Grant List
Represents Grant table in the DB
GET /v1/grants?page%5Bnumber%5D=1394&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=1397&sort=-program_officials", "next": "https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=1395&sort=-program_officials", "prev": "https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=1393&sort=-program_officials" }, "data": [ { "type": "Grant", "id": "11579", "attributes": { "award_id": "5U01CK000643-02", "title": "RFA-CK-22-003, Emerging Infections Sentinel Networks (EISN) Research - 2022", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [], "program_reference_codes": [], "program_officials": [], "start_date": "2022-05-01", "end_date": "2027-04-30", "award_amount": 250000, "principal_investigator": { "id": 23853, "first_name": "DAVID ANDREW", "last_name": "TALAN", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [ { "id": 818, "ror": "", "name": "UNIVERSITY OF CALIFORNIA LOS ANGELES", "address": "", "city": "", "state": "CA", "zip": "", "country": "United States", "approved": true } ] }, "other_investigators": [], "awardee_organization": { "id": 818, "ror": "", "name": "UNIVERSITY OF CALIFORNIA LOS ANGELES", "address": "", "city": "", "state": "CA", "zip": "", "country": "United States", "approved": true }, "abstract": "In 1995, following the Institute of Medicine’s report, “Emerging Infections,” and in response to the CDC’s strategic plan to enhance surveillance, EMERGEncy ID NET was established. EMERGEncy ID NET's goal was to address the threat of emerging infectious diseases by assessing disease prevalence, risk factors, and management practices for acute presentations from the community among a diverse and underserved population of patients presenting to US emergency departments (EDs). A CDC cooperative grant has funded the network for the last 25 years. Due to the ability to prospectively collect clinical data and specimens for on-site laboratory analysis 24/7 from acutely ill patients from the community, EMERGEncy ID NET has been able to produce translational research that has impacted physician practices and informed public health policy. The research network demonstrated its capability to successfully address an urgent public health threat during the COVID-19 pandemic by rapidly implementing 20-site and 16-site public health surveillance projects of ED patient care-related infection risk and vaccine effectiveness among frontline health care personnel. Numerous peer-reviewed publications have resulted from EMERGEncy ID NET research, including in high-impact journals such as The New England Journal of Medicine, the Journal of the American Medical Association, Clinical Infectious Diseases, Emerging Infectious Diseases, and Annals of Emergency Medicine. Aims of EMERGEncy ID NET for the next 5 years are to: 1) identify emerging infections and risk factors for these conditions affecting US ED patients, including among underserved groups; 2) leverage EMERGEncy ID NET’s findings to create new collaborations to develop and improve diagnostic tests, treatments, and vaccines; and 3) disseminate results at national medical conferences, in high-impact journals, and on infectious diseases, public health, and emergency medicine social media outlets to inform treatment and public health policy, and educate the public. Support of EMERGEncy ID NET for the next 5 years will ensure that the network can continue to answer new questions about emerging infections and related areas of high public health priority.", "keywords": [], "approved": true } }, { "type": "Grant", "id": "11607", "attributes": { "award_id": "5U19AI167899-02", "title": "Research Project 1 - The pregnancy ImmunOME", "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": [], "start_date": "2022-04-19", "end_date": "2027-03-31", "award_amount": 747949, "principal_investigator": { "id": 21683, "first_name": "Andrea Goldberg", "last_name": "Edlow", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [ { "id": 736, "ror": "https://ror.org/002pd6e78", "name": "Massachusetts General Hospital", "address": "", "city": "", "state": "MA", "zip": "", "country": "United States", "approved": true } ] }, "other_investigators": [], "awardee_organization": { "id": 210, "ror": "https://ror.org/042nb2s44", "name": "Massachusetts Institute of Technology", "address": "", "city": "", "state": "MA", "zip": "", "country": "United States", "approved": true }, "abstract": "Project 1: Summary The COVID-19 pandemic has revolutionized our ability to decode the rules of maternal immunity. There is a significant gap in knowledge regarding innate and adaptive immune responses over the course of pregnancy and how trimester-specific perturbations in the maternal immunological signature might manifest in attributable risk or benefit to the maternal-fetal dyad. The COVID-19 vaccines and their real-world use by pregnant women present a unique opportunity to define the baseline, trimester-specific immune signature, and to examine the maternal immune response after in vivo perturbation with both de novo (never before seen by the immune system) and recall (boosted responses such as influenza and pertussis) vaccines across the trimesters of pregnancy. In Project 1 in the Maternal ‘Omics to Maximize Immunity (MOMi) consortium, we propose to apply a multi-‘OMICs approach to deeply and comprehensively capture shifts in the maternal immune response before and after maternal immunization across pregnancy. We will profile maternal peripheral blood mononuclear cells, plasma, placental cell isolates, and stool from pre- and post-maternal vaccination, using single cell RNA-Seq (scRNA-Seq), Assay for Transposase-Accessible Chromatin with high-throughput sequencing (scATAC-Seq), Cellular Indexing of Transcriptomes and Epitopes by Sequencing (scCITE-Seq), proteomics, metabolomics, and metagenomics, and integrate all data through the Data Management and Analysis Core (DMAC). In collaboration with Project 2, the ultimate goal is to define maternal immunity longitudinally across pregnancy trimesters in the normal baseline and vaccinated state, in order to build the most comprehensive Pregnancy Immune Atlas of innate and adaptive immune profiling across the maternal-fetal dyad. We will examine how the cellular transcriptome, microbiome, metabolome, and