Grant List
Represents Grant table in the DB
GET /v1/grants?page%5Bnumber%5D=2&sort=-program_officials
https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=1&sort=-program_officials", "last": "https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=1392&sort=-program_officials", "next": "https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=3&sort=-program_officials", "prev": "https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=1&sort=-program_officials" }, "data": [ { "type": "Grant", "id": "15623", "attributes": { "award_id": "1R13CA298641-01", "title": "2025 RNA Nanotechnology Gordon Research Conference and Seminar", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "National Cancer Institute (NCI)" ], "program_reference_codes": [], "program_officials": [ { "id": 32121, "first_name": "YALI", "last_name": "FU", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2025-01-01", "end_date": "2025-12-31", "award_amount": 5000, "principal_investigator": { "id": 32122, "first_name": "Dolores", "last_name": "Di Vizio", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 226, "ror": "https://ror.org/05rad4t93", "name": "Gordon Research Conferences", "address": "", "city": "", "state": "RI", "zip": "", "country": "United States", "approved": true }, "abstract": "This R13 application requests partial funding to support the upcoming Gordon Research Conference (GRC) on RNA Nanotechnology, which will be held in Ventura, CA on Jan 5–10, 2025, and the associated Gordon Research Symposium (GRS), to precede it on Jan 4–5. The meeting will be co-chaired by Dolores Di Vizio and Mark Bathe, who served as the co-vice chairs of the third GRC in January 2023, elected in 2019 at the same GRC Meeting. Vice-chairs are Nils Walter and Alissa Weaver. RNA nanotechnology continues to grow as an important field, with active research being carried out to elucidate fundamental principles of, and develop innovative approaches to, the design and construction of RNA assemblies for diagnostic, therapeutic, and structural applications in biotechnology and medicine. The pace of discovery and innovation in RNA nanotechnology has significantly accelerated in the past two years with the success of the mRNA vaccines that helped mitigate the spread of COVID19, which has spurred major research activity in other forms of mRNA vaccines including principally also cancer vaccines. RNA nanotechnology is built on the unique properties of RNA, including its structural versatility, its known biological functions, its ease of production and functionalization, and its programmability, which altogether make it an ideal platform on which to build self-assembling architectures with a variety of functional attributes for biomedical applications. This GRC focuses on linking fundamental studies of RNA sequence, modifications, and structure to self-assembly and packaging to inform translational studies using RNA nanotechnology in therapeutics and diagnostics with in situ characterization. The 2025 meeting will cover a range of topics in nine sessions. Sessions on Structure-based Elucidation of RNA Function, RNA Therapeutics, RNA Modifications, RNA Delivery, RNA Imaging & Characterization \"In Situ\", RNA Immunology, RNA Cargo Selection, Transport, and Processing, RNA-based Diagnostics and Biomarkers, and RNA Synthetic Biology will feature cutting-edge research on the molecular basis of nucleic acid architectures and modifications, design and characterization of RNA for cancer vaccines and therapeutics, and biophysical approaches to study and predict RNA function. Sessions on RNA cargo selection in extracellular vesicles, RNA immunology, and RNA therapeutics will provide perspectives on the discovery, isolation, and application of RNA from extracellular vesicles and learnings from viruses with an emphasis on translational aspects of natural and synthetic RNAs in cancer diagnostics and therapeutics. The 2025 conference will continue to emphasize robust representation of women and minority scientists. The invited speakers and discussion leaders will include a mix of established researchers in the field and outstanding junior-level investigators from around the world. The meeting will be advertised to a diverse audience to include early career and under-represented minority scientists in the RNA nanotechnology and related fields. A Power Hour will feature topics of diversity, inclusion, and opportunity for under-represented scientists.", "keywords": [ "Academia", "Acceleration", "Address", "Architecture", "Area", "Base Pairing", "Bathing", "Biological Assay", "Biological Markers", "Biological Process", "Biology", "Biophysics", "Biotechnology", "COVID-19", "Cancer Diagnostics", "Cancer Vaccines", "Characteristics", "Chemicals", "Chemistry", "Collaborations", "Collection", "Communication", "Communities", "Computational Biology", "Dedications", "Development", "Diagnosis", "Diagnostic", "Disease", "Drug Industry", "Early Diagnosis", "Engineering", "Ensure", "Environment", "Feedback", "Free Energy", "Funding", "Future", "Genetic Transcription", "Genome", "Government", "Growth", "Hour", "Image", "Immunology", "In Situ", "Knowledge", "Learning", "Link", "Malignant Neoplasms", "Medicine", "Messenger RNA", "Modification", "Molecular", "Nanostructures", "Nanotechnology", "Nucleic Acids", "Participant", "Postdoctoral Fellow", "Principal Investigator", "Production", "Productivity", "Property", "Proteins", "RNA", "RNA Sequences", "RNA chemical synthesis", "RNA delivery", "RNA vaccine", "Reporting", "Request for Applications", "Research", "Research Activity", "Research Personnel", "Role", "Schedule", "Scientist", "Screening for cancer", "Structure", "Technology", "Therapeutic", "Time", "Translating", "Underrepresented Minority", "Untranslated RNA", "Vaccines", "Vertebral column", "Virus", "Woman", "Work", "base", "biophysical techniques", "biotypes", "cancer biomarkers", "career", "clinical application", "clinical diagnostics", "design", "design and construction", "drug development", "extracellular", "extracellular vesicles", "fundamental research", "graduate student", "innovation", "interdisciplinary collaboration", "interest", "meetings", "minority scientist", "new growth", "novel", "novel therapeutics", "preclinical trial", "programs", "rational design", "self assembly", "success", "supportive environment", "symposium", "synthetic biology", "therapeutic RNA", "translational applications", "translational approach", "translational potential", "translational study", "undergraduate student" ], "approved": true } }, { "type": "Grant", "id": "15613", "attributes": { "award_id": "1K99ES036168-01A1", "title": "Environmental