Represents Grant table in the DB

GET /v1/grants?page%5Bnumber%5D=1383&sort=start_date
HTTP 200 OK
Allow: GET, POST, HEAD, OPTIONS
Content-Type: application/vnd.api+json
Vary: Accept

{
    "links": {
        "first": "https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=1&sort=start_date",
        "last": "https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=1397&sort=start_date",
        "next": "https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=1384&sort=start_date",
        "prev": "https://cic-apps.datascience.columbia.edu/v1/grants?page%5Bnumber%5D=1382&sort=start_date"
    },
    "data": [
        {
            "type": "Grant",
            "id": "15519",
            "attributes": {
                "award_id": "1R13AI189261-01",
                "title": "HIV-DART 2024",
                "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": 22396,
                        "first_name": "Martin",
                        "last_name": "Gutierrez",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-11-08",
                "end_date": "2025-10-31",
                "award_amount": 30000,
                "principal_investigator": {
                    "id": 22933,
                    "first_name": "Raymond Felix",
                    "last_name": "Schinazi",
                    "orcid": null,
                    "emails": "",
                    "private_emails": "",
                    "keywords": null,
                    "approved": true,
                    "websites": null,
                    "desired_collaboration": null,
                    "comments": null,
                    "affiliations": [
                        {
                            "id": 265,
                            "ror": "https://ror.org/03czfpz43",
                            "name": "Emory University",
                            "address": "",
                            "city": "",
                            "state": "GA",
                            "zip": "",
                            "country": "United States",
                            "approved": true
                        }
                    ]
                },
                "other_investigators": [],
                "awardee_organization": {
                    "id": 265,
                    "ror": "https://ror.org/03czfpz43",
                    "name": "Emory University",
                    "address": "",
                    "city": "",
                    "state": "GA",
                    "zip": "",
                    "country": "United States",
                    "approved": true
                },
                "abstract": "Abstract/ Project Summary Thanks to advances in antiretroviral therapy, the face of HIV and AIDS has been gradually and meaningfully transformed from a deadly disease, into a chronic condition that is managed with careful adherence to life-long treatment regimens. The need for improved drugs, with fewer side effects, improved adherence, and more favorable pharmacokinetics increases as the number of infected individuals on long-term therapy continues to rise. Importantly, the quest for an HIV cure continues to gain momentum as we learn more about viral reservoirs and mechanisms of viral persistence. HIV-DART is a biennial conference for physicians, clinicians, nurses, scientists, and clinical researchers that will focus on the latest discoveries in these areas. Specific Aims: 1. To gather cross-disciplinary professionals such as clinicians, health care workers, researchers, and basic scientists together in an interactive workshop setting to share and discuss the latest developments in the field of HIV; 2. To facilitate the neutral and balanced exchange of scientific knowledge with regard to antiviral drug development, vaccine development, and recent progress in curative strategies; 3. To enhance the training of young scientists such as graduate students and post-doctoral fellows and provide increased opportunities for underrepresented scientists and health care workers through attendee scholarships. Educational Objectives: 1. Advances in antiretroviral therapy, long-acting formulations, and injectable drugs for HIV; 2. Novel approaches to suppress and eliminate HIV reservoirs; 3. HIV cure strategies; 4. HIV in the aging population; 5. Novel vaccines and immunological therapies in development; 6. Treatment as prevention and PrEP; 7. HIV co-infections with HCV, HBV, and/or SARS-CoV-2. Design & Methods: HIV-DART 2024 will take place over 2.5 days, at a location that is easily accessed by both national and international delegates. The program will include plenary lectures, oral abstract presentations, discussion panels, and a debate. This ACCME-accredited program will provide ample time for formal and informal networking and discussion amongst the delegates during the poster session, coffee breaks, and meal breaks.",
                "keywords": [],
                "approved": true
            }
        },
        {
            "type": "Grant",
            "id": "15521",
            "attributes": {
                "award_id": "1R21AI181742-01A1",
                "title": "SAMHD1-mediated regulation of innate immunity during SARS-CoV-2 infection",
                "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": 6173,
                        "first_name": "QIAN",
                        "last_name": "Liu",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-11-12",
                "end_date": "2026-10-31",
                "award_amount": 188544,
                "principal_investigator": {
                    "id": 21045,
                    "first_name": "Li",
                    "last_name": "Wu",
                    "orcid": null,
                    "emails": "",
                    "private_emails": "",