proteome shifts over the course of pregnancy, and how they are modified in response to different vaccine platforms (mRNA, adenovirus, adjuvanted protein) and types (de novo versus recall), providing a unique opportunity to profile the maternal immune response with unprecedented resolution. This detailed map of pregnancy immunity will generate critical data to open previously unrecognized therapeutic windows in this unusual and understudied area of human immunology.", "keywords": [ "Address", "Adenoviruses", "Adjuvant", "Age", "Antigens", "Area", "Atlases", "Biological Assay", "Body mass index", "COVID-19 pandemic", "COVID-19 vaccination", "COVID-19 vaccine", "Cell Separation", "Cells", "Cellular Assay", "Cellular Indexing of Transcriptomes and Epitopes by Sequencing", "Characteristics", "Child", "Chromatin", "Clinical", "Collaborations", "Complex", "Data", "Data Analyses", "Disease", "Enrollment", "Ensure", "Equilibrium", "Fc Receptor", "Feces", "Future", "Goals", "High-Throughput Nucleotide Sequencing", "Human", "Immune", "Immune Tolerance", "Immune response", "Immune system", "Immunity", "Immunologics", "Immunology", "In Vitro", "Infant", "Infection", "Inflammation", "Influenza", "Infrastructure", "Innate Immune Response", "Knowledge", "Maps", "Maternal Mortality", "Maternally-Acquired Immunity", "Messenger RNA", "Metabolic Diseases", "Metagenomics", "Mothers", "Peripheral Blood Mononuclear Cell", "Pertussis", "Pertussis Vaccine", "Plasma", "Population", "Pregnancy", "Pregnancy Trimesters", "Pregnant Women", "Proliferating", "Proteins", "Proteome", "Proteomics", "Recording of previous events", "Research Project Grants", "Resolution", "Risk", "SARS-CoV-2 negative", "Sampling", "Shapes", "Shotguns", "Systems Biology", "Therapeutic", "Time", "Transposase", "Vaccinated", "Vaccination", "Vaccine Design", "Vaccines", "Woman", "Work", "adaptive immune response", "antibody transfer", "booster vaccine", "cell type", "cohort", "data integration", "data management", "early pregnancy", "emerging pathogen", "experimental study", "fetal", "gut microbiome", "healthy pregnancy", "high dimensionality", "in vivo", "influenza virus vaccine", "insight", "maternal immune system", "maternal vaccination", "metabolome", "metabolomics", "metagenomic sequencing", "microbial", "microbiome", "multidisciplinary", "neonate", "next generation", "novel", "pandemic disease", "pathogen", "placental transfer", "prospective", "receptor expression", "response", "sex", "single-cell RNA sequencing", "therapeutic development", "transcriptome", "vaccine development", "vaccine platform", "vaccine response", "vaccine strategy" ], "approved": true } }, { "type": "Grant", "id": "11631", "attributes": { "award_id": "5I01HX003401-02", "title": "COVID-19 Impact on Pain management: Highlighting, Explaining, and Realigning services (CIPHER)", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [], "program_reference_codes": [], "program_officials": [], "start_date": "2022-04-01", "end_date": "2025-03-31", "award_amount": null, "principal_investigator": { "id": 23769, "first_name": "Marianne", "last_name": "Matthias", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [ { "id": 1636, "ror": "", "name": "RLR VA MEDICAL CENTER", "address": "", "city": "", "state": "IN", "zip": "", "country": "United States", "approved": true } ] }, "other_investigators": [ { "id": 23770, "first_name": "Alan Benjamin", "last_name": "McGuire", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] } ], "awardee_organization": { "id": 1636, "ror": "", "name": "RLR VA MEDICAL CENTER", "address": "", "city": "", "state": "IN", "zip": "", "country": "United States", "approved": true }, "abstract": "Background: The COVID-19 pandemic severely disrupted healthcare. These changes are likely to have especially profound implications for Veterans obtaining regular care for chronic conditions, such as chronic pain—particularly when Veterans are obtaining nonpharmacologic treatments, which often require regular, in- person visits. Although virtual care delivery in VA increased dramatically in response to the pandemic and is unlikely to diminish, it is unclear what services Veterans with pain have received via virtual delivery and how Veterans have been affected by these shifts. The goal of this project is to understand changes in VA chronic pain care after COVID-19 and their associated impacts—and use these data to develop strategies to optimize delivery of VA pain services in a post-COVID-19 environment. Significance: This project addresses priorities in HSR&D's Targeted Solicitation on Opioid Safety and Opioid Use Disorder. Specifically, this project is responsive to priority area E, “examination of the impact of the COVID-19 public health emergency on the effective treatment of chronic pain, including effects on access to care, medication and treatment modalities, patient function and satisfaction.” Despite the clear impact on care delivery exerted by the COVID-19 pandemic, little is known about how these changes and disruptions have affected Veterans and their chronic pain care, including their ability to access care after the onset of the pandemic. These concerns are reflected in our pilot data, which indicate that Veterans have felt the impact of service disruptions on their pain management. Innovation and Impact: The effects of the COVID-19 pandemic are not yet well-understood. This project takes a mixed-methods approach to examine changes in chronic pain care since the onset of the pandemic, working with an expert stakeholder panel at each stage of the research to ensure results and recommendations are disseminated rapidly to maximize impact. Specific Aims: Aim 1 (quantitative): Describe changes in pain management visits for Veterans with chronic low- back pain (cLBP) following the onset of the COVID-19 pandemic. Aim 2 (qualitative): Explain differential patterns of receipt of services identified in Aim 1 and perceived impacts. Aim 3: Share findings throughout the project, including final, integrated findings from Aims 1 and 2, and develop short- and long-term strategies to optimize delivery of pain care for Veterans in a post-COVID-19 environment. Methodology: This study uses an embedded mixed-methods design. In Aim 1 a retrospective cohort of Veterans