Therapeutic and Device Innovations Win Accelerator Grants

October 16, 2023

Eight research teams led by Columbia University faculty, across diverse disciplines, have received a Columbia Life Science Accelerator Pilot Grant for their innovative lab-to-market projects. The winning projects focus on research or inventions that are on a path to commercialization and have real-world applications in the clinic that can impact patient care and diagnosis.

The researchers are addressing a diverse array of problems in domains ranging from heart disease to diagnostics to fertility and more. The teams participated in a rigorous semester-long bootcamp and pitched their innovation to expert judges. Awardees were selected for their novel scientific solution, clinical merit, and potential to translate their research from the lab to commercial market. This year’s teams share a total of $490,000 in pilot funding. 

The annual Life Science Accelerator pilots are co-funded by the Irving Institute for Clinical and Translational Research through its Translational Therapeutics Accelerator (TRx); the Herbert Irving Comprehensive Cancer Center through its Accelerating Cancer Therapeutics (ACT) program; and Columbia Engineering through its Biomedical Engineering Technology Accelerator (BiomedX). The three groups work closely with Columbia Technology Ventures (CTV), a central office at Columbia for technology development initiatives, entrepreneurial activities and industry collaborations, and the Columbia Lab-to-Market Accelerator Network, which serves as a framework to successfully develop, launch and execute initiatives to help commercialize academic research. 

Congratulations to the 2023 Life Science Accelerator Pilot Grant awardees (categorized by funding arm): 

Columbia Biomedical Engineering Technology Accelerator (BiomedX): 

CardiAIc: Artificial intelligence for atrial isthmus prediction 

Dr. Saluja

Deepak Saluja, MD

Lead Investigators:  

  • Deepak Saluja, MD, Associate Professor of Medicine, Division of Cardiology 
  • Christine Hendon, PhD, Associate Professor of Electrical Engineering 

Cardiac arrhythmias are common and can be fatal. Cardiac ablation targeting the critical component or isthmus of an arrhythmia is a commonly used treatment; however, effectively targeting the isthmus is challenging due to functional and anatomic barriers such as scar tissue. The team, led by Drs. Deepak Saluja and Christine Hendon, is applying deep learning to recognize electroanatomic features of target regions for ablation in patients with arrythmias. This technology has the potential to improve ablation outcomes for a substantial number of patients, by accurately identifying where to ablate within the critical circuit. 
 

Vibrant Bio: A comprehensive diagnostic method for all human infectious pathogens from one sample (Co-funded by ACT and TRx)

Dr. Min

Wei Min, PhD

Lead Investigators:  

  • Wei Min, PhD, Professor of Chemistry 
  • Zhilun Zhao, PhD, Postdoctoral Scientist, Department of Chemistry 

Current methods for infectious disease diagnostics are slow, costly, and low throughput, limiting timely treatment for many patients. To tackle the need for rapid, comprehensive detection, Professor Wei Min and team developed an innovative approach to screen for numerous pathogens in one patient sample. This Vibrant MicroBead method utilizes optical barcoding of microbeads, proprietary nucleic acid-based bioassays, and a high-efficiency, cost-effective hardware platform to rapidly read barcodes. This adaptable system can also extend to other applications like serological tests, pathogen screening, and phenotyping. This technology holds promise for early detection of infectious disease, rapid intervention to improve patient outcomes, and prevention of future pandemics. 
 

Rapid point-of-care assays for detection of poliovirus and enterovirus D68 

Dr. Mishra

Nischay Mishra, PhD

Lead Investigators:  

  • Nischay Mishra, PhD, Assistant Professor of Epidemiology 
  • Kiran T. Thakur, MD, Assistant Professor of Neurology 

In recent years, risk of paralytic poliomyelitis and enterovirus D68 (EV-D68) associated acute flaccid myelitis has increased. Early symptoms of both conditions are similar and include fever, headache, congestion, and cough. However, within one week, symptoms can worsen to include neurological sequelae. Clinicians and public health practitioners need rapid, inexpensive point-of-care diagnostic assays with high sensitivity and specificity to identify infected patients who may benefit from therapeutic antibodies and antivirals. Dr. Mishra and team have developed high-density peptide microarrays and immunoreactive peptides for EV-D68 and poliovirus. Using these technologies, the team will build lateral-flow assays for rapid differential diagnosis of early infections with EV-D68 or poliovirus. The lateral-flow assays will be used as point-of-care diagnostics for testing both patients and environmental samples to improve response time for future cases and outbreaks. 


FoliSeq: Stool-based host RNA profiling for gut disease diagnosis and monitoring (Co-funded by TRx)

Dr. Wang

Harris Wang, PhD

Lead Investigators: 

  • Harris Wang, PhD, Associate Professor of Systems Biology 
  • Alejandro Chavez, MD, Associate Professor of Pediatrics, UCSD 

Development of real-time, low-burden, and cheap monitoring methods for gut health, signaling, and inflammatory status can improve the treatment and management of inflammatory bowel disease (IBD). Current methods for clinical management of IBD involve colonoscopies, which are resource intensive and invasive, and stool or blood tests, which only measure a limited number of biomarkers such as calprotectin and lactoferrin. However, these strategies do not provide the necessary sensitivity nor insight into disease pathology required to inform clinical decision making. Professor Harris Wang and team have developed a non-invasive and inexpensive method, FoliSeq, to measure the gastrointestinal transcriptome directly from stool to track disease state and response to therapy over time in large patient cohorts for less than $30 per sample. FoliSeq can be used to profile gut health, level of inflammation, disease pathway activity, and therapeutic response, and holds great potential as a non-invasive method to guide IBD management. 
 