                    "keywords": null,
                    "approved": true,
                    "websites": null,
                    "desired_collaboration": null,
                    "comments": null,
                    "affiliations": [
                        {
                            "id": 220,
                            "ror": "https://ror.org/036jqmy94",
                            "name": "University of Iowa",
                            "address": "",
                            "city": "",
                            "state": "IA",
                            "zip": "",
                            "country": "United States",
                            "approved": true
                        }
                    ]
                },
                "other_investigators": [],
                "awardee_organization": {
                    "id": 220,
                    "ror": "https://ror.org/036jqmy94",
                    "name": "University of Iowa",
                    "address": "",
                    "city": "",
                    "state": "IA",
                    "zip": "",
                    "country": "United States",
                    "approved": true
                },
                "abstract": "The COVID-19 pandemic caused by severe acute respiratory syndrome-related coronavirus 2 (SARS-CoV-2) remains a threat to public health, particularly in aging populations. The top priority for COVID-19 research is to improve fundamental knowledge of SARS-CoV-2 and viral pathogenesis, including studies to characterize the virus and to understand the host immunity. Our proposed study aims to better understand the molecular mechanisms underlying SARS-CoV-2 infection and innate immune regulation, which is important for controlling COVID-19 and potential future threats of other emerging coronaviruses. Sterile alpha motif (SAM) and HD-domain containing protein 1 (SAMHD1) is the unique mammalian dNTP hydrolase (dNTPase) that also regulates innate and adaptive immunity in hosts. Importantly, SAMHD1 phosphorylation negatively regulates its dNTPase activity and antiviral function. We found that SAMHD1 negatively regulates antiviral innate immune responses and inflammation through interacting with various key proteins in innate immune signaling in macrophages. SAMHD1 transcript is upregulated in SARS-CoV-2 infected primary human bronchial epithelial cells. Our preliminary data showed that SARS-CoV-2 infection of human lung epithelial cell lines or primary human airway epithelial (HAE) cultures significantly upregulated SAMHD1 phosphorylation. However, the function of SAMHD1 in regulating SARS-CoV-2 replication and innate immunity remains unclear. Our hypothesis is that SARS-CoV-2 infection increases inflammation in lung and airway epithelial cells by upregulating phosphorylation of SAMHD1, thereby reducing SAMHD1-mediated anti- inflammation effects. Our established primary HAE cultures provided a physiologically relevant in vitro model to study SARS-CoV-2 infection and cellular responses. We will also use an established mouse model of SARS-CoV-2 infection as a complementary in vivo approach in our studies. We designed two specific aims to test our hypothesis: Aim 1. To examine the role of SAMHD1 in SARS-CoV-2-induced inflammation of primary HAE cultures and SAMHD1- knockout mice; and Aim 2. To investigate the mechanisms of SARS-CoV-2-upregulated phosphorylation of human SAMHD1. Accomplishing our multidisciplinary studies will help define the mechanisms by which SAMHD1 regulates SARS-CoV-2 replication, inflammation, and antiviral innate immunity.",
                "keywords": [],
                "approved": true
            }
        },
        {
            "type": "Grant",
            "id": "15517",
            "attributes": {
                "award_id": "1R01AI183645-01A1",
                "title": "Assessing functional immunity to influenza infection by quantifying BCR binding avidities",
                "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": 26918,
                        "first_name": "Michelle Marie",
                        "last_name": "Arnold",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-11-13",
                "end_date": "2029-10-31",
                "award_amount": 811009,
                "principal_investigator": {
                    "id": 22692,
                    "first_name": "Nicole",
                    "last_name": "Baumgarth",
                    "orcid": null,
                    "emails": "",
                    "private_emails": "",
                    "keywords": null,
                    "approved": true,
                    "websites": null,
                    "desired_collaboration": null,
                    "comments": null,
                    "affiliations": [
                        {
                            "id": 746,
                            "ror": "",
                            "name": "UNIVERSITY OF CALIFORNIA AT DAVIS",
                            "address": "",
                            "city": "",
                            "state": "CA",
                            "zip": "",
                            "country": "United States",
                            "approved": true
                        }
                    ]
                },
                "other_investigators": [
                    {
                        "id": 22693,
                        "first_name": "Steven CARL",
                        "last_name": "George",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    },
                    {
                        "id": 32060,
                        "first_name": "Venktesh",
                        "last_name": "Shirure",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    },