receiving care for cLBP will be established. Receipt of guideline-recommended, nonpharmacological pain management services, including changes in service receipt following the onset of COVID-19, will be examined. Veteran and facility factors associated with differential changes will be tested. Once the first phase of Aim 1 analyses is complete, qualitative sampling for Aim 2 will begin. Thus, consistent with an embedded design, most Aim 1 and Aim 2 activities will take place concurrently. Qualitative interviews with clinicians, administrators, and Veterans will be conducted to better understand how they experienced pandemic-related changes, including any effects on their current practice/service delivery, and to identify positive changes made since the pandemic that could serve as examples for other facilities. In Aim 3 we will work with our stakeholder advisory panel, led by the VA Office of Pain Management, to develop comprehensive recommendations to guide VA on policies to optimize delivery of pain management services in a post-COVID-19 environment. Next Steps: Findings will be directly applied to VA care, as directed by the CIPHER stakeholder panel (led by the VA National Pain Management Office). Future research will examine implementation strategies aimed at supporting VA-wide implementation.", "keywords": [ "Address", "Administrator", "Affect", "Appointment", "Area", "COVID-19", "COVID-19 impact", "COVID-19 pandemic", "COVID-19 pandemic effects", "Caring", "Characteristics", "Chronic Care", "Chronic low back pain", "Data", "Data Collection", "Diagnosis", "Ensure", "Environment", "Event", "Funding", "Future", "Goals", "Guidelines", "Health Services Accessibility", "Healthcare", "Height", "Interruption", "Interview", "Light", "Low Back Pain", "Methodology", "Methods", "Modality", "Office Management", "Opioid", "Opioid Analgesics", "Outcome", "Pain", "Pain management", "Participant", "Patients", "Pattern", "Persons", "Pharmaceutical Preparations", "Phase", "Physical therapy", "Policies", "Publishing", "Race", "Recommendation", "Reporting", "Research", "Retrospective cohort", "Safety", "Sampling", "Services", "Site", "Surveys", "Telephone", "Testing", "Time", "Veterans", "Visit", "Work", "care delivery", "chiropracty", "chronic pain", "chronic pain management", "chronic painful condition", "data warehouse", "design", "disability", "effective therapy", "evidence base", "experience", "health care delivery", "implementation strategy", "innovation", "interest", "multimodality", "opioid epidemic", "opioid use disorder", "pandemic disease", "post-COVID-19", "prescription opioid", "public health emergency", "response", "satisfaction", "service delivery", "telehealth", "virtual delivery", "virtual healthcare", "virtual visit" ], "approved": true } }, { "type": "Grant", "id": "11663", "attributes": { "award_id": "5U19AI167903-02", "title": "Systems biological assessment of B cell responses to vaccination", "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": [], "start_date": "2022-03-07", "end_date": "2027-02-28", "award_amount": 409240, "principal_investigator": { "id": 23528, "first_name": "Scott Dexter", "last_name": "Boyd", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [ { "id": 266, "ror": "https://ror.org/00f54p054", "name": "Stanford University", "address": "", "city": "", "state": "CA", "zip": "", "country": "United States", "approved": true } ] }, "other_investigators": [], "awardee_organization": { "id": 266, "ror": "https://ror.org/00f54p054", "name": "Stanford University", "address": "", "city": "", "state": "CA", "zip": "", "country": "United States", "approved": true }, "abstract": "– Project 3 The focus of Project 3 is to study antigen-specific B cell and plasma cell responses in the context of two timely and fundamental topics in vaccinology: (i) Immunology of COVID-19 vaccines, and (ii) the impact of the microbiota on immune responses to vaccination. The COVID-19 pandemic caused by the novel coronavirus SARS-CoV-2 (CoV-2), and the vaccines developed to combat this pathogen, have underscored a need for greater understanding of primary antibody responses in humans. We will use a systematic panel of cutting- edge humoral immunity analyses to thoroughly characterize antibodies elicited by two CoV-2 vaccines, and the B cell and plasma cell clonal populations required for B cell memory and sustained antibody titers. Our focus will be on the serological, B cell and plasma cell responses elicited by a lipid nanoparticle mRNA vaccine (Pfizer-BioNTech), and a Matrix M-adjuvanted recombinant protein vaccine (Novavax). Combining these analyses with studies of innate immunity (Project 1) and T cell (Project 2) responses to these vaccines should highlight cellular mechanisms correlated with the strength and durability of antibody responses. Rare serious anaphylactoid adverse reactions have been reported for mRNA vaccines, particularly in individuals with a history of food allergy, and those with IgG antibodies specific for polyethylene glycol (PEG). We will examine potential B cell contributions to these anaphylactoid reactions, using specimens from affected individuals who received SARS-COV-2 mRNA vaccines. Finally, we will address the role of the microbiome on humoral immunity to vaccination, with a similar strategy of serological, memory B cell and plasma cell analyses in participants with or without temporarily ablated microbiota following antibiotic treatment. Of particular importance in the aforementioned studies, we will not only analyze peripheral blood B cells and plasmablasts, but also monitor lymph node germinal center reactions by fine-needle aspiration, and sample bone marrow plasma cells in the same participants, to comprehensively study humoral immunity to vaccination in humans. The combined impact of these investigations will likely be clinically significant in guiding the development of future vaccination strategies by uncovering the B cell and plasma cell specificities, differentiation pathways, and longevity stimulated by new SARS-CoV-2 vaccine platforms, and in clarifying the role of the microbiome in vaccine responses to novel antigens.", "keywords": [ "2019-nCoV", "Ablation", "Acute", "Address", "Adjuvant", "Adverse reactions", "Affect", "Affinity", "Allergic", "Antibiotic