ecosystem partners and programs (e.g. Connaught Innovation Award, UHN Innovation Acceleration Fund, CCRM, TIAP/LAB150, industry partners, etc.).

Project activities can include but are not limited to:

  • Generation of preclinical, pivotal in vivo efficacy and/or proof-of-concept data.
  • Demonstration of a technology for a regenerative medicine application.
  • Critical, pivotal experiments to support the early stages of target validation.
  • Drug screening on a validated target.
  • Demonstrating feasibility of cell manufacturing scale-up.

Investment Review Committee

Bharat Srinivasa

Co-Founder and Principal, Amplitude Ventures
Learn more

Glenn Maclean

Director, Technology and Venture Development, CCRM

Jacques Sayegh

CEO and Managing Partner, Samuel Capital Partners
Learn more

Jamie Stiff

Managing Director, Genesys Capital
Learn more

Matthew Mistry

Market and Equity Analyst, CCRM Enterprises Inc.
Learn more

Parimal Nathwani

CEO, Toronto Innovation Acceleration Partners (TIAP)
Learn more

Go to Top