                    {
                        "id": 32061,
                        "first_name": "Xiangdong",
                        "last_name": "Zhu",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "awardee_organization": {
                    "id": 746,
                    "ror": "",
                    "name": "UNIVERSITY OF CALIFORNIA AT DAVIS",
                    "address": "",
                    "city": "",
                    "state": "CA",
                    "zip": "",
                    "country": "United States",
                    "approved": true
                },
                "abstract": "Activation, clonal expansion, and affinity maturation of antigen-specific B cells are considered the hallmarks of adaptive immunity; hence, the assessment of B cell responses is thought to provide correlates of immune protection. Antigen-specific B cell responses have traditionally been observed indirectly, by examining the serum antibody pool using ELISA, or in the case of influenza the hemagglutinin inhibition assay (HIA). Direct measurements of antibody-secreting cells have also been done using ELISPOT, or more recently by flow cytometry. However, there are currently no tools to assess the functionality of the non-antibody (Ab) secreting memory B cell (Bmem) pool. This is an important limitation as Bmem, more than the circulating Ab pool, contain cross-reactive and cross-protective cells that could respond to a challenge infection with a mutated pathogen variant, such as occurs during the seasonal yearly influenza surges. Knowing the extent to which Bmem cells exist and can bind to the original or emerging variants of a pathogen would greatly help, for example, with decisions about when to update vaccines to the circulating seasonal influenza or SARS-COV-2 strains. The primary objective of this proposal is to generate such a tool. Our recent work demonstrates the proof-of- concept that a simple microfluidic platform can capture the full avidity spectrum of antigen-specific B cells, and that the equilibrium binding avidity of the BCR correlates strongly with the binding affinity of the secreted antibody. We have dubbed this approach and technology the Shear-force Avidity-based Cell Selector (SACS). These findings provide the foundation of our central hypothesis: capture, separation, and quantification of peripheral B cells based on the force-dependent BCR binding avidity can be used as a measure of functional immune status. To test our hypothesis, and fully develop the SACS technology, the specific aims of this project are to: 1) enhance the design and optimize a microfluidic device with the capacity to capture and separate a complex pool of B cells based on BCR binding avidity to a defined antigen; and 2) characterize the relationship between binding avidity dynamics of influenza-specific peripheral B cells and immune status following influenza virus infection. Our platform will demonstrate the functionality of a rapid and easily adaptable system to evaluate the dynamics of the B cell response to influenza, but is completely adaptable to host of other pathogens such as SARS-Cov-2. Expected results would provide a new measurement platform (SACS) that would allow for the first time the rapid assessment of the range of functional BCR-antigen interaction strengths of individual B cells within the complex pools of peripheral B cells and B cell subsets to predict functional immunity.",
                "keywords": [],
                "approved": true
            }
        },
        {
            "type": "Grant",
            "id": "15665",
            "attributes": {
                "award_id": "2444914",
                "title": "I-Corps: Translation Potential of an Online Healthcare Information (OHI) Trust Badge",
                "funder": {
                    "id": 3,
                    "ror": "https://ror.org/021nxhr62",
                    "name": "National Science Foundation",
                    "approved": true
                },
                "funder_divisions": [
                    "Technology, Innovation and Partnerships (TIP)",
                    "I-Corps"
                ],
                "program_reference_codes": [],
                "program_officials": [
                    {
                        "id": 602,
                        "first_name": "Ruth",
                        "last_name": "Shuman",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-11-15",
                "end_date": null,
                "award_amount": 50000,
                "principal_investigator": {
                    "id": 32174,
                    "first_name": "Ankur",
                    "last_name": "Chattopadhyay",
                    "orcid": null,
                    "emails": "",
                    "private_emails": "",
                    "keywords": null,
                    "approved": true,
                    "websites": null,
                    "desired_collaboration": null,
                    "comments": null,
                    "affiliations": []
                },
                "other_investigators": [
                    {
                        "id": 32173,
                        "first_name": "Seth A",
                        "last_name": "Adjei",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "awardee_organization": {
                    "id": 1396,
                    "ror": "https://ror.org/01k44g025",
                    "name": "Northern Kentucky University",
                    "address": "",
                    "city": "",
                    "state": "KY",
                    "zip": "",
                    "country": "United States",
                    "approved": true
                },