Therapy", "Antibiotics", "Antibodies", "Antibody Response", "Antibody titer measurement", "Antigens", "Avidity", "B-Cell Antigen Receptor", "B-Lymphocytes", "B-cell receptor repertoire sequencing", "Basophils", "Binding", "Biological Assay", "Blood", "Bone Marrow", "COVID-19", "COVID-19 pandemic", "COVID-19 vaccine", "Cells", "Clinical", "Clone Cells", "Collaborations", "DNA", "Data Analyses", "Development", "Ensure", "Epitopes", "Fine needle aspiration biopsy", "Food Hypersensitivity", "Future", "Glycoproteins", "Human", "Humoral Immunities", "Immune response", "Immunity", "Immunoglobulin G", "Immunoglobulin M", "Immunology", "Individual", "Infection", "Investigation", "Label", "Longevity", "Measures", "Memory B-Lymphocyte", "Methods", "Monitor", "Monoclonal Antibodies", "Natural Immunity", "Participant", "Pathogenicity", "Pathway interactions", "Phenotype", "Plasma Cells", "Plasmablast", "Polyethylene Glycols", "Population", "RNA vaccination", "RNA vaccine", "Rabies", "Rabies Vaccines", "Reaction", "Recombinant Proteins", "Recording of previous events", "Reporting", "Role", "SARS-CoV-2 antigen", "Sampling", "Serology", "Shapes", "Specificity", "Specimen", "Structure of germinal center of lymph node", "T-Lymphocyte", "Vaccination", "Vaccinee", "Vaccines", "Variant", "Viral", "Virus", "Virus Diseases", "adaptive immunity", "biological systems", "clinically significant", "combat", "coronavirus disease", "data management", "in vitro Assay", "lipid nanoparticle", "lymph nodes", "microbiome", "microbiota", "monoclonal antibody production", "multiplex assay", "novel", "novel coronavirus", "pathogen", "peripheral blood", "recruit", "response", "transcriptome", "vaccination strategy", "vaccine development", "vaccine platform", "vaccine response", "vaccinology", "variants of concern" ], "approved": true } }, { "type": "Grant", "id": "11668", "attributes": { "award_id": "5U19AI168631-02", "title": "Immune Phenotyping Core", "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": [], "start_date": "2022-03-22", "end_date": "2027-02-28", "award_amount": 522189, "principal_investigator": { "id": 21760, "first_name": "Ana", "last_name": "Fernandez-Sesma", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [ { "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 } ] }, "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": "The goal of the Immune Phenotyping Core (Core C) is to provide tools and reagents to the different projects that will be used to characterize the immune responses induced by vaccinations and infections with coronaviruses, influenza viruses and dengue viruses. The Core will leverage existing state-of-the-art serological techniques established in the Krammer laboratory, as well as multiplex analysis of cytokine and chemokine plasma profile and Cytek Aurora Spectral Flow Cytometry profiling of PBMCs and human tonsillar histocultures (HC) currently used and optimized in the Fernandez-Sesma laboratory. The following aims re proposed: Aim 1: Characterization of antibody responses to coronavirus, influenza virus and dengue virus vaccination and infection. The Core will provide assays, reagents and protocols to measure binding and functional antibody responses against SARS-CoV-2 for Project 1 and influenza viruses for Project 2. Additionally, the secretion of IgM, IgG and IgA in the supernatant of human tonsil histocultures (HC) treated with the different SARS-CoV-2 vaccines types (Project 1), influenza virus vaccines and viruses (Project 2) will be assessed. Aim 2: Characterization of cytokine/chemokine responses to coronavirus, influenza virus and dengue virus vaccination and infection. The Core will analyze the levels of cytokines and chemokines in the plasma of vaccinated/infected individuals and the supernatant of human tonsil histocultures treated with different vaccines for Projects 1, 2 and 3. Aim 3: Characterization of cellular responses to coronavirus, influenza virus and dengue virus vaccination and infection. Analysis of the cellular immune profiles of PBMCs from vaccinated/infected individuals over time, using Spectral Flow cytometry. Human tonsillar HC will be also analyzed by Cytek Aurora Spectral Flow Cytometry in order to capture early immune signatures and changes in cell populations corresponding to adaptive immune responses in those HC after treatment with different vaccines. We will obtain high-resolution data at the single-cell level to resolve the most challenging cell populations including cells expressing viral antigens. PBMCs will also be subjected to a complementary transcriptomics analysis by RNAseq conducted by the Genomics Core. These tools will serve to generate immune signatures representative of the longitudinal immune responses to vaccination and/or infection in study participants enrolled in observational non-interventional cohort studies in coordination with the Data Management and Analysis Core (Core E). Data obtained using these immunological techniques will be analyzed by the Data management and Analysis Core comparing them across the different systems used in the projects as well as in combination with the genomic data obtained in the Genomics Core (Core D) from the same samples. All data generated by the VIVA Projects and Cores, including the Immune Phenotyping Core, will be deposited by the Data Analysis Core into ImmPort.", "keywords": [ "2019-nCoV", "Aftercare", "Antibody Response", "Assessment tool", "Binding", "Biological Assay", "COVID-19 vaccine", "Cells", "Clinical", "Cohort Studies", "Collaborations", "Coronavirus", "Data", "Data Analyses", "Data Set", "Dengue Virus", "Deposition", "Enzyme-Linked Immunosorbent Assay", "Flow Cytometry", "Genomics", "Goals", "Human", "Immune", "Immune response", "Immunity", "Immunoglobulin A", "Immunoglobulin G", "Immunoglobulin M", "Immunologic Techniques", "Immunology", "Individual", "Infection", "Laboratories", "Letters", "Measures", "Mission", "Peripheral Blood Mononuclear Cell", "Phenotype", "Plasma", "Population", "Process", "Protocols documentation", "Reagent", "Resolution", "Sampling", "Serology", "Serum", "Services", "Standardization", "System", "Techniques", "Testing", "Time", "Tonsil", "Vaccinated", "Vaccination", "Vaccinee", "Vaccines", "Viral", "Viral Antigens", "Virus", "adaptive