mixtures, immune function, and vaccine antibody response in children", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "National Institute of Environmental Health Sciences (NIEHS)" ], "program_reference_codes": [], "program_officials": [ { "id": 32109, "first_name": "Ashlinn Ko", "last_name": "Quinn", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2025-02-04", "end_date": "2027-01-31", "award_amount": 126985, "principal_investigator": { "id": 32110, "first_name": "Mike Zhongyu", "last_name": "He", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 625, "ror": "https://ror.org/04a9tmd77", "name": "Icahn School of Medicine at Mount Sinai", "address": "", "city": "", "state": "NY", "zip": "", "country": "United States", "approved": true }, "abstract": "I am an environmental epidemiologist and exposure scientist by training with a primary research interest in the impact of environmental exposures on immune function and vaccine response – a critical public health issue highlighted by COVID-19. The goal of this proposal is to obtain training and acquire the skills necessary to become an independent investigator at the air pollution-immunology-infectious disease nexus. I have assembled a world-class mentoring team with interdisciplinary expertise in immunology, toxicology, biostatistics, data science, exposure science, exposomics, and epidemiology. The proposed training plan includes formal coursework, field work, clinical observations, laboratory rotations, academic meetings and conferences, and a variety of activities designed for leadership and professional development. I am confident that by the end of the K99 phase of this proposal, my existing expertise combined with the training proposed in this project will allow me to establish myself as an independent investigator in a tenure-track faculty position, with the skillset to build my future independent research program integrating exposure science, epidemiology, and infectious diseases. For this proposal, I will leverage the Programming Research in Growth, Environment and Social Stress (PROGRESS) cohort, an ongoing longitudinal pregnancy/birth cohort based in Mexico City to investigate the impact of ambient environmental pollutants on the antibody response to routine vaccinations. Specifically, I will: 1) build a daily ozone model for Mexico City, which will be integrated with our existing models for fine particulate matter, nitrogen dioxide, and temperature that already exists for the area; 2) assess the association between these ambient environmental pollutants with vaccine antibody levels to routine vaccinations; 3) assess the association between the same pollutants and levels of several specific cell types involved in vaccine response; and 4) explore whether the associations between ambient environmental pollutants and vaccine antibody levels are partially or fully mediated through the immune cell population subsets assessed in #3). This research proposal has several innovations, including the generation of new exposure data (the first of its kind in Mexico City), the use of cutting-edge statistical approaches, and its leverage of both extant data and new measurements that addresses a unique research question while maximizing cost effectiveness. If successful, this work will be able to significantly foster interdisciplinary research and advance our understanding of a critical and urgent public health issue.", "keywords": [ "Address", "Adult", "Affect", "Age", "Air Pollution", "Antibodies", "Antibody Repertoire", "Antibody Response", "Applied Skills", "Area", "Attenuated", "Attenuated Vaccines", "Biological", "Biometry", "Birth", "COVID-19", "COVID-19 pandemic", "Cells", "Child", "Child Development", "Childhood", "Cities", "Climate", "Clinical", "Collaborations", "Communicable Diseases", "Complement", "Data", "Data Science", "Development", "Developmental Process", "Disease", "Environment", "Environmental Exposure", "Environmental Impact", "Environmental Pollutants", "Environmental Risk Factor", "Epidemiologist", "Epidemiology", "Epigenetic Process", "Exposure to", "Faculty", "Flow Cytometry", "Fostering", "Future", "Gases", "Generations", "Goals", "Growth", "Health", "Immune", "Immune system", "Immunity", "Immunologics", "Immunology", "Immunophenotyping", "Individual", "Innate Immune Response", "Innate Immune System", "Interdisciplinary Study", "Joints", "Laboratories", "Leadership", "Leukocytes", "Life", "Life Cycle Stages", "Link", "Literature", "Long-Term Effects", "Measurement", "Mediating", "Mentors", "Metabolic", "Mexico", "Modeling", "Modification", "Nitrogen Dioxide", "Ozone", "Participant", "Pathway interactions", "Perinatal", "Peripheral Blood Mononuclear Cell", "Phase", "Population", "Positioning Attribute", "Predictive Factor", "Pregnancy", "Public Health", "Research", "Research Personnel", "Research Proposals", "Resolution", "Risk Factors", "Role", "Rotation", "Sampling", "Science", "Scientist", "Statistical Methods", "Systems Development", "T-Lymphocyte Subsets", "Temperature", "Tissue Differentiation", "Toxicology", "Training", "Vaccination", "Vaccines", "Work", "adaptive immune response", "ambient air pollution", "career", "cell type", "cohort", "cost effectiveness", "design", "early childhood", "exposomics", "fine particles", "future pandemic", "immune function", "in utero", "infancy", "innovation", "insight", "interest", "late life", "meetings", "methylomics", "nanoparticle", "pandemic disease", "peripheral blood", "pollutant", "prenatal", "preservation", "prevent", "programs", "response", "skill acquisition", "skills", "social stress", "symposium", "tenure track", "vaccine failure", "vaccine response", "vaccine-induced antibodies" ], "approved": true } }, { "type": "Grant", "id": "15610", "attributes": { "award_id": "1R03AI185421-01A1", "title": "Optimizing Strongyloides Testing in an At-Risk US Population", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "National Institute of Allergy and Infectious Diseases (NIAID)" ], "program_reference_codes": [], "program_officials": [ { "id": 32106, "first_name": "Deirdre A.", "last_name": "Joy", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2025-02-01", "end_date": "2027-01-31", "award_amount": 77843, "principal_investigator": { "id": 32107, "first_name": "Eva H", "last_name": "Clark", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 566, "ror": "https://ror.org/02pttbw34", "name": "Baylor College of Medicine", "address": "", "city": "", "state": "TX", "zip": "", "country": "United States", "approved": true }, "abstract": "Strongyloidiasis, caused by infection with the parasitic nematode