                "abstract": "The broader impact of this I-Corps project is the development of an online knowledge recommender tool and trust badge for consumers. Health misinformation remains a serious societal threat. Since the emergence of the COVID-19 pandemic, reports show on average that 8 out of 10 Americans search for online healthcare information (OHI), and 4 out of 10 Americans cannot correctly identify false healthcare claims. The goal of the new technology is to help alleviate confusion amongst consumers caused by the overwhelming amount of OHI, and to help OHI providers boost their reputation as a trustworthy source. The tool is designed to combat misinformation by proactively serving a wide spectrum of stakeholders who regularly deal with OHI content. The I-Corps project will focus on the specific issues and public challenges of endorsements in addition to fact checking of OHI content and contributing to a better understanding of the needs of people who use and/or provide OHI content. This solution serves as a foundation for a consultancy service providing platform offering advice plus training to OHI consumers and OHI providers.    This I-Corps project utilizes experiential learning coupled with a first-hand investigation of the industry ecosystem to assess the translation potential of a software tool that will serve online healthcare information (OHI) users by providing machine learning-based classification and certification of OHI content trustworthiness. Research has shown that machine learning-based classifiers can process OHI claims and classify them as fact or fake, but such solutions have not been directly integrated into web browsers and have been trained with primarily textual cues from mostly unimodal datasets. This technology addresses these limitations and is designed as a machine learning driven online knowledge recommender tool, prototyped as a web extension utility, which can be directly embedded into web browsers to seamlessly report trustworthiness of any OHI content. the solution is designed as a trust badge model for easy certification of web content and can function both as an online content classifier. This capability may allow both OHI consumers and OHI providers to validate and tag OHI websites' trustworthiness. Additionally, the solution is trained with multimodal data, that includes both textual and visual cues (e.g., image elements, graphic contents, and infographics), unlike existing solutions that do not include visual cues or image artifacts.    This award reflects NSF's statutory mission and has been deemed worthy of support through evaluation using the Foundation's intellectual merit and broader impacts review criteria.",
                "keywords": [],
                "approved": true
            }
        },
        {
            "type": "Grant",
            "id": "15516",
            "attributes": {
                "award_id": "1R01AI182043-01A1",
                "title": "Harnessing nutrition to enhance vaccine responses",
                "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": 6181,
                        "first_name": "Deborah",
                        "last_name": "Hodge",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-11-18",
                "end_date": "2029-10-31",
                "award_amount": 631977,
                "principal_investigator": {
                    "id": 32059,
                    "first_name": "Nicholas",
                    "last_name": "Collins",
                    "orcid": null,
                    "emails": "",
                    "private_emails": "",
                    "keywords": null,
                    "approved": true,
                    "websites": null,
                    "desired_collaboration": null,
                    "comments": null,
                    "affiliations": []
                },
                "other_investigators": [],
                "awardee_organization": {
                    "id": 825,
                    "ror": "",
                    "name": "WEILL MEDICAL COLL OF CORNELL UNIV",
                    "address": "",
                    "city": "",
                    "state": "NY",
                    "zip": "",
                    "country": "United States",
                    "approved": true
                },
                "abstract": "Novel pathogens (e.g., SARS-CoV2, monkeypox, respiratory syncytial virus, and influenza) are constantly emerging, creating an urgent, unmet need to identify strategies that enhance immunological memory. Our previous work demonstrated that caloric restriction (CR) is a nutritional intervention that enhances immunological memory in mice, resulting in 500-fold greater pathogen control (Collins N, Cell, 2019 and Han SJ…Collins N*, PNAS, 2023). However, the molecular determinants, signaling pathways, and cell-cell interactions involved are unknown. The objective of this proposal is to generate fundamental mechanistic information that fills these gaps in knowledge, which will advance the field and ultimately reduce the burden of infectious diseases by (i) revealing how CR can be utilized to enhance pathogen- and vaccine-elicited immunological memory, and (ii) leading to the development of novel therapies that pharmacologically mimic the immune-enhancing effect of CR. Our recent publication indicated that CR enhances immunity by increasing memory CD8+ T cell-derived IFN and gut microbiota-derived acetate, which converge on myeloid cells to enhance their capacity to kill pathogens. Our preliminary metabolomics data first show that CR potently modulates systemic nutrient abundance. Memory CD8+ T cells metabolically adapt by switching their intracellular signaling pathways to promote the utilization of nutrient sources that are abundant in this setting, as shown