immune response", "base", "chemokine", "cytokine", "data management", "genomic data", "influenza virus vaccine", "influenzavirus", "instrument", "participant enrollment", "response", "tool", "transcriptome sequencing", "transcriptomics" ], "approved": true } }, { "type": "Grant", "id": "11681", "attributes": { "award_id": "1U01DA057849-01", "title": "Supported employment to create a community culture of SARS-CoV-2 rapid testing among people who inject drugs: PeerConnect2Test", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [], "program_reference_codes": [], "program_officials": [], "start_date": null, "end_date": null, "award_amount": null, "principal_investigator": { "id": 26966, "first_name": "Camille C", "last_name": "Cioffi", "orcid": "https://orcid.org/0000-0003-2424-7473", "emails": "[email protected]", "private_emails": null, "keywords": "[]", "approved": true, "websites": "['psi.uoregon.edu']", "desired_collaboration": "", "comments": "", "affiliations": [] }, "other_investigators": [], "awardee_organization": null, "abstract": "", "keywords": [], "approved": true } }, { "type": "Grant", "id": "11707", "attributes": { "award_id": "1I01CX002400-01A2", "title": "Consequences of social isolation during the COVID-19 pandemic in older adults with and without Alzheimer's disease", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [], "program_reference_codes": [], "program_officials": [], "start_date": "2023-07-01", "end_date": "2027-06-30", "award_amount": null, "principal_investigator": { "id": 27576, "first_name": "Andrew", "last_name": "Budson", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 1521, "ror": "", "name": "VA BOSTON HEALTH CARE SYSTEM", "address": "", "city": "", "state": "MA", "zip": "", "country": "United States", "approved": true }, "abstract": "The COVID-19 pandemic has caused over half a million deaths in the United States alone. Even for those individuals whose physical health and financial security have not been impacted, the consequences of pandemic-induced isolation on mental and cognitive health may be far-reaching— especially for our Veterans. Prior to the pandemic, approximately 10% of older Veterans reported feeling lonely often and 40% indicated feeling lonely some of the time. Due to social distancing guidelines and other measures that limit social contact, loneliness during the pandemic has likely been far greater than these estimates. Decades of research have supported the protective effect of social relationships on cognition in older adulthood; thus, the disruption of social support related to the pandemic may impact cognitive function in the older Veteran population. To address this possibility, we propose a prospective study to examine the relationships between isolation in the pandemic and subsequent cognitive function in healthy older adult Veterans and in older Veterans with Alzheimer’s disease (AD) (Aim 1). In addition, forced isolation is likely not the only factor with important consequences in the pandemic. We propose investigating two other pandemic-related contexts that may either mitigate the effects of isolation in the pandemic or exacerbate the consequences of cognitive impairment on health-related outcomes for older Veterans. First, older Veterans may be able to cope with isolation through use of features of the built environment, such as porches, window views, and public neighborhood space. However, the extent the built environment may be able to mitigate pandemic-related loneliness in older adults is unknown. In Aim 2, we test the hypothesis that built environment factors will mitigate or contribute to social isolation in older Veterans with and without AD. In addition, individuals’ cognition may be impacted by their belief in misconceptions related to social isolation itself. In Aim 3, we test the relationship between cognitive function and the ability to correct misconceptions related to social isolation and AD—and whether such corrections engender changes in behavior. To accomplish Aims 1-3, we will apply questionnaires and neuropsychological tests administered during the first wave of the pandemic (June 2020-June 2021) to 196 participants over three additional one-year time intervals from October 2022 to September 2025. In Aim 1, we will test the relationship between social isolation in the pandemic and cognitive function one, two, and three years later. In Aim 2, we will test whether older Veterans with fewer built environment resources will demonstrate a worsening of social isolation, loneliness, depression, anxiety, and cognition over time. In Aim 3, two objectives will be tested. First, we will determine the extent that individuals with and without cognitive impairment due to AD can successfully change their belief in misconceptions related to social isolation and AD through a correction procedure over time. Second, we will test whether corrections related to changes in behavior at delayed intervals of 1 and 2 years. Preliminary cross-sectional data from our already recruited final sample of older adults (N=196) with and without AD support the relationships between social isolation and cognitive impairment (Aim 1), the built environment and social isolation (Aim 2), and cognitive impairment and misconception correction (Aim 3). As the ramifications of the pandemic continue to unfold for years—possibly decades—it is imperative that we understand the relationship between social isolation and future cognitive impairment in Veterans with and without Alzheimer’s disease.", "keywords": [ "Address", "Adult", "Affect", "Alzheimer&apos", "s Disease", "Alzheimer&apos", "s disease risk", "Anxiety", "Behavior", "Belief", "COVID-19 pandemic", "COVID-19 pandemic effects", "Cessation of life", "Cognition", "Cognitive", "Cognitive Science", "Complex", "Data", "Dementia", "Elderly", "Environment", "Feeling", "Future", "Goals", "Grant", "Guidelines", "Health", "Hippocampus (Brain)", "Home", "Impaired cognition", "Individual", "Light", "Loneliness", "Measures", "Memory Loss", "Mental Depression", "Misinformation", "Modernization", "Moods", "Neighborhoods", "Neuropsychological Tests", "Outcome", "Participant", "Personality", "Personality Character", "Persons", "Procedures", "Prospective Studies", "Psyche