Department of Health and Human Services

Part 1. Overview Information

Participating Organization(s)

National Institutes of Health (NIH)

Components of Participating Organizations

National Institute on Aging (NIA)

Funding Opportunity Title
Alzheimer's Drug-Development Program (U01 Clinical Trial Optional)
Activity Code

U01 Research Project – Cooperative Agreements

Announcement Type
Reissue of PAR-22-047
Related Notices

See Notices of Special Interest associated with this funding opportunity

  • August 31, 2022- Implementation Changes for Genomic Data Sharing Plans Included with Applications Due on or after January 25, 2023. See Notice NOT-OD-22-198.
  • August 5, 2022- Implementation Details for the NIH Data Management and Sharing Policy. See Notice NOT-OD-22-189.
Funding Opportunity Number (FON)
PAR-24-223
Companion Funding Opportunity
None
Number of Applications

See Section III. 3. Additional Information on Eligibility.

Assistance Listing Number(s)
93.866
Funding Opportunity Purpose

This Notice of Funding Opportunity (NOFO) invites applications proposing pre-clinical and early stage clinical (Phase I) development of novel small-molecule and biologic drug candidates that aim to prevent Alzheimer's disease (AD), slow its progression, or treat its cognitive and behavioral symptoms. Grant recipients will receive funding for therapy development activities, such as the following:

  • Medicinal chemistry
  • Pharmacokinetics (PK)
  • Absorption, Distribution, Metabolism, Excretion, Toxicology (ADMET)
  • Efficacy in animal models
  • Development of biomarkers for target engagement
  • Formulation development chemical synthesis under Good Manufacturing Practices (GMP)  
  • Investigational New Drug (IND) enabling studies
  • Initial Phase I clinical testing

Key Dates

Posted Date
June 25, 2024
Open Date (Earliest Submission Date)
January 03, 2025
Letter of Intent Due Date(s)

30 days before each application due date

The following table includes NIH standard due dates marked with an asterisk.
Application Due Dates Review and Award Cycles
New Renewal / Resubmission / Revision (as allowed) AIDS - New/Renewal/Resubmission/Revision, as allowed Scientific Merit Review Advisory Council Review Earliest Start Date
February 05, 2025 * March 05, 2025 * Not Applicable July 2025 October 2025 December 2025
June 05, 2025 * July 05, 2025 * Not Applicable November 2025 January 2026 April 2026
October 05, 2025 * November 05, 2025 * Not Applicable March 2026 May 2026 July 2026
February 05, 2026 * March 05, 2026 * Not Applicable July 2026 October 2026 December 2026
June 05, 2026 * July 05, 2026 * Not Applicable November 2026 January 2027 April 2027
October 05, 2026 * November 05, 2026 * Not Applicable March 2027 May 2027 July 2027
February 05, 2027 * March 05, 2027 * Not Applicable July 2027 October 2027 December 2027
June 05, 2027 * July 05, 2027 * Not Applicable November 2027 January 2028 April 2028
October 05, 2027 * November 05, 2027 * Not Applicable March 2028 May 2028 July 2028

All applications are due by 5:00 PM local time of applicant organization. 

Applicants are encouraged to apply early to allow adequate time to make any corrections to errors found in the application during the submission process by the due date.

Expiration Date
November 06, 2027
Due Dates for E.O. 12372

Not Applicable

Required Application Instructions

It is critical that applicants follow the instructions in the Research (R) Instructions in the How to Apply - Application Guide, except where instructed to do otherwise (in this NOFO or in a Notice from NIH Guide for Grants and Contracts).

Conformance to all requirements (both in the Application Guide and the NOFO) is required and strictly enforced. Applicants must read and follow all application instructions in the Application Guide as well as any program-specific instructions noted in Section IV. When the program-specific instructions deviate from those in the Application Guide, follow the program-specific instructions.

Applications that do not comply with these instructions may be delayed or not accepted for review.