by preliminary single-cell RNA-seq and flow cytometry results. The metabolic state adopted by memory T cells during CR has been shown to enhance their function, providing a potential explanation for increased IFN production. A second essential effect of CR demonstrated by metagenomics sequencing in our recent publication is a ~1000-fold enrichment of the commensal Bifidobacteria that produce the short-chain fatty acid acetate. Ex vivo data indicates that the combination of IFN and acetate is essential to enhance the ability of myeloid cells from mice on CR to kill pathogens. Further, our RNA-seq identified that myeloid cells upregulate multiple genes related to pathogen killing in vivo during CR. This proposal builds on our recent publication and extensive preliminary data and will test the central hypothesis that CR induces memory CD8+ T cell metabolic rewiring to enhance the production of IFN, which combines with Bifidobacteria-derived acetate to increase the capacity of myeloid cells to kill pathogens. Based on our new data, this proposal will determine if (i) CR-induced metabolic reprogramming is essential to enhance memory T cell IFN production, and (ii) how the combination of memory T cell-derived IFN and microbiota-derived acetate enhances myeloid cells in this context. Altogether, we will define how CR enhances and promotes cooperation between distinct immune cells for increased pathogen control. This will be essential to achieving our long-term goal of reducing the burden of infectious disease by optimizing immunological memory.",
                "keywords": [],
                "approved": true
            }
        },
        {
            "type": "Grant",
            "id": "15518",
            "attributes": {
                "award_id": "1R01AI179729-01A1",
                "title": "Defining mucosal plasma cell origin, residence, and longevity in the upper airway",
                "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": 9207,
                        "first_name": "Michael",
                        "last_name": "Minnicozzi",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-11-18",
                "end_date": "2029-10-31",
                "award_amount": 640221,
                "principal_investigator": {
                    "id": 27602,
                    "first_name": "E. Ashley",
                    "last_name": "Moseman",
                    "orcid": null,
                    "emails": "",
                    "private_emails": "",
                    "keywords": null,
                    "approved": true,
                    "websites": null,
                    "desired_collaboration": null,
                    "comments": null,
                    "affiliations": []
                },
                "other_investigators": [],
                "awardee_organization": {
                    "id": 246,
                    "ror": "https://ror.org/00py81415",
                    "name": "Duke University",
                    "address": "",
                    "city": "",
                    "state": "NC",
                    "zip": "",
                    "country": "United States",
                    "approved": true
                },
                "abstract": "The upper respiratory tract is lined by two distinct mucosal tissues. Respiratory mucosa humidifies and modulates incoming air temperature as it passes toward the lungs. Olfactory mucosa on the other hand is specialized for chemosensation and supports detection of airborne odorants by olfactory sensory neurons (OSNs). We have recently described an endothelial level barrier, a “blood-olfactory barrier” (BOB), that prevents high molecular weight proteins like antibodies (~150kD) from accessing the olfactory mucosa. Antibodies are absolutely required to prevent olfactory reinfection by airway viral infections, but serum neutralizing antibody provides no protection. Our studies have found that antibody secreting plasma cells (PCs) must reside within olfactory tissues- beyond the BOB- to provide locally neutralizing antibody in order to prevent olfactory infection. While vaccines generate robust circulating antibody titers, we found that they generally fail to differentiate OlfPC and as such, fail to protect against olfactotropic pathogens (that infect the olfactory mucosa), including SARS-Cov-2. Tissue specific PCs are generally understudied and this proposal will specifically establish a foundational understanding of when OlfPCs establish tissue residence, how long and where they reside within olfactory tissues, and how these cells relate to the peripheral BM PC pool. These data will empower mechanistic improvements to vaccines against upper respiratory infection.",
                "keywords": [],
                "approved": true
            }
        },
        {
            "type": "Grant",
            "id": "15520",
            "attributes": {
                "award_id": "1R13AI188976-01",
                "title": "Respi-DART: Frontiers in drug development against respiratory viruses and emerging viruses",
                "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": 22573,
                        "first_name": "Barbara L.",
                        "last_name": "Mulach",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-11-18",
                "end_date": "2025-10-31",
                "award_amount": 5000,
                "principal_investigator": {
                    "id": 25153,
                    "first_name": "Asuncion",
                    "last_name": "Mejias",
                    "orcid": null,
                    "emails": "",
                    "private_emails": "",
                    "keywords": null,
                    "approved": true,
                    "websites": null,