structure", "Questionnaires", "Recommendation", "Reporting", "Research", "Resources", "Sampling", "Security", "Shapes", "Social Behavior", "Social Distance", "Social isolation", "Social support", "Stereotyping", "Surveys", "Techniques", "Testing", "Time", "United States", "Variant", "Veterans", "behavior change", "built environment", "cognitive function", "cognitive testing", "coping", "coronavirus disease", "dementia risk", "design", "effective intervention", "emotional factor", "experience", "follow-up", "human old age (65+)", "improved", "long term consequences of COVID-19", "mild cognitive impairment", "military veteran", "neuroinflammation", "pandemic disease", "pandemic response", "physical conditioning", "protective effect", "psychosocial", "recruit", "social cognition", "social contact", "social factors", "social relationships", "time interval", "video chat" ], "approved": true } }, { "type": "Grant", "id": "11710", "attributes": { "award_id": "1I01BX005651-01A2", "title": "Establishing the Therapeutic Efficacy of Alpha-1-Antitrypsin and Enoxaparin Against COVID-19", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [], "program_reference_codes": [], "program_officials": [], "start_date": "2023-07-01", "end_date": "2027-06-30", "award_amount": null, "principal_investigator": { "id": 27580, "first_name": "EDWARD D", "last_name": "CHAN", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 1828, "ror": "https://ror.org/04d7ez939", "name": "VA Eastern Colorado Health Care System", "address": "", "city": "", "state": "CO", "zip": "", "country": "United States", "approved": true }, "abstract": "The clinical outcome for patients with severe COVID-19 remains poor due to the lack of highly efficacious treatment for such individuals. Finding a better remedy for them is an important niche and immediate unmet need. The aim of this pre-clinical project is to substantiate the therapeutic effect of combined alpha-1- antitrypsin (AAT) + enoxaparin (a low molecular weight heparin) against SARS-CoV-2 infection and its consequences. Establishing the efficacy of AAT + enoxaparin combination will provide a necessary foundation for future clinical trials with the goal of employing effective therapy for those with severe COVID-19. We have novel biological evidence supported by Artificial Intelligence-based molecular modeling that enoxaparin synergizes with AAT to inhibit TMPRSS2 (a cell surface protease that activates the spike protein of SARS-CoV-2) and to reduce SARS-CoV-2 burden in primary human airway epithelial cells (hAEc) and monocyte-derived macrophages (MDM). Because both AAT and enoxaparin embrace a panoply of activities that antagonize other pathogenic mechanisms of severe COVID-19 – including anti-inflammatory, anti-thrombotic, pro-autophagy (known to kill MERS-CoV), and endothelial cell protection – we hypothesize that the AAT + enoxaparin combination will be most effective (compared to each alone) in mitigating SARS- CoV-2 infection and its consequences. We will use three complementary models to elucidate the efficacy of AAT, enoxaparin, and combination of both against SARS-CoV-2 infection: (i) primary hAEc since they express high ACE2 levels, fulminant airway disease occurs, and infection of ciliated hAEc and breach of their defense initiates a portal of entry into the lower airways / alveoli to cause COVID-19 pneumonia; (ii) MDM + plasma derived from AAT-deficient individuals immediately before and immediately after receiving routine intravenous AAT since macrophages are key orchestrators of the hyper-inflammatory response seen with COVID-19; and (iii) two murine models, one with wildtype AAT and another with AAT knocked out. Aim 1: Determine in primary hAEc the mechanisms by which AAT, enoxaparin, and both reduce SARS- CoV-2 infection and its consequences. Approach: hAEc will be infected with SARS-CoV-2 followed by no treatment or treatment with AAT, enoxaparin, or combination of both and assayed for viral load, autophagic flux, pro-inflammatory cytokines, and hAEc viability and barrier integrity. Aim 2: Determine in macrophages the mechanisms by which AAT (given in vivo) ± enoxaparin mitigate SARS-CoV-2 infection. Approach: infect human MDM cultured in autologous plasma – prepared from AAT- deficient individuals before and after AAT infusions ± ex vivo enoxaparin – with SARS-CoV-2 and determine viral load, autophagic flux, and pro-inflammatory cytokine / macrophage extracellular trap (METs) production. Aim 3: Determine if SARS-CoV-2 infection of mice is mitigated by AAT, enoxaparin, and combination of both. Approach: transgenic (Tg) mice bred to express human ACE2 with or without deletion of AAT (to represent humans who are AAT-replete and AAT-deficient, respectively) will either be left untreated or treated with AAT, enoxaparin, or the combination along with SARS-CoV-2 infection. From the lungs and spleens, we will quantify viral load and the phenotypes of macrophages, dendritic cells, CD4+ T cells, and CD8+ T cells in both organs, as well as analyze the lungs for epithelial and endothelial injury, co-localization of SARS-CoV-2 with airway and alveolar epithelial cells, and neutrophil extracelluar trap (NETs) formation (both METs and NETs implicated in immunothrombosis of severe COVID-19). Establishing the efficacy of AAT + enoxaparin combination will provide a foundation for future clinical trials with the goal of employing more effective therapy for veterans and non-veterans with severe COVID-19.", "keywords": [ "2019-nCoV", "ACE2", "Airway Disease", "Alveolus", "Anti-Inflammatory Agents", "Artificial Intelligence", "Autologous", "Autophagocytosis", "Biochemical", "Biological", "Biological Assay", "Blood Vessels", "Breeding", "CD4 Positive T Lymphocytes", "CD8-Positive T-Lymphocytes", "COVID-19", "COVID-19 impact", "COVID-19 pneumonia", "Cell Survival", "Cell surface", "Cells", "Clinical", "Clinical Trials", "Cytoprotection", "Data", "Dendritic Cells", "Endothelial Cells", "Endothelium", "Enoxaparin", "Enzymes", "Epithelial", "Epithelial Cells", "Foundations", "Future", "Goals", "Human", "Individual", "Infection", "Inflammatory", "Inflammatory Response", "Inflammatory Response Pathway", "Infusion procedures", "Injury", "Intravenous", "Knock-out", "Left", "Low-Molecular-Weight Heparin", "Lung", "Middle East Respiratory Syndrome Coronavirus", "Modeling", "Mus", "Organ", "Pathogenicity", "Patient-Focused Outcomes", "Peptide Hydrolases", "Phenotype", "Plasma", "Pre-Clinical Model", "Production", "Property", "Proteins", "SARS-CoV-2 infection", "SARS-CoV-2 inhibitor", "SARS-CoV-2 spike protein", "Safety", "Spleen", "TMPRSS2 gene", "Testing", "Therapeutic Effect", "Thrombosis", "Transgenic Mice", "Treatment Efficacy", "Veterans", "Viral Load result", "Virus", "airway epithelium", "alpha 1-Antitrypsin", "alpha 1-Antitrypsin Deficiency", "alveolar epithelium", "antagonist", "cell injury", "cell type", "cytokine", "cytotoxicity", "effective therapy", "efficacious treatment", "extracellular", "immunothrombosis", "in vivo", "macrophage", "molecular modeling", "monocyte", "mouse model", "neutrophil", "novel", "pre-clinical", "prevent", "severe COVID-19", "synergism", "therapeutic evaluation", "thrombotic" ], "approved": true } }, { "type": "Grant", "id": "11718", "attributes": { "award_id": "1I21RX004371-01A1", "title": "Fatigue and Fatigability in Veterans Following SARS-CoV-2 Infection", "funder": { "id": 4, "ror": "https://ror.org/01cwqze88", "name": "National Institutes of Health", "approved": true }, "funder_divisions": [], "program_reference_codes": [], "program_officials": [], "start_date": "2023-07-01", "end_date": "2025-06-30", "award_amount": null, "principal_investigator": { "id": 27588, "first_name": "JARED M.", "last_name": "GOLLIE", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 2041, "ror": "", "name": "U.S. DEPT/VETS AFFAIRS MEDICAL CENTER", "address": "", "city": "", "state": "DC", "zip": "", "country": "United States", "approved": true }, "abstract": "The chronic presence of elevations in fatigue suggests that individuals with post-COVID-19 fatigue (PCF) may be at risk for experiencing increased fatigability when engaging in physical activity. An individual’s level of fatigability is determined by the interactions between objective declines in performance (performance fatigability) and perceptual changes regulating activity performance (i.e., perceived fatigability). Skeletal muscle alterations including reduced force capacity, fiber atrophy, and metabolic dysfunction have been observed in patients following SARS-CoV-2 infection. In addition, depression and anxiety have been identified as common pre-existing conditions in patients with PCF. Thus, psychological as well as physiological consequences of SARS-CoV-2 may provide insight into co-occurring mechanisms by which increased fatigability occurs and persists following SARS-CoV-2 infection. While there have been widespread calls for the implementation of rehabilitation for individuals recovering from SARS-CoV-2 infection, relatively little is known about the potential benefits of physical exercise in this population. Home-based exercise programs maybe particularly advantageous due to its ability to provide an exercise option to combat sedentary behavior imposed by lockdowns, quarantines, and restrictions on community activity caused by the COVID-19 pandemic. To date, the safety and feasibility of home-based exercise has yet to be thoroughly investigated in people previously infected by SARS-CoV-2. The overall goal of this project is to advance our understanding of underlying mechanisms impacting performance fatigability and perceived fatigability in Veterans with PCF and explore the safety and feasibility of a home-based “minimal-dose” resistance exercise program in this population. The central hypothesis is that declines in force capacity, skeletal muscle oxygen extraction, and affective responses to physical activity offer potential mechanisms through which fatigability is increased in Veterans with PCF. Moreover, home-based resistance exercise delivered remotely may provide a safe and feasible treatment option for this population. To address this hypothesis, the following aims are proposed: Aim 1: Compare fatigability, and the underlying neuromuscular and neurobiological factors, in Veterans with post- COVID-19 fatigue to Veterans previously infected by SARS-CoV-2 without fatigue. Hypothesis 1a. Veterans with post-COVID-19 fatigue will exhibit greater performance fatigability and perceived fatigability compared to Veterans previously infected with SARS-CoV-2 not reporting post-COVID-19 fatigue. Hypothesis 1b. Veterans with post-COVID-19 fatigue will exhibit reductions in knee extensor force capacity, muscle oxygen extraction, and affective valence compared to Veterans previously infected with SARS-CoV-2 not reporting post-COVID-19 fatigue. Aim 2: Determine the safety and feasibility of an 8-week home-based “minimal-dose” resistance exercise program for Veterans with post-COVID-19 fatigue. Hypothesis 2a. 8 weeks of home-based minimal- dose resistance exercise will be safe and feasible in Veterans with post-COVID-19 fatigue. To test aim 2, we will use a randomized controlled feasibility trial. Skeletal muscle force will be assessed during isometric and dynamic knee extensor contractions using dynamometry and muscle oxygen extraction of the vastus lateralis will be estimated using near-infrared spectroscopy. Affective responses will be measured using the Feeling Scale. Secondary measures include motor unit properties, functional outcomes, and health-related quality of life.", "keywords": [ "2019-nCoV", "Address", "Affective", "Anxiety", "Atrophic", "Award", "Awareness", "COVID-19 pandemic", "Chronic", "Clinical", "Clinical Trials", "Communities", "Data", "Dose", "Effectiveness", "Exercise", "Exhibits", "Extensor", "Fatigue", "Feeling", "Fiber", "Goals", "Health", "Home", "Individual", "Isometric Exercise", "Knee", "Measures", "Mental Depression", "Metabolic dysfunction", "Motor", "Muscle", "Near-Infrared Spectroscopy", "Neurobiology", "Oxygen", "Patients", "Performance", "Persons", "Physical Exercise", "Physical activity", "Physiological", "Population", "Prevalence", "Property", "Quarantine", "Randomized", "Rehabilitation therapy", "Reporting", "Risk", "SARS-CoV-2 infection", "Severities", "Skeletal Muscle", "Symptoms", "Testing", "Veterans", "appropriate dose", "base", "combat", "design", "exercise program", "experience", "feasibility trial", "functional outcomes", "health related quality of life", "improved", "insight", "neurobehavioral", "neuromuscular", "novel", "patient