IMPORTANT: Per NOT-OD-24-086 updated application forms (FORMS-I) will be used for this opportunity. The updated forms are not yet available and will be posted 30 calendar days or more prior to the first application due date. Once posted, you will be able to access the forms using one of the following submission options:

  1. NIH ASSIST
  2. An institutional system-to-system (S2S) solution
  3. Grants.gov Workspace
Table of Contents

Part 2. Full Text of Announcement

Section I. Notice of Funding Opportunity Description

Overview

AD is the most common cause of dementia in older persons and is among the greatest healthcare challenges of the 21st century. The disease currently affects an estimated 6.7 million in the United States; by 2050, this number could rise as high as 14 million. To avert this public health crisis, effective therapies that prevent AD, slow its progression, or treat its cognitive and behavioral symptoms are urgently needed. To catalyze drug development for AD, NIA is reissuing, with this NOFO, the Alzheimer's Drug Development Program (ADDP). The ADDP offers researchers funding for drug development activities that can be conducted in their own laboratories, in collaboration with contract research organizations (CROs) that specialize in various drug development activities including the following:

  • Medicinal chemistry
  • PK
  • ADMET  
  • Formulations development
  • Efficacy in animal models
  • Development of biomarkers for target engagement
  • Chemical synthesis under GMP
  • IND enabling safety-toxicology
  • Initial Phase I clinical testing

Scope

The overarching goal of the ADDP is the development of a broad range of novel drug candidates for AD, including small molecules, natural products, and biologics which will be eligible for Food and Drug Administration (FDA) approval. 

Projects can enter the ADDP at one of the following stages: 

  1. Early Stage - To optimize the agent's potency, drug-like properties, specificity, pharmacological properties, and/or ADMET properties and undergo dose-range finding toxicology and initial IND-enabling safety toxicology, or 
  2. Late Stage - To advance development of candidates through all required IND-enabling toxicology studies, IND document preparation, and initial single dose, single ascending dose, or multiple ascending dose Phase I clinical testing

Applicants are encouraged to contact the NIA Scientific/Research contact listed in Section VII. Agency Contacts of this NOFO regarding the suitability of their projects for the ADDP and to clarify which entry stage is most appropriate for their project.

Stage Specific Entry Criteria

Early Stage Entry Criteria for Small Molecule Projects

Small molecule projects must meet the following requirements prior to entering the Early Stage:

  1. Rigorous data supporting the hypothesis that modulating the putative drug target/affected pathway will produce a desirable outcome for the intended AD indication (i.e., pre-clinical, prodromal/Mild Cognitive Impairment (MCI), early, mid and late stage).
  2. A bioactive compound, in hand, that will serve as a starting point for optimization with proof of identity and purity (typically greater than 95%, as determined by, e.g., Nuclear Magnetic Resonance (NMR), melting point, or Liquid Chromatography/Mass Spectrometry (LC/MS), with no single impurity greater than 0.5%).
  3. In vitro biological activity (typically less than 1 micromolar (μM) in biochemical assays and less than 10 μM in cell-based assays relevant to the drug target), confirmed by repeat dose-response testing, with more than one batch of compound.
  4. Selectivity for the intended target over closely related targets, if desired (and when the target is known).
  5. Availability of primary and secondary in vitro bioactivity assays that can be used or optimized for driving Structure-Activity Relationship (SAR) studies.
  6. Availability of preclinical animal model(s) that can be used to assess in vivo efficacy or target engagement (i.e., measurement of target binding or proximal downstream effects).
  7. Studies using animal models presented to justify the choice of therapeutic target, drug candidate or, to determine efficacy, must be in compliance with NIH guidance on rigor and reproducibility. In particular, investigators proposing animal model studies are expected to follow the general ARRIVE guidelines for rigorous animal research and the best practices guidelines for AD preclinical efficacy studies. These studies should include a power analysis and associated assumptions for the determination of sample size, statistical handling of the data (such as criteria for data inclusion or exclusion), procedures used for blinding and randomization, and whether studies were balanced for sex and were replicated.
  8. Availability of selectivity and counter-screening assays to address potential activity at related targets and other undesirable activities or artifacts.
  9. Demonstration that the proposed secondary bioactivity assays and counter-screening and selectivity assays have sufficient reliability and throughput for their proposed use in the project.
  10. Demonstration of a clear correlation between activity in the primary assay and activity in confirmatory assays and models, sufficient to justify advancement criteria in a testing funnel.
  11. No obvious legal (e.g., intellectual property) constraints to pursuing the proposed chemical scaffold(s) and using the proposed assays and models for research purposes and/or commercial development.