Translational Therapeutics Accelerator (TRx):

Preventing Genetic Abnormalities to Improve ART/IVF 

Dr. Egli

Dieter Egli, PhD

Lead Investigators: 

  • Dieter Egli, PhD, Associate Professor of Developmental Cell Biology​ in Pediatrics and Obstetrics and Gynecology, Columbia Stem Cell Initiative 
  • Zev Williams, MD, PhD, Wendy D. Havens Associate Professor of Women's Health in Obstetrics and Gynecology

 The inefficiency of human embryo development is a major bottleneck for fertility treatments, which use assisted reproductive technology (ART) and in vitro fertilization (IVF) to help people have a biological child. The human embryo has an extraordinarily unstable genome, and abnormal DNA replication and the resulting chromosomal abnormalities are thought to be a primary cause of embryo failure. Drs. Egli and Willliams and team are developing a messenger RNA therapeutic that will introduce genome stabilizing proteins to fertilized eggs to decrease the mutations and chromosomal abnormalities that result in unusable blastocytes. The ultimate goal is to develop a method that increases the efficiency and accessibility of fertility treatments by reducing the costs and the number of eggs needed to achieve a live birth for millions of people worldwide who struggle to conceive. 


Corvyv: Keeping Heart Cells Alive Through a Heart Attack 

Barry Fine, MD, PhD

Barry Fine, MD, PhD

Lead Investigators:

  • Barry Fine, MD, PhD, Assistant Professor of Medicine
  • Donald Landry, MD, PhD, Hamilton Southworth Professor of Medicine

During a heart attack, blood flow to the heart is interrupted leading to irreversible cell death and loss of cardiac muscle. Approximately 12 percent of patients in the United States who have a heart attack will die, and survivors face significant complications including heart failure and arrhythmias. Current therapies focus on mitigating the damage to cardiac muscle by reestablishing blood flow, but there is no therapy that improves muscle survival. To address this gap, Drs. Fine and Landry have developed an approach to treat heart cells with the small molecule APS-062 to prevent cardiac cell death and preserve heart muscle during a heart attack. APS-062 is a small molecule inhibitor of STK25, a kinase that the team has shown to regulate cardiac cell survival. This novel approach to pharmacological inhibition of STK25 has the potential to prevent cardiomyocyte death and improve heart attack outcomes for patients.


Cell Therapy for Lung Diseases

Hans-Willem Snoeck, MD, PhD

Hans-Willem Snoeck, MD, PhD

Lead Investigators:

  • Hans-Willem Snoeck, MD, PhD, Byron M. Thomashow Professor of Medicine in Microbiology and Immunology
  • N. Valerio Dorrello, MD, PhD, Associate Professor of Pediatrics

For some lung diseases, including lethal neonatal surfactant deficiency and idiopathic pulmonary fibrosis (IPF), lung transplantation is the only curative treatment available. Yet lung transplantation is costly, is hampered by donor organ shortages, and has a 5-year survival rate of only 60 percent. The team, led by Drs. Snoeck and Dorrello, is designing a one-time cell replacement therapy (CRT) for these conditions that would repair damaged lungs and correct genetic defects. Their approach uses distal lung epithelial progenitors (DLEPs) generated from induced pluripotent stem cells (iPSCs) or embryonic stem cells (ESCs) coupled to conditioning of the recipient lung using de-epithelialization and local irradiation. This approach will address some of the key challenges of cell therapy, including generation of suitable engrafting cells and creating a receptive environment in the recipient. CRT could provide a one-time, life-saving treatment for patients for whom lung transplantation is currently the only curative option that is more universally available and offers a higher probability of long-term survival, and it could potentially be expanded to other lung diseases.


ProteoClear: Combatting Neurodegenerative Disease 

Ai Yamamoto, PhD

Ai Yamamoto, PhD

Lead Investigators: 

  • Ai Yamamoto, PhD, Associate Professor of Neurology and Pathology and Cell Biology 
  • Katherine Croce, PhD, Postdoctoral Research Fellow, Department of Neurology 

Many incurable adult onset neurodegenerative diseases, such as Huntington’s Disease and Parkinson’s Disease, are characterized by abnormal accumulation of protein aggregates in the brain, which results in debilitating symptoms. Drs. Yamamoto and Croce are developing an innovative approach to help cells find and then clear the protein accumulation. They will introduce lncRNA to increase expression of Alfy, a cellular label that directs cells to clear protein aggregates via autophagy—the body’s process of reusing damaged cell parts. The team will initially focus on Huntington’s Disease, a rare, heritable disease that results in motor and cognitive impairment and renders patients unable to care for themselves, with the goal of slowing disease progression and increasing quality of life for patients and families. This technology also has the potential to expand to combat more prevalent disorders such as Parkinson’s Disease and Alzheimer’s Disease.