                    "desired_collaboration": null,
                    "comments": null,
                    "affiliations": [
                        {
                            "id": 265,
                            "ror": "https://ror.org/03czfpz43",
                            "name": "Emory University",
                            "address": "",
                            "city": "",
                            "state": "GA",
                            "zip": "",
                            "country": "United States",
                            "approved": true
                        }
                    ]
                },
                "other_investigators": [
                    {
                        "id": 22933,
                        "first_name": "Raymond Felix",
                        "last_name": "Schinazi",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": [
                            {
                                "id": 265,
                                "ror": "https://ror.org/03czfpz43",
                                "name": "Emory University",
                                "address": "",
                                "city": "",
                                "state": "GA",
                                "zip": "",
                                "country": "United States",
                                "approved": true
                            }
                        ]
                    }
                ],
                "awardee_organization": {
                    "id": 265,
                    "ror": "https://ror.org/03czfpz43",
                    "name": "Emory University",
                    "address": "",
                    "city": "",
                    "state": "GA",
                    "zip": "",
                    "country": "United States",
                    "approved": true
                },
                "abstract": "Abstract/ Project Summary Since 1995, the DART conference series has primarily focused on the latest advancements in antiviral therapy against viruses like HIV, hepatitis B, and hepatitis C. The DART series is continually evolving to tackle emerging challenges that the global community faces. In 2018, the series began a new program called Respi- DART and Emerging Viruses to address respiratory viruses like RSV and influenza that pose a significant threat to human health, particularly in vulnerable groups like infants, the elderly, and people with compromised immune systems. However, nobody knew that COVID-19 would strike the world two years later. The recent coronavirus pandemic has shaken the world and reminded humanity of the importance of continued research and development in this arena. By bringing together world leaders and key opinion holders, Respi-DART 2024 will tackle current challenges in drug development and vaccines and against respiratory and other emerging viruses. Specific Aims: 1. Provide an interactive workshop setting where clinicians, basic scientists, and health care workers can share and discuss the latest medical and scientific developments in the field of therapeutics development against respiratory viruses; 2. Develop a cross-disciplinary program that facilitates the neutral and balanced exchange of scientific knowledge in the areas of biology, chemistry, pharmacology, clinical research and public health as related to topics such as epidemiology and barriers to treatment, vaccine development, and recent clinical advances of small molecules; 3. Enhance the training of young scientists such as graduate students and post- doctoral fellows and provide increased opportunities for underrepresented scientists and healthcare workers through attendee scholarships in a CME-accredited setting. Educational Objectives: 1. Epidemiology, screening, and diagnosis of all respiratory viruses, 2. Drug discovery, development, modeling, and structural biology, 3. Pre-clinical and clinical evaluation of novel therapeutics in development against respiratory viruses such as SARS-CoV-2, influenza virus, RSV, parainfluenza virus, and metapneumovirus, 4. Long COVID-19, 5. Respiratory virus enzymology, host-virus interactions, and virus replication, 6. Immunology and vaccine development, 7. Impact of variant emergence on therapeutics, vaccines, and epidemiology. 8. Drug resistance, vaccine resistance, and viral evolution, 9. Novel assay and animal model development, 10. Viral infections and respiratory diseases in adults and pediatric populations. Design & Methods: Respi-DART 2024 will take place in Los Cabos, Mexico over three days, at a location that is easily accessed by both national and international delegates. The program will include lectures, oral abstract presentations, discussion panels, and a poster session. We believe that Respi-DART 2024 is a much-needed and timely platform for the discussion of the latest developments in this fast-evolving area of global health.",
                "keywords": [],
                "approved": true
            }
        },
        {
            "type": "Grant",
            "id": "15508",
            "attributes": {
                "award_id": "1K23MH136367-01A1",
                "title": "Integrated Cognitive Rehabilitation and Cognitive Behavioral Therapy Intervention for patients with Post-COVID-19 Cognitive Impairment and Depression",
                "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": 26585,
                        "first_name": "Maggie",
                        "last_name": "Sweeney",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-12-01",
                "end_date": "2029-11-30",
                "award_amount": 194157,
                "principal_investigator": {
                    "id": 32053,
                    "first_name": "Jacqueline Helcer",