Strongyloides stercoralis, remains an important public health problem in tropical and sub-tropical regions, with an estimated 30-100 million infected persons globally. Because S. stercoralis infection is acquired from contact with contaminated soil, those most at risk include the socioeconomically disadvantaged, those with agricultural occupations, and those living in areas with poor sewage control practices. S. stercoralis larval migration during chronic infection is limited to the gastrointestinal tract and lungs and causes few if any symptoms. However, larvae can disseminate and cause severe disease in immunosuppressed individuals. The importance of identifying S. stercoralis infection before immunosuppressive treatments are started has been highlighted by increased use of corticosteroids during the COVID-19 pandemic and related increases in disseminated strongyloidiasis diagnoses. Because disseminated strongyloidiasis is often diagnosed late, after larvae have caused significant and irreversible end-organ damage, the mortality rate of disseminated strongyloidiasis is near 90%. Regional studies and case reports indicate that strongyloidiasis is commonly diagnosed in tropical and sub- tropical regions of the US. We have found 4-10% seropositivity in a Houston solid organ transplant cohort and, more recently, even higher seropositivity (16.5%) in a central Texas community. Other small studies of US immigrants from low- and middle-income countries have indicated infection rates as high as 46.1%. Screening for strongyloidiasis is suboptimal due to poor knowledge of the disease among US healthcare providers and imperfect performance of available diagnostic tests. The clinical gold standard of strongyloidiasis diagnosis is microscopic evaluation of multiple stool specimens for S. stercoralis larva. Unfortunately, it is challenging for patients to submit even one stool specimen in a timely manner, and US clinical laboratory technicians may not have enough training and expertise to reliably identify larvae. Several serologic tests have been developed, but commercially available tests have imperfect test performance and are less sensitive in immunocompromised individuals who may not be able to mount a reliable humoral immune response. Newer serologic and molecular tests based on recombinant antigens have been developed but are not yet commercially available. We propose a prospective study of adult HHS patients undergoing a panel of parasitologic, serologic, and molecular Strongyloides assays, with the following specific aims: (1) to identify a high-performing strongyloidiasis screening strategy for at-risk individuals living in the US and (2) to evaluate each assay as a test-of-cure for strongyloidiasis. Improvement in Strongyloides testing would improve screening and management practices, thereby allowing for timely treatment and reduction of strongyloidiasis related morbidity.", "keywords": [ "Adrenal Cortex Hormones", "Adult", "Aftercare", "Agar", "Agriculture", "Antigens", "Area", "Biological Assay", "COVID-19 pandemic", "Case Study", "Cessation of life", "Clinical", "Communities", "County", "Data", "Death Rate", "Diagnosis", "Diagnostic", "Diagnostic tests", "Disease", "Enrollment", "Enzyme-Linked Immunosorbent Assay", "Epidemiology", "Evaluation", "Feces", "Functional disorder", "Future", "Gastrointestinal tract structure", "Goals", "HIV", "Head", "Health Care Systems", "Health Personnel", "Health system", "Helminths", "Hematologic Neoplasms", "Hispanic", "Human", "Iatrogenesis", "Immigrant", "Immune response", "Immunocompetent", "Immunocompromised Host", "Immunoglobulin G", "Immunosuppression", "Immunosuppressive Agents", "Immunotherapy", "Individual", "Infection", "Knowledge", "Laboratories", "Laboratory Technicians", "Larva", "Lung", "Lymphocyte", "Measures", "Microscopic", "Microscopy", "Molecular", "Morbidity - disease rate", "Occupations", "Organ", "Organ Transplantation", "Ovum", "Parasites", "Parasitic nematode", "Patients", "Performance", "Persons", "Peruvian", "Population", "Practice Management", "Prospective Studies", "Public Health", "Recombinants", "Reference Standards", "Reporting", "Research", "Resources", "Risk", "Serology", "Serology test", "Serum", "Sewage", "Soil", "Solid", "Specificity", "Specimen", "Strongyloides", "Strongyloides stercoralis", "Strongyloidiasis", "Symptoms", "T-Cell Lymphoma", "Testing", "Texas", "Therapeutic immunosuppression", "Training", "Uninsured", "United States National Institutes of Health", "Virus", "Visit", "chronic infection", "co-infection", "cohort", "cost", "cross reactivity", "culture plates", "disability", "follow-up", "immunosuppressed", "improved", "infection rate", "low and middle-income countries", "metropolitan", "migration", "mortality", "performance tests", "prevent", "reduce symptoms", "response", "safety net", "screening", "seropositive", "socioeconomic disadvantage", "stool sample" ], "approved": true } }, { "type": "Grant", "id": "15596", "attributes": { "award_id": "4R44HD114323-02", "title": "A Translational Research Approach to Healthy Technology Usage in Language-Minority Families with Young Children", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD)" ], "program_reference_codes": [], "program_officials": [ { "id": 32093, "first_name": "Courtney Leigh", "last_name": "Gallen", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2023-09-12", "end_date": "2026-08-31", "award_amount": 1245366, "principal_investigator": { "id": 32094, "first_name": "Yuan", "last_name": "D'Antilio", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 2093, "ror": "", "name": "TRANSCENDENT INTERNATIONAL, LLC", "address": "", "city": "", "state": "NY", "zip": "", "country": "United States", "approved": true }, "abstract": "Technology use among young children in the U.S. has become increasingly prevalent over the past decade, and the transition to online learning during the COVID-19 pandemic has renewed concerns among parents and educators about screen time. Recommendations for choosing high-quality apps and setting boundaries around device use are often conveyed through position statements, white papers, and blog posts, but the information is rarely presented in an engaging, family-friendly manner that can be readily adopted and sustained in young children’s everyday routines. As a matter of health equity, this challenge is felt even more acutely among language-minority households, who additionally face language barriers when attempting to access relevant resources. This project delivers an interactive, bilingual, hybrid virtual-and-physical world approach to address these translational gaps. It presents