population", "post SARS-CoV-2 infection", "post-COVID-19", "psychologic", "remote delivery", "resistance exercise", "response", "safety and feasibility", "sedentary lifestyle", "skeletal muscle weakness", "vastus lateralis" ], "approved": true } }, { "type": "Grant", "id": "11725", "attributes": { "award_id": "1P30GM145500-01A1", "title": "Lentivirus Construct Core", "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": [], "start_date": "2023-07-01", "end_date": "2028-06-30", "award_amount": 213200, "principal_investigator": { "id": 27595, "first_name": "OLIN D.", "last_name": "Liang", "orcid": null, "emails": "", "private_emails": "", "keywords": null, "approved": true, "websites": null, "desired_collaboration": null, "comments": null, "affiliations": [] }, "other_investigators": [], "awardee_organization": { "id": 1416, "ror": "https://ror.org/01aw9fv09", "name": "Rhode Island Hospital", "address": "", "city": "", "state": "RI", "zip": "", "country": "United States", "approved": true }, "abstract": "Project Summary/Abstract: The past 20 years have seen a rapid expansion in the use of viral gene transfer vectors, with approved therapies and late stage clinical trials underway for the treatment of genetic disorders and multiple forms of cancer, as well as prevention of infectious diseases through vaccination. Major innovations in vector design and virus production have been accomplished for the three most widely used viral vector systems based on adenovirus, adeno- associated virus (AAV), and lentivirus. For laboratory investigators, cell and molecular biology methods to stably over-express and knockout a gene in cells and tissues have become indispensable in modern biomedical research. Lentivirus-mediated over-expression and Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) knockout techniques are particularly powerful due to their efficiency and the capability of infecting dividing and non-dividing cells. Given the significant need and demand to use these viral gene transfer technologies and the lack of expert service providers in Rhode Island and the rest Southern New England region, we propose a Lentivirus Construct Core for the Stem Cells and Aging (SCA) COBRE Phase 3. Our long-term goal is to provide cutting-edge viral gene transfer technologies to the greater biomedical research community in Rhode Island and beyond. To accomplish this goal, we propose the following 4 Specific Aims: Specific Aim 1. To provide lentivirus technologies for easy access and efficient use. Specific Aim 2. To enhance the competitiveness of Rhode Island investigators to secure federal research funding. Specific Aim 3. To align our Core with translational research. Specific Aim 4. To become an independent self-sustainable service research facility. Innovations and impact: Recombinant viral vectors are powerful gene delivery tools for cells, animal models, and clinical applications. The lentiviral constructs from our Core will differ in their suitability for different applications, and will allow investigators to monitor cell functions, replace, correct, express or block expression of target genes, tag cells for fate determination, and change the physiological state of specific cell populations. The timely development of COVID-19 pseudovirus variants by our Core was a prime example of innovation. To genetically engineer oncolytic adenovirus encoding bispecific T cell engagers is cutting-edge, and the novel immunovirotherapies have the potential to make a profound impact in cancer treatments. The current exponential growth of clinical trials using AAV vectors suggests that we are only at the beginning of what is achievable for AAV as the leading platform for gene therapies. These innovations can potentially address diseases that have no other treatment options. In this vein, the Lentivirus Construct Core has already successfully made and will continue to make a positive impact as a catalyst on basic and translational research to improve human health.", "keywords": [ "2019-nCoV", "Address", "Adenoviruses", "Adoption", "Aging", "Animal Model", "Basic Science", "Biology", "Biomedical Research", "COVID-19", "COVID-19 pandemic", "Caliber", "Cancer Center", "Cardiovascular system", "Cell physiology", "Cells", "Cellular biology", "Centers of Research Excellence", "Clinical Trials", "Clustered Regularly Interspaced Short Palindromic Repeats", "Collaborations", "Communicable Diseases", "Communities", "Country", "Data", "Dependovirus", "Development", "Disease", "Faculty", "Fees", "Funding", "Gene Delivery", "Gene Transduction Agent", "Genes", "Genetic Diseases", "Genetic Engineering", "Goals", "Grant", "Growth", "Health", "Health Services Research", "Hematologic Neoplasms", "Hospitals", "Human", "Infrastructure", "Institution", "Interphase Cell", "K-Series Research Career Programs", "Knock-out", "Laboratories", "Lentivirus", "Malignant Neoplasms", "Mediating", "Medical center", "Methods", "Modernization", "Molecular Biology", "Monitor", "Neurosciences", "New England", "Orthopedics", "Pathogenesis", "Phase", "Physicians", "Physiological", "Population", "Prevention", "Production", "Research", "Research Activity", "Research Personnel", "Rest", "Rhode Island", "Role", "Scientist", "Secure", "Series", "Services", "Solid Neoplasm", "Strategic Planning", "System", "Techniques", "Technology", "Technology Transfer", "Tissues", "Translational Research", "United States National Institutes of Health", "Universities", "Vaccination", "Variant", "Veins", "Viral", "Viral Genes", "Viral Vector", "Virus", "adeno-associated viral vector", "bi-specific T cell engager", "cancer therapy", "catalyst", "clinical application", "cost", "design", "diagnostic tool", "experimental study", "gene therapy", "gene transfer vector", "immunotherapeutic virotherapy", "improved", "innovation", "interest", "laboratory equipment", "medical schools", "meetings", "member", "novel", "novel diagnostics", "novel therapeutics", "oncolytic adenovirus", "overexpression", "recombinant viral vector", "research facility", "service providers", "sound", "stem cells", "tool", "translational research program", "vector" ], "approved": true } } ], "meta": { "pagination": { "page": 1394, "pages": 1397, "count": 13961 } } }{ "links": { "first": "