                    "last_name": "Becker",
                    "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": "Research: The post-acute sequelae of SARS-CoV-2 infection (PASC) continues to pose an unprecedented public health burden, and cognitive impairment (CI) and depression are among the most common symptoms, which profoundly impacts functioning and quality of life. The purpose of this study is to develop and pilot a novel intervention to treat post-COVID CI and depression in patients with PASC by integrating two evidence-based interventions, cognitive rehabilitation (CR) based on Goal Management Training (GMT) and cognitive behavior therapy (CBT), and adapting the intervention specifically for ethnoculturally diverse patients with PASC. The specific aims are to: 1) Develop a manualized intervention for diverse patients with post- COVID CI and depression by adapting and enhancing a GMT CR+CBT approach and 2) Determine the feasibility and preliminary efficacy of the intervention on diverse patients with post- COVID CI and depression. To accomplish these aims, we will employ a mixed methods, design. Qualitative findings from focus groups with patients with PASC will guide the development of the intervention, which will be iteratively refined through additional semi-structured individual interviews and evaluated for feasibility and preliminary impact in a pilot randomized controlled trial (RCT). Candidate: The primary objective of this application is to support Dr. Jacqueline Becker's career development into an independent investigator studying the bidirectional impact of cognitive and mental health on chronic illnesses. Dr. Becker's proposed training activities are: 1) qualitative methodology; 2) psychosocial intervention development; and 3) RCT design, implementation, and evaluation. To achieve these goals, she has assembled a multidisciplinary mentoring team. Dr. Wisnivesky, her primary mentor, is a health services researcher with expertise in PASC, health disparities research, biostatistics, and RCT design and implementation. Her co-mentors include Dr. Lin, a clinician-investigator with expertise in qualitative research and chronic disease self- management in diverse adults, Dr. Feldman, a psychologist and expert in behavioral medicine psychosocial interventions, Dr. Murrough, a world-renowned psychiatrist with expertise in clinical trials for depression, and Dr. Bagiella, an expert in biostatistics and RCT design and evaluation. Environment: The Icahn School of Medicine at Mount Sinai has a strong tradition of outstanding research and the Department of Medicine is ranked 12th nationwide in NIH funding. The Division of General Internal Medicine has a well-established research infrastructure with an exceptionally strong record of successful and well-funded, mentored and independent investigators.",
                "keywords": [],
                "approved": true
            }
        },
        {
            "type": "Grant",
            "id": "15509",
            "attributes": {
                "award_id": "1R01MH138347-01",
                "title": "Understanding Circuit Dysfunction in Post-Acute Sequelae of SARS-CoV-2 Infection",
                "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": 27469,
                        "first_name": "Andrea",
                        "last_name": "Wijtenburg",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-12-01",
                "end_date": "2029-10-31",
                "award_amount": 797157,
                "principal_investigator": {
                    "id": 32054,
                    "first_name": "R Todd",
                    "last_name": "Constable",
                    "orcid": null,
                    "emails": "",
                    "private_emails": "",
                    "keywords": null,
                    "approved": true,
                    "websites": null,
                    "desired_collaboration": null,
                    "comments": null,
                    "affiliations": []
                },
                "other_investigators": [
                    {
                        "id": 7486,
                        "first_name": "SERENA S",
                        "last_name": "SPUDICH",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "awardee_organization": {
                    "id": 452,
                    "ror": "https://ror.org/03v76x132",
                    "name": "Yale University",
                    "address": "",
                    "city": "",
                    "state": "CT",
                    "zip": "",
                    "country": "United States",
                    "approved": true
                },
                "abstract": "Individuals with neuropsychiatric manifestations of post-acute sequelae of SARS-CoV-2 infection (neuro-PASC) report cognitive impairment, new or worsening anxiety and/or depression, altered sleep, autonomic dysfunction, headache, dizziness, post-exertional malaise, and/or neuropathy, among other symptoms. Currently, an estimated 10% of individuals who contract SARS-CoV-2 go onto develop PASC, which represents an enormous disability as well as clinical, social, and economic burden worldwide. Yet, there is a large gap in our understanding of how neuro-PASC alters the function of the brain leading to persistent symptoms and deficits. In response to RFA-NS-23-021, this proposal is aimed at merging two powerful lines of investigation here at Yale to provide a biological basis for the clinical symptoms observed. The first strength is the NeuroCOVID Clinic at Yale, which has seen over 600 patients with neuro-PASC. We then created The COVID Mind Study at Yale under the leadership of Dr. Spudich and have enrolled over 90 participants