Early Stage Entry Criteria for Biologics Projects

Biologics projects should meet the following qualifications prior to entering the Early Stage:

  1. Applicants should have one or more drug lead(s) from which a candidate can be derived.
  2. The drug lead(s) should be sufficiently characterized so that the parameters to be optimized can be quantitatively specified.
  3. Established preliminary in-vivo efficacy and target engagement data using agent(s) in relevant animal model(s). The agent(s) should show in-vivo efficacy using clinically relevant outcome measures (e.g., biochemical, neuropathological, behavioral, and/or functional, when possible) and in-vivo target engagement (e.g., measurement of target binding or proximal downstream effects) at the clinically intended site of action using sufficient experimental and statistical rigor in a relevant animal in-vivo proof-of-concept model.
  4. Applicants should have pre-existing data demonstrating that the key in vitro and in vivo assays proposed to optimize the leads are suitable for the proposed purpose and available in either the applicant's or collaborator's laboratories.
  5. Availability of animal and/or cell-based model(s) that can be used to assess in vivo efficacy or target engagement (i.e., measurement of target binding or proximal downstream effects).
  6. Studies using animal models presented to justify the choice of therapeutic target, drug candidate or to determine efficacy must be in compliance with NIH guidance on rigor and reproducibility. In particular, investigators proposing animal model studies are expected to follow the general ARRIVE guidelines for rigorous animal research and the best practices guidelines for AD preclinical efficacy studies. These studies should include a power analysis and associated assumptions for the determination of sample size, statistical handling of the data (such as criteria for data inclusion or exclusion), procedures used for blinding and randomization, and whether studies were balanced for sex and were replicated.
  7. No obvious legal (e.g., intellectual property) constraints to pursuing the proposed biologic agent(s) using the proposed assays and models for research purposes and/or commercial development.

Late Stage Entry Criteria for Small Molecule Projects

Small molecule projects should meet the following qualifications prior to entering the Late Stage:

  1. A strong data package linking the putative drug target/affected pathway to the proposed disease indication and supporting the hypothesis that altering the target activity as proposed will produce desirable outcomes for the disease.
  2. Proposed compounds should have in vitro and in vivo biological activity and ADMET properties appropriate for the intended clinical use (i.e., the disease indication, patient population, delivery mode, treatment duration, and treatment regimen) and outcomes.
  3. In vivo study results that include assessments of PK, bioavailability at the relevant site of action, and the pharmacokinetics-pharmacodynamics (PK/PD) relationship.
  4. Rigorous evidence that the agent is a blood-brain-barrier penetrant (unless the agent is proposed to be delivered directly to the Central Nervous System (CNS)).
  5. A data package showing a clear and convincing demonstration of preclinical efficacy. The data package can include in vivo efficacy in an established AD animal model and/or in vivo target engagement.
  6. Studies using animal models presented to justify the choice of therapeutic target, drug candidate or, efficacy must be in compliance with NIH guidance on rigor and reproducibility. In particular, investigators proposing animal model studies are expected to follow the general ARRIVE guidelines for rigorous animal research and the best practices guidelines for AD preclinical efficacy studies. These studies should include a power analysis and associated assumptions for the determination of sample size, statistical handling of the data (such as criteria for data inclusion or exclusion), procedures used for blinding and randomization, and whether studies were balanced for sex and were replicated.
  7. A data package that includes Ames mutagenicity, hERG activity, microsome stability, cytochrome P450 (CYP) inhibition, plasma protein binding, and aqueous solubility.
  8. A data package that includes counter-screening aimed at determining selectivity across a broad panel of closely related and unrelated pharmacological targets (e.g., G protein-coupled receptors, kinases, etc.).
  9. No obvious legal (e.g., intellectual property) constraints to pursuing the proposed small molecule(s) using the proposed assays and models for research purposes and/or commercial development.