recommendations from the academic and medical spheres through bilingual storytelling and a suite of interactive, co-play activities in an online virtual world, then transitions the gameplay to caretaker-child interactions in the physical world, empowering users to first practice healthy technology through interactive online games and then implement those rehearsed practices in their day-to-day lives. The effectiveness of the online platform and interactive content will be evaluated through a randomized controlled study. Improvements in knowledge, attitudes, and confidence related to healthy technology use will be assessed among children and their caretakers. The platform will also be evaluated for its ability to facilitate bilingual interactions between child and caretaker that support learning and language development. Overall, our product will serve not only as a means to inform families of best technology use practices, but as a catalyst to broaden and reimagine joint play with digital devices especially among linguistic minority families.", "keywords": [ "6 year old", "Academy", "Acute", "Address", "Adopted", "Advocacy", "Advocate", "Age", "American", "Area", "Articulation", "Attitude", "Awareness", "Behavior", "Behavioral", "Books", "COVID-19 pandemic", "Caregiver support", "Caregivers", "Child", "Child Rearing", "Code", "Communication", "Computer software", "Consumption", "Data", "Decision Making", "Development", "Devices", "E-learning", "Education", "Educational process of instructing", "Effectiveness", "Emotional", "Ensure", "Environment", "Evaluation", "Face", "Family", "Feedback", "Flowers", "Fostering", "Friends", "Generations", "Goals", "Habits", "Health education", "Home environment", "Household", "Hybrids", "Informatics", "Intervention", "Interview", "Joints", "Knowledge", "Language", "Language Development", "Learning", "Libraries", "Linguistics", "Literature", "Measures", "Medical", "Methodology", "Minority", "Modeling", "National Institute of Child Health and Human Development", "Neurocognitive", "Outcome", "Pamphlets", "Paper", "Parent-Child Relations", "Parents", "Participant", "Pediatrics", "Phase", "Play", "Positioning Attribute", "Process", "Proliferating", "Provider", "Questionnaires", "Recommendation", "Research", "Research Priority", "Resources", "Respondent", "Role", "Scheme", "Series", "Small Business Innovation Research Grant", "Social Interaction", "Strategic Planning", "Stream", "Surveys", "Technology", "Testing", "Translating", "Translational Research", "Video Recording", "age group", "bilingualism", "catalyst", "design", "digital", "digital media", "digital technology", "electronic book", "empowerment", "evidence base", "health care disparity", "health equity", "improved", "information gathering", "interest", "media use", "meetings", "prototype", "randomized controlled study", "skills", "social", "tv watching", "underserved community", "usability", "verbal", "virtual", "virtual world", "web platform" ], "approved": true } }, { "type": "Grant", "id": "15577", "attributes": { "award_id": "5F99AG083306-02", "title": "The Impact of SARS-CoV-2 Infection and Vaccination on the Risk of Alzheimer’s Disease and Related Dementias", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "National Institute on Aging (NIA)" ], "program_reference_codes": [], "program_officials": [ { "id": 32092, "first_name": "Tanisha Anne", "last_name": "Jackson", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2023-12-01", "end_date": "2025-11-30", "award_amount": 42800, "principal_investigator": { "id": 31473, "first_name": "Jingxuan", "last_name": "Wang", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 768, "ror": "https://ror.org/043mz5j54", "name": "University of California, San Francisco", "address": "", "city": "", "state": "CA", "zip": "", "country": "United States", "approved": true }, "abstract": "The growing prevalence of Alzheimer’s disease and Alzheimer’s related disorders (ADRD) is a critical public health concern, potentially exacerbated by the COVID-19 pandemic. Infectious diseases may increase ADRD risk by causing neuroinflammation and oxidative damage in the central nervous system, promoting atherosclerosis and endothelial dysfunction, or via other inflammatory, immune-response, and vascular mechanisms. Despite intriguing links between several infectious and neurodegenerative conditions, whether and how infectious diseases influence ADRD risk remains underexplored. This question is urgent especially during the COVID-19 pandemic, where the widespread SARS-CoV-2 infection has heavily impacted global public health. Mounting evidence suggests a significant fraction of those infected with SARS-CoV-2 may experience substantial and long-lasting sequelae, including cognitive decline and neurologic deficits. Many unknowns remain, including long-term outcomes and the role of COVID-19 vaccination and viral variants in modifying the effect of SARS-CoV-2 infection on cognitive decline and ADRD risk. This proposed F99/K00 project seeks to address these gaps with two specific aims using complementary longitudinal datasets and rigorous, advanced epidemiological and statistical methods. Aim 1 (F99 dissertation phase: 2023-2025) will use the previously identified COVID-19 brain magnetic resonance imaging (MRI) signature region to infer the long-term effects of infection on ADRD risk via brain structure changes. The COVID-19 MRI signature region comprises brain regions where cortical thickness and gray-white matter contrast was reduced after SARS- CoV-2 infection (identified in a longitudinal MRI study). The candidate will quantify the association between the COVID-19 MRI signature region and future ADRD risk among UK Biobank participants. Aim 2 (K00 postdoctoral phase: 2025-2029) will evaluate the effects of vaccination status and timing of SARS-CoV-2 infection on cognitive outcomes and ADRD among COVID-19 survivors. The candidate will use data from electronic health records (EHR) and a nationally representative cohort. Methodological innovations include the use of neuroimaging, causal survival analysis, machine learning methods for classification algorithms, as well as multiple data sources (i.e., clinical data from EHR and survey data from cohort studies). This proposal directly responds to the call to study the impact of COVID-19 on risk of ADRD and cognition from the National Alzheimer's Project Act (NAPA). The proposal extends the candidate’s quantitative expertise with advanced training in clinical and biological perspectives on ADRD as well as causal inference methods for aging research. Strong interdisciplinary mentorship teams