with neuro-PASC and 60 controls with no PASC. The clinic and study will serve as key sources of subject recruitment. The second strength is the robust and innovative neuroscience and neuroimaging group at Yale under the leadership of Dr. Constable. His group has leveraged the NIH interest in the human connectome and developed methods that link circuit-level (dys)function in the brain to cognitive profiles measured through standardized testing and clinical symptoms. This framework uses a powerful predictive modeling approach and machine learning strategies to localize circuit (dys)function and quantify the contributions of different networks to clinical symptoms and behavior. With both of these strengths, we are ideally positioned to investigate this critical public health issue. Our preliminary data from The COVID Mind Study demonstrates significant cognitive deficits in individuals with neuro-PASC, particularly in language, working memory, declarative memory, non-dominant motor function, and perception compared to COVID negative controls. They also demonstrate greater negative valence issues, including depression, rumination, apathy, anxiety, and perceived stress. We also demonstrated the neuro-PASC group had significantly lower cerebral blood flow in the left superior parietal lobule compared to PASC negative controls using MRI perfusion imaging. To date, there have been no comprehensive assessments of the impact of neuro-PASC on brain function. Using novel methodology developed for this project centered on predictive models for generating reliable associations, we will identify the circuit level brain changes, potential compensatory mechanisms in each cognitive domain, and understand the distinct role of mood alterations in neuro-PASC. We will generate targets for the development of future treatment strategies in neuro-PASC. Our overarching hypothesis is that there are distinct changes in the brain-behavior circuits in individuals with neuro- PASC that will provide key insight into the pathophysiology of this disorder and guide future treatments. The outcome of these studies will have a major impact in how such patients are perceived and managed.",
                "keywords": [],
                "approved": true
            }
        },
        {
            "type": "Grant",
            "id": "15510",
            "attributes": {
                "award_id": "1F31AI189116-01",
                "title": "Studies on the SARS-CoV-2 Main Protease Dimerization Mechanism",
                "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": 27645,
                        "first_name": "Leshawndra Nyrae",
                        "last_name": "Price",
                        "orcid": null,
                        "emails": "",
                        "private_emails": "",
                        "keywords": null,
                        "approved": true,
                        "websites": null,
                        "desired_collaboration": null,
                        "comments": null,
                        "affiliations": []
                    }
                ],
                "start_date": "2024-12-01",
                "end_date": "2027-11-30",
                "award_amount": 38674,
                "principal_investigator": {
                    "id": 32055,
                    "first_name": "Renee",
                    "last_name": "Delgado",
                    "orcid": null,
                    "emails": "",
                    "private_emails": "",
                    "keywords": null,
                    "approved": true,
                    "websites": null,
                    "desired_collaboration": null,
                    "comments": null,
                    "affiliations": []
                },
                "other_investigators": [],
                "awardee_organization": {
                    "id": 748,
                    "ror": "",
                    "name": "UNIVERSITY OF TEXAS HLTH SCIENCE CENTER",
                    "address": "",
                    "city": "",
                    "state": "TX",
                    "zip": "",
                    "country": "United States",
                    "approved": true
                },
                "abstract": "The main protease enzyme (Mpro) of SARS-CoV-2 cleaves the viral polyprotein into functional units responsible for virus replication and pathogenesis. Interestingly, Mpro functions only as a homodimer. However, the structure- function relationship and conformational plasticity of the Mpro dimerization mechanism remains poorly understood. To address these gaps in knowledge, I have developed quantitative luciferase-based reporter assays for Mpro dimerization in living cells and in vitro. I propose first to use these assays to study the Mpro dimerization mechanism through the construction and analysis of a panel structure-guided and evolution- informed mutants, as well as through targeted deep-mutational scanning of dimerization interface residues. Second, I will use my luciferase-based reporter assays to identify candidate dimerization inhibitors. Third, I additionally propose to use my luciferase-based reporter assays to perform studies on the (likely allosteric) mechanism of inhibitor-facilitated dimerization. Altogether, the proposed studies will yield new assays for studying coronavirus protease biology, unique insights into the mechanisms of Mpro dimerization and its allosteric modulation, and novel compounds that can be used as chemical probes of these molecular mechanisms.",
                "keywords": [],
                "approved": true
            }
        }
    ],
    "meta": {
        "pagination": {
            "page": 1383,
            "pages": 1397,
            "count": 13961
        }
    }
}