Late Stage Entry Criteria for Biologics Projects

Biologics projects should meet the following qualifications prior to entering the Late Stage:

  1. A data package demonstrating completed optimization and final characterization of the candidate, such as structure/identity, selectivity, bioavailability stability, formulation, manufacturability, sufficient purity, clinically intended route of administration, minimal effective dose, optimal effective dose, time and duration of treatment as determined in relevant in vivo assays using clinically relevant functional and/or pathological outcome measures, and/or in vivo target engagement assays.
  2. Proposed compounds should have in vitro and in vivo biological activity and ADMET properties appropriate for the intended clinical use (i.e., the disease indication, patient population, delivery mode, treatment duration, and treatment regimen) and outcomes.
  3. In vivo study results that include assessments of PK, bioavailability at the relevant site of action, and the PK/PD relationship.
  4. Details concerning special formulations, if proposed (i.e., slow release, liposomes, nanoparticles, etc.).
  5. Rigorous evidence that the agent is a blood-brain-barrier penetrant (unless the agent is proposed to be delivered directly to the CNS).
  6. A data package showing a clear and convincing demonstration of efficacy. The data package can include in vivo efficacy in an established AD animal model and/or in vivo target engagement at the clinically intended site of action.
  7. Studies using animal models presented to justify the choice of therapeutic target, drug candidate or, efficacy must be in compliance with NIH guidance on rigor and reproducibility. In particular, investigators proposing animal model studies are expected to follow the general ARRIVE guidelines for rigorous animal research and the best practices guidelines for AD preclinical efficacy studies. These studies should include a power analysis and associated assumptions for the determination of sample size, statistical handling of the data (such as criteria for data inclusion or exclusion), procedures used for blinding and randomization, and whether studies were balanced for sex and were replicated.
  8. A data package that includes Ames mutagenicity, hERG activity, microsome stability, CYP inhibition, plasma protein binding, and aqueous solubility.
  9. No obvious legal (e.g., intellectual property) constraints to pursuing the proposed biologic agent(s) using the proposed assays and models for research purposes and/or commercial development.

Drug Development Activities Supported Through this NOFO

Supported Early Stage Activities for Small Molecule Projects

Examples of Early Stage activities for small molecule projects include, but are not limited to, the following:

  • Establishment of the SAR
  • Testing of analogs in bioactivity assays and animal models
  • Testing of analogs in selectivity and counter-screening assays
  • Assessment of efficacy and/or target engagement. Studies using animal models to measure target engagement and/or efficacy must be in compliance with NIH guidance on rigor and reproducibility. In particular, investigators proposing animal model studies are expected to follow the general ARRIVE guidelines for rigorous animal research and the best practices guidelines for AD preclinical efficacy studies. These studies should include a power analysis and associated assumptions for the determination of sample size, statistical handling of the data (such as criteria for data inclusion or exclusion), procedures used for blinding and randomization, and whether studies were balanced for sex and were replicated
  • Identification of biomarkers for target engagement
  • Testing of analogs in ADMET assays (e.g., microsome stability, CYP induction and inhibition, solubility, permeability in Madin-Darby Canine Kidney (MDCK) or Caco-2 cells, plasma protein binding, brain/plasma ratio, PK in multiple species)
  • Scale-up synthesis
  • Compound stability studies
  • Pre-formulation studies
  • Multiple dose rodent PK testing with PD correlations, if possible
  • Dose-range finding toxicology studies
  • Initial IND-enabling toxicology