and outstanding supportive training environments at the University of California, San Francisco (F99) and the Massachusetts General Hospital (K00) provide a foundation for the candidate to fill an important scientific gap on infectious disease and immune-related determinants of cognitive aging and ADRD, including infection with SARS-CoV-2.", "keywords": [ "2019-nCoV", "Address", "Affect", "Aging", "Alzheimer disease prevention", "Alzheimer&apos", "s Disease", "Alzheimer&apos", "s disease diagnosis", "Alzheimer&apos", "s disease related dementia", "Alzheimer&apos", "s disease risk", "Atherosclerosis", "Biological", "Biology", "Blood Vessels", "Brain", "Brain region", "COVID-19", "COVID-19 diagnosis", "COVID-19 impact", "COVID-19 pandemic", "COVID-19 survivors", "COVID-19 vaccination", "California", "Caring", "Central Nervous System", "Clinical", "Clinical Data", "Clinical assessments", "Cognition", "Cognitive", "Cognitive aging", "Cohort Studies", "Communicable Diseases", "Data", "Data Set", "Dementia", "Diagnosis", "Disease", "Early treatment", "Electronic Health Record", "Emotional", "Environment", "Epidemiologic Methods", "Foundations", "Future", "General Hospitals", "Goals", "Health Care", "Health Surveys", "Health and Retirement Study", "Hypoxia", "Immune", "Immune response", "Impaired cognition", "Individual", "Infection", "Inflammatory", "Knowledge", "Link", "Long-Term Effects", "Magnetic Resonance Imaging", "Massachusetts", "Mediating", "Medicare claim", "Memory", "Mentorship", "Methodology", "Methods", "Nerve Degeneration", "Neurologic Deficit", "Outcome", "Parahippocampal Gyrus", "Participant", "Pathology", "Persons", "Phase", "Postdoctoral Fellow", "Prevalence", "Prevention", "Public Health", "Recording of previous events", "Research", "Research Activity", "Research Personnel", "Resource Allocation", "Risk", "Risk Reduction", "Role", "SARS-CoV-2 B.1.617.2", "SARS-CoV-2 infection", "SARS-CoV-2 variant", "San Francisco", "Science", "Shapes", "Smell Perception", "Statistical Methods", "Structure", "Survival Analysis", "Symptoms", "Thick", "Time", "Training", "Training Activity", "Universities", "Vaccination", "Variant", "Viral", "Virus", "Work", "aging population", "biobank", "brain health", "brain magnetic resonance imaging", "career", "classification algorithm", "clinical diagnosis", "cognitive change", "cohort", "dementia risk", "disorder risk", "diverse data", "electronic health data", "endothelial dysfunction", "epidemiology study", "experience", "high risk", "improved", "innovation", "late life", "longitudinal dataset", "machine learning method", "multiple data sources", "neuroimaging", "neuroinflammation", "novel", "older adult", "oxidative damage", "pandemic disease", "post SARS-CoV-2 infection", "post-COVID-19", "skills", "w" ], "approved": true } }, { "type": "Grant", "id": "15564", "attributes": { "award_id": "5R01CA276107-02", "title": "Control of cancer cachexia via stimulation of resolution of inflammation", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "National Cancer Institute (NCI)" ], "program_reference_codes": [], "program_officials": [ { "id": 32090, "first_name": "Marjorie", "last_name": "Perloff", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2024-01-01", "end_date": "2028-12-31", "award_amount": 622526, "principal_investigator": { "id": 31454, "first_name": "Steven David", "last_name": "Freedman", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [ { "id": 31455, "first_name": "Dipak", "last_name": "Panigrahy", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, { "id": 31456, "first_name": "Charles Nicholas", "last_name": "Serhan", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "awardee_organization": { "id": 771, "ror": "https://ror.org/04drvxt59", "name": "Beth Israel Deaconess Medical Center", "address": "", "city": "", "state": "MA", "zip": "", "country": "United States", "approved": true }, "abstract": "While cancer cachexia is a devastating syndrome characterized by progressive muscle wasting, inflammation, and metabolic disruption, the underlying mechanisms remain poorly characterized. There are currently no treatments for cachexia. Cancer cachexia is driven by systemic inflammation, pro-inflammatory cytokines, and apoptosis (“cellular debris”). A paradigm shift is emerging in understanding the resolution of inflammation as an active biochemical process with our discovery of novel specialized pro-resolving mediators (SPMs), such as resolvins and endogenous resolution programs. SPMs stimulate macrophage-mediated clearance of debris, promote tissue/muscle regeneration, and counter-regulate pro-inflammatory cytokines at nanogram doses without immunosuppression. Despite approaches to block systemic inflammation, there are no current “pro- resolving” therapies in cancer or cancer cachexia. Moreover, the impact of cancer cachexia on eicosanoids and the novel pro-resolving lipid mediators, both key endogenous regulators of initiation and resolution of inflammation, are unknown. This proposal builds on our recent finding that stimulating resolution of inflamma- tion prevents tumor growth by counter-regulating pro-inflammatory cytokines/eicosanoids and clearing debris. Therefore, the overarching theme is to elucidate the underlying processes of failed resolution of inflamma- tion that drive cancer cachexia. We will rely on a set of established experimental systems, including genetic and pharmacological manipulation of SPMs and their receptors in animal models and macrophages. We shall test this in a multi-pronged approach. In Specific Aim 1, the mechanisms of dysregulated resolution of in- flammation in cachexia will be investigated. We will profile lipid autacoid mediators, including eicosanoids and SPMs, to test our innovative hypothesis that failed resolution of inflammation is necessary and sufficient to cause cachexia, which induces a rapid eicosanoid storm with dysregulated SPMs that leads to an un- controlled cytokine storm. We will evaluate pro-resolving lipid mediators as interventional targets in ca- chexia. In Specific Aim 2, we will optimize the delivery of SPM mimetics and humanized nano-pro- resolving medicines (NPRMs) carrying SPM cargo to cachectic tissues and determine whether SPMs can prevent chemotherapy-induced cachexia. These studies will complement Specific Aim 3, which seeks to pre- vent cancer cachexia via stimulation of resolution by precision nutrition. Parabiosis studies will determine whether cancer-induced cachexia is caused by an effect on the primary tumor or the metastatic site as most patients with cachexia die from metastases. These studies can offer a new animal