Supported Early Stage Activities for Biologics Projects

Examples of Early Stage activities for biologics projects include, but are not limited to, the following:

  • Optimization of leads for improvement in potency, specificity, bioavailability, and suitability for human testing
  • Selection of the best promoter or viral serotype for a gene therapy product
  • Humanization of a mouse monoclonal antibody
  • Characterization of identity and properties (e.g., cell phenotype, aggregation, epitope mapping, post-translational modifications, sequences, viscosity, stability)
  • Assessment of in vitro activities (e.g., affinity, specificity, activity in cells, cellular uptake)
  • Assessment of efficacy and/or target engagement. Studies using animal models to measure target engagement and/or efficacy must be in compliance with NIH guidance on rigor and reproducibility. In particular, investigators proposing animal model studies are expected to follow the general ARRIVE guidelines for rigorous animal research and the best practices guidelines for AD preclinical efficacy studies. These studies should include a power analysis and associated assumptions for the determination of sample size, statistical handling of the data (such as criteria for data inclusion or exclusion), procedures used for blinding and randomization, and whether studies were balanced for sex and were replicated
  • Identification of biomarkers for target engagement
  • Assessment of in vivo pharmacology such as determination of dose range, dosing regimen, duration of treatment
  • Assessment of PK/PD properties
  • Evaluation of metabolism
  • Minimizing a predicted or previously encountered toxicity
  • Optimization of delivery systems and special formulations (i.e., slow release, liposomes, etc.)
  • Optimization to have better suitability for the route of administration
  • Developing a production plan
  • Dose-range finding toxicology studies
  • Initial IND-enabling toxicology

Supported Late Stage Activities for Small Molecule Projects

Examples of Late Stage activities for small molecule projects include, but are not limited to, the following:

  • Chemistry, Manufacturing, and Control (CMC) activities (e.g., purification development, CMC analytical development, formulation development, salt and polymorph screening, scale-up manufacturing, compound stability studies, and GMP manufacturing) for IND-enabling pharmacology/toxicology testing
  • Multiple dose rodent PK testing with PD correlations, if possible
  • Dose-range finding toxicology studies
  • Metabolite identification
  • IND-enabling toxicology, with toxicokinetics, if applicable
  • Pre-IND meeting
  • IND document preparation (the PD/PI will be responsible for the submission of the IND application and scheduling meetings with the Food and Drug Administration (FDA))
  • GMP manufacturing of material for Phase I clinical testing
  • First-in-human Phase I clinical trial (i.e., single dose, single ascending, or multiple ascending study. Clinical trial outcomes may include safety, tolerability, PK and PD, target engagement, and target modulation endpoints)

Supported Late Stage Activities for Biologics Projects

Examples of Late Stage activities for biologics projects include but are not limited to the following:

  • CMC activities (e.g., master and working cell banks development, purification development, CMC analytical development, formulation development, scale-up manufacturing, compound stability studies, and cGMP manufacturing) for IND-enabling pharmacology/toxicology testing
  • PK evaluations in species relevant for toxicology or human dose-prediction
  • Preliminary safety such as safety pharmacology and/or dose-range finding toxicology
  • IND-enabling toxicology, with toxicokinetics, if applicable
  • Tumorigenicity evaluations particularly for gene therapies and cell therapies
  • Immunogenicity evaluations
  • Biodistribution studies
  • Pre-IND meetings
  • IND document preparation
  • GMP manufacturing of material for Phase I clinical testing
  • First in-human Phase I clinical trial (i.e., single dose, single ascending or multiple ascending dose study. Clinical trial outcomes may include safety, tolerability, PK and PD, target engagement, and target modulation endpoints)

Non-Responsiveness Criteria:

The following types of applications will be considered non-responsive to this NOFO and will be administratively withdrawn prior to scientific peer review:

  • Basic research and studies of disease mechanism
  • Studies aimed at mechanisms of drug action
  • Animal model development studies 
  • Stand-alone preclinical efficacy studies in animal models
  • Screening studies to identify hit compounds
  • Projects aimed at anti-amyloid therapies (e.g., beta- and gamma-secretase inhibitors, amyloid aggregation inhibitors, and anti-amyloid immunotherapies)
  • Projects aimed at developing anti-tau immunotherapies
  • Projects that propose to develop generic or off-patent compounds for AD
  • Projects that propose to repurpose or develop new formulations or drug delivery systems for compounds that have previously been approved for another indication by FDA or any other worldwide regulatory agency
  • Projects that propose to develop and/or test combination therapies
  • Development of risk, detection, diagnostic, prognostic, predictive, and prevention biomarkers projects
  • Development of diagnostics and diagnostic devices projects
  • Stand-alone clinical trials
  • Studies directed beyond Phase I clinical testing 

Milestones

Milestone-driven research is used to ensure research is focused on a well-defined goal and achieving that goal with the greatest efficiency. As drug development research is inherently high risk, the use of milestones provides clear indicators of a project's continued success or emergent difficulties. The milestones must provide objective and quantitative success criteria which are recognizable as appropriate endpoints for a specific scientific goal and can be used to monitor the progress made by a research project. The milestones will serve as a basis for go/no-go decision making between NIA program staff and the project research team. Therefore, the application must include a table or list of go/no-go milestones with quantitative success criteria for each year of funding. Prior to funding of an application, NIA program staff will contact the applicant to discuss the proposed milestones and any modifications to the milestones recommended by the review committee or NIA Program staff. A final set of approved milestones will be specified in the Notice of Award. Progress towards achievement of the established milestones will be evaluated by a committee composed of NIA program staff. If warranted, the milestones for future years may be revised based on data and research progress during the preceding year.

Note: If a funded project does not make satisfactory progress toward the agreed upon milestones at any stage during the funding period, future year grant funding may be discontinued. 

Quality and Compliance Requirements

Since one of the goals of the ADDP is to generate drug candidates which will be eligible for FDA approval, adherence to compliance and quality criteria is required.

Intellectual Property

The creation and protection of intellectual property (IP) that will make drug candidates attractive to potential licensing and commercialization partners is an important activity to be undertaken by the PD/PI in consultation with their institutions' technology transfer officials. This program is structured so that the awardee institution retains their assignment of IP and thereby controls the patent prosecution and licensing negotiations for drug candidates developed in this program. It is expected that the awardee institution will take responsibility for patent filings, maintenance, and licensing efforts toward eventual commercialization.

Evaluation of this Program

In carrying out its stewardship of this NOFO, NIA may request information essential to an assessment of the effectiveness of this program from the participants. Participants may be contacted during and after the completion of this award for periodic updates on information helpful in evaluating the impact of the program. In carrying out its stewardship of this NOFO, NIA may request information essential to an assessment of the effectiveness of this program from the participants. Participants may be contacted during and after the completion of this award for periodic updates on information helpful in evaluating the impact of the program. NIA will use this information to determine overall success of the program in developing drugs for AD. 


OutreachPro

NIA is committed to ensuring that clinical research studies have diverse and inclusive representation, particularly of those communities understudied in research. NIA’s National Strategy for Recruitment and Participation in Alzheimer’s and Related Dementias Clinical Research provides an emphasis on the urgent need for more research volunteers and from various populations and it encourages examining  research study populations across multiple factors such as age, race, ethnicity, sex, education, socioeconomic status, geographic location, comorbidities, and cognitive status. To increase participation in AD/ADRD clinical trials, NIA developed a public facing tool, OutreachPro. OutreachPro is an online recruitment materials generator that enables grantees to develop customizable and culturally appropriate materials in multiple languages to raise awareness of, and interest in, AD/ADRD clinical trials. Grantees and research teams are encouraged but not required to use this free online resource to enhance their recruitment and retention plans and related activities for their NIA-funded clinical studies.

Clinical Research Operations Management System

NIA uses a central resource to NIA staff and extramural investigators to facilitate/support the conduct and management