system to evaluate cachexia at an early stage. We will connect our preclinical findings to clinical disease phenotypes using a computational framework to understand failed inflammation resolution in cachexia. Since SPMs have proven safe and effec- tive in human inflammatory disorders, these studies shall provide the basis for rapid translation of resolution- directed treatments in humans as a new direction to potentially prevent and/or reverse cancer cachexia.", "keywords": [ "Acceleration", "Adipocytes", "Advanced Malignant Neoplasm", "Animal Model", "Animals", "Anti-Inflammatory Agents", "Apoptosis", "Apoptotic", "Autacoids", "Biochemical Process", "Body Weight decreased", "CD59 Antigen", "Cachexia", "Cancer Cachexia", "Cancer Control", "Cancer Etiology", "Cancer Patient", "Catabolism", "Cell Death", "Cells", "Cessation of life", "Clinical", "Clinical Trials", "Complement", "Deterioration", "Disease", "Dose", "Eicosanoids", "Excision", "Exhibits", "Fatty acid glycerol esters", "Genetic", "Heart", "Homeostasis", "Human", "Human body", "Immunosuppression", "Induction of Apoptosis", "Inflammation", "Inflammation Mediators", "Inflammatory", "Intervention", "Lipids", "Liver", "Lung", "Lymphocyte", "Macrophage", "Malignant Neoplasms", "Mediating", "Mediator", "Medical", "Medicine", "Metabolic", "Metabolic dysfunction", "Micrometastasis", "Modeling", "Molecular", "Multiple Organ Failure", "Mus", "Muscle", "Muscular Atrophy", "Necrosis", "Neoplasm Metastasis", "Nutritional", "Parabiosis", "Patients", "Phagocytosis", "Phase", "Plasma", "Prevention", "Primary Neoplasm", "Process", "Recurrent tumor", "Regulation", "Research Personnel", "Resolution", "Role", "Site", "Skeletal Muscle", "Spleen", "Syndrome", "System", "Testing", "Tissues", "Toxic effect", "Translations", "Wasting Syndrome", "adipokines", "cancer therapy", "chemotherapy", "clinical translation", "computer framework", "control trial", "cytokine", "cytokine release syndrome", "cytotoxic", "disease phenotype", "innovation", "lipid mediator", "mimetics", "muscle regeneration", "nano", "novel", "novel strategies", "pharmacologic", "pre-clinical", "precision nutrition", "prevent", "programs", "protein degradation", "receptor", "repaired", "restoration", "skeletal muscle wasting", "small molecule", "systemic inflammatory response", "tissue regeneration", "tumor", "tumor growth", "tumor progression" ], "approved": true } }, { "type": "Grant", "id": "15535", "attributes": { "award_id": "7R34MH129542-03", "title": "Adaptation of Critical Time Intervention for Young Adults with Mental Health Challenges in the Transition from Homelessness to Housing", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "National Institute of Mental Health (NIMH)" ], "program_reference_codes": [], "program_officials": [ { "id": 32081, "first_name": "Shahrzad", "last_name": "Mavandadi", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2024-09-15", "end_date": "2025-11-30", "award_amount": 146354, "principal_investigator": { "id": 26858, "first_name": "Sarah Carter", "last_name": "Narendorf", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 167, "ror": "https://ror.org/0190ak572", "name": "New York University", "address": "", "city": "", "state": "NY", "zip": "", "country": "United States", "approved": true }, "abstract": "Young adults experiencing homelessness (YAEH) have high prevalence of mental disorders, however, they face unique access barriers to mental health services combined with low recognition of mental health symptoms, leading to low rates of mental health service utilization. The point of transition from homelessness to housing presents a period of opportunity to identify mental health symptoms and connect and engage YAEH into mental health services to reduce symptoms and promote housing stability. The COVID-19 pandemic has heightened mental health symptoms and increased the need for services among YAEH, but it has also resulted in recovery funds to increase housing supports in many communities. There is a critical need to develop interventions that can support YAEH with mental health challenges as they make the transition from homelessness to housing as more supported housing becomes available. Critical Time Intervention (CTI), a structured, time limited case management intervention has demonstrated success with adults with serious mental illness in multiple randomized controlled trials but it has not been tested in YAEH. The goal of this study is to adapt CTI for the context of transition from homelessness to supported housing within the rapid rehousing program, integrating mental health specific content from a young adult treatment model, C4, to develop CTI-YAMH (young adult mental health), and then test the new intervention in a feasibility pilot. Specifically, we aim to: Aim 1: Refine the draft CTI- YAMH intervention (treatment, training and assessment protocols) to ensure the target mechanisms are adequately addressed for stabilizing housing and mental health, utilizing an iterative stakeholder feedback process to finalize the manuals for pilot testing, then Aim 2: Conduct an open trial of the adapted CTI- YAMH intervention to assess the feasibility of randomization procedures, refine outcome measures, assess acceptability, and examine the preliminary signal of impact of the intervention. This innovative study targets a critical point of intervention, the transition from homelessness to housing, for an extremely marginalized group (YAEH), utilizing an innovative adaptation framework, ADAPT-ITT, to systematically adapt CTI in partnership with youth with lived expertise and community providers. The CTI-YAMH intervention aims to support a population with high unmet need for mental health services through a model that can be paired with rapid rehousing, a supportive housing model widely used in communities across the U.S. Results from this R34 study lay the foundation for a fully powered RCT of the CTI-YAMH intervention in a future R01 study.", "keywords": [ "Address", "Administrator", "Adult", "Age", "Anxiety", "COVID-19 pandemic", "Caring", "Case Management", "Case Manager", "Characteristics", "Clinic", "Communities", "Community Health Care", "Continuity of Patient Care", "Coupled", "Development", "Diagnosis", "Effectiveness", "Elements", "Ensure", "Evidence based intervention", "Evidence based practice", "Face", "Feedback", "Focus Groups", "Foundations", "Funding", "Future", "Goals", "Health", "High Prevalence", "Homelessness", "Housing", "Individual", "Institute of Medicine (U.S.)", "Intervention", "Intervention Studies", "Investigational Therapies", "Manuals", "Mental Health", "Mental Health Services", "Mental disorders", "Mentors", "Modeling", "Moods", "National Institute of Mental Health", "Outcome Measure", "Outcomes Research", "Outpatients", "Phase", "Population", "Population Heterogeneity", "Procedures", "Process", "Protocols documentation", "Provider", "Randomized", "Randomized Controlled Trials", "Recommendation", "Recovery", "Recurrence", "Research Priority", "Self Efficacy", "Services", "Signal Transduction", "Social support", "Structure", "Symptoms", "Testing", "Time", "Training", "Trauma", "United States", "Variant", "Work", "Youth", "care coordination", "experience", "family support", "federal policy", "health service use", "housing instability", "improved", "innovation", "marginalized population", "member", "peer support", "person centered", "pilot test", "pilot trial", "preference", "primary outcome", "programs", "reduce symptoms", "service utilization", "severe mental illness", "skill acquisition", "social stigma", "substance use", "success", "supported housing", "therapy design", "therapy development", "working group", "young adult" ], "approved": true } }, { "type": "Grant", "id": "15522", "attributes": { "award_id": "1F30GM153101-01A1", "title": "Targeting Both Hyperinflammation and Pyroptosis Via Nanotherapeutics to Improve Outcomes in Gram-Negative Sepsis", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "National Institute of General Medical Sciences (NIGMS)" ], "program_reference_codes": [], "program_officials": [ { "id": 32062, "first_name": "DONNA M", "last_name": "KRASNEWICH", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2024-07-18", "end_date": "2026-07-17", "award_amount": 53974, "principal_investigator": { "id": 32063, "first_name": "Jennifer M", "last_name": "Messina", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 1863, "ror": "", "name": "UPSTATE MEDICAL UNIVERSITY", "address": "", "city": "", "state": "NY", "zip": "", "country": "United States", "approved": true }, "abstract": "Sepsis is a complex, heterogenous disease that results in a dysregulated immune response. It is associated with hyperinflammation, known as “cytokine storm”, causing multiple organ failure and death, as well as simultaneous compensatory anti-inflammatory responses, leading to immune suppression by abundant anti- inflammatory cytokine secretion and immune cell death. Sepsis survivors may suffer chronic critical illness, known as Persistent Inflammation, Immunosuppression, and Catabolism Syndrome (PIICS), leading to serious complications and later death. In sepsis triggered by Gram negative bacterial infection, lipopolysaccharide (LPS), an endotoxin, promotes uncontrolled inflammation and cytokine storm through TLR4 activation. It can also trigger significant cell-lytic death of immune cells, known as inflammasome-mediated pyroptosis, which promotes further immune response for pathogen clearance. However, excessive pyroptosis during hyperinflammation results in prolonged immunosuppression and increased organ dysfunction, contributing to the development of PIICS. Currently, there are no available therapies to directly target both excessive cytokine production and immune cell pyroptosis in sepsis, a gap we will fill with our novel immune modulating itaconate- loaded telodendrimer nanoparticles (ITA:TD NP). In Luo lab, we have developed a series of bioactive immune modulating TD nanodrugs, which mimic the molecular pattern of LPS (multivalent charge and fatty acid tails). As a result, the optimized TD nanodrug attenuates LPS-induced inflammation via competitive binding with TLR4. Itaconate, a metabolite produced by activated macrophages, is known inhibit immune cell pyroptosis. Unfortunately, the in vivo application of ITA is limited by the unfavorable pharmacokinetic properties and cytotoxicity. Our LPS-antagonizing TD nanodrug can form a nanocarrier for efficient encapsulation of itaconate, thus to synergize the in vivo application and immune modulation. We hypothesize that concurrent inhibition of LPS signals, inflammasome activation, and membrane pore formation will effectively control both early phase hyperinflammation and pyroptosis-mediated later stage PIICS for improved sepsis treatment. Preliminary results indicate a survival benefit with ITA:TD NP treatment in septic mice induced by LPS and effective inhibition of both inflammation and pyroptosis. Thus, further studies to evaluate ITA:TD NP mechanism of action and efficacy to prevent both acute and late death in sepsis are needed. The aims of this study are: 1) investigate ITA:TD NP-mediated inhibition of hyperinflammation in sepsis, and 2) elucidate the protective effect of ITA:TD NP to prevent PIICS-associated morbidity and mortality. Results from these studies will provide insight to the mechanism of action of ITA:TD NP and its therapeutic potential for concurrent treatment of hyperinflammation and prevention of PIICS in sepsis to reduce mortality and fill this critical gap in patient care.", "keywords": [], "approved": true } }, { "type": "Grant", "id": "15470", "attributes": { "award_id": "5R01CA276040-02", "title": "Immunoglobulin Replacement Therapy and Infectious Complications After CD19-Targeted CAR-T-Cell Therapy", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "National Cancer Institute (NCI)" ], "program_reference_codes": [], "program_officials": [ { "id": 32038, "first_name": "Nancy J.", "last_name": "Emenaker", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2023-09-01", "end_date": "2028-08-31", "award_amount": 858502, "principal_investigator": { "id": 27940, "first_name": "Joshua Aiden", "last_name": "Hill", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [ { "id": 27941, "first_name": "MIGUEL-ANGEL", "last_name": "PERALES", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "awardee_organization": { "id": 2062, "ror": "", "name": "FRED HUTCHINSON CANCER CENTER", "address": "", "city": "", "state": "WA", "zip": "", "country": "United States", "approved": true }, "abstract": "Chimeric antigen receptor T cell therapy (CARTx) has transformed treatment for B cell malignancies. However, the effects of CARTx on humoral immunity and infection risk are incompletely understood. The high prevalence of hypogammaglobulinemia in CARTx recipients has driven frequent use of prophylactic immunoglobulin G (IgG) replacement therapy (IGRT) to prevent infections in this patient population. However, limited data exist to support this practice, and shortages, side effects, and cost necessitate careful stewardship of IGRT. Emerging data indicate that pathogen-specific antibodies often persist after CD19-CARTx, potentially contesting the need for IGRT. Well controlled studies are needed to ascertain the clinical utility of IGRT in CARTx recipients. Within this clinical context, other important and connected questions remain about how IGRT affects CAR-T cell function, in addition to the possible costs versus benefits of the effect of IGRT on healthcare resource utilization. This timely and unique proposal will be the first randomized, controlled trial of IGRT use in CARTx recipients and provide critical insights into the potential risks and benefits of IGRT in this patient population. The key objectives of this study are to evaluate whether IGRT in CARTx recipients reduces infection rates compared to placebo, and to understand the impact of IGRT on previously unexplored outcomes such as CAR-T cell expansion, CAR- T cell persistence, CAR-T cell function, and healthcare resource utilization. For the proposed study, we have assembled an interdisciplinary group of physicians and scientists from high-volume CARTx centers who will leverage our expertise in immuno-oncology, infectious diseases, and cancer outcomes research. We propose a randomized trial of IGRT versus placebo in 150 adults with serum total IgG ≤400 mg/dL prior to CD19-CARTx. Participants will be randomized 1:1 to receive IGRT or placebo within 14 days prior to CARTx and at 28-day intervals after CARTx for 4 months. Aim 1 will compare between study arms the incidence rate of infections through 6 months after CD19-CARTx; we will also longitudinally characterize and compare total and pathogen-specific IgG levels and their association with infections. Aim 2 will explore the association of IGRT with healthcare resource utilization, cytokine release syndrome, and CARTx-associated neurotoxicity. Aim 3 will characterize the impact of IGRT on CAR-T cell expansion, persistence, and function. This will be the first randomized controlled study of IGRT after CARTx and will provide foundational data to establish evidence-based estimates of the clinical efficacy and risk-benefit of IGRT in CD19-CARTx recipients. In parallel, this study will explore other potential effects of IGRT on CAR-T cell dynamics and healthcare resource utilization. The data generated by this proposal will provide the groundwork for future studies to refine infection prevention strategies in the growing population of CARTx recipients.", "keywords": [], "approved": true } }, { "type": "Grant", "id": "15424", "attributes": { "award_id": "1SB1AG087755-01", "title": "Ryan® CompanionBot for Assisting Older Adults with Early-Stage Alzheimer's Disease and Dementia", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [ "National Institute on Aging (NIA)" ], "program_reference_codes": [], "program_officials": [ { "id": 32031, "first_name": "DINESH", "last_name": "John", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "start_date": "2024-03-01", "end_date": "2026-05-31", "award_amount": 1424731, "principal_investigator": { "id": 32032, "first_name": "Mohammad", "last_name": "Mahoor", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 2541, "ror": "", "name": "DREAM FACE TECHNOLOGIES, LLC", "address": "", "city": "", "state": "CO", "zip": "", "country": "United States", "approved": true }, "abstract": "The population of Americans age 65 years or older will increase from 58 million in 2021 to 88 million by 2050. By 2050, nearly 14 million older adults are expected to have AD/ADRD. For these individuals, significant care is required, and that care is often provided by family members. A 2022 study estimated that 11 million American family members are providing 16 billion hours of care valued at more than $272 billion. Senior care facilities are another option. The COVID-19 pandemic severely affected senior care facility residents. Despite representing only about 1% of the total population in the U.S., COVID-19 deaths in senior care facilities have made up nearly 40% of total COVID-19 deaths. Senior care facilities often face staffing shortages during and after the pandemic. Currently, 3 in 5 assisted living facilities are concerned that they may have to close due to staffing shortages. The situation with these two caregiving options is alarming; increasing demand for caregivers coupled with short supply has led to higher costs, unfilled needs, and fierce competition for resources. While computer technologies, such as wearable devices, are beginning to partially alleviate the shortage of caregivers, more powerful and personalized tools are needed. To address this urgent need, DreamFace Technologies, LLC invented Ryan® CompanionBot, a novel humanoid socially-assistive robot expertly tailored to the specific needs of older adults with early-stage AD/ADRD. The development of Ryan®, with the support of one NSF and two NIA/NIH SBIR grants, has been informed by 100 customer interviews and several subsequent field tests and clinical trials involving more than 50 older adults with early-stage AD/ADRD. In these tests, Ryan® has effectively delivered companionship, engaging conversations, physical and mental stimulation, daily activity reminders, and valuable assistance to the AD/ADRD-afflicted seniors powered by state-of-the-art artificial intelligence technologies such as facial expression recognition and synthesis, brain games, and empathic conversations while we also learned about several additional capabilities required for commercial success. Furthermore, in the pre-launch phase of the initial version of Ryan®, it has been deployed on a subscription basis at the esteemed senior care facility, Morningstar, which has served as a valuable beta site. In this Commercialization Readiness Pilot (CRP) program, we plan to complete the preparation of Ryan® for full commercialization. Specifically, we will: (1) refine and enhance Ryan®'s software and hardware, making mass manufacturing more efficient and cost-effective while making Ryan®'s operation more robust and easier to adopt by family members, staff, administrators, and caregivers in senior care facilities, (2) develop robust, integrated marketing and sales strategies, (3) develop an Intellectual Property strategy and required privacy policy and legal documents and (4) develop a financing and fundraising strategy for the successful commercialization of Ryan®. Upon the completion of the CRP project, we will have all the essential elements in place for the full commercialization of Ryan® as a transformative solution for senior care, benefiting both individuals diagnosed with early-stage AD/ADRD and caregivers alike.", "keywords": [], "approved": true } } ], "meta": { "pagination": { "page": 2, "pages": 1392, "count": 13920 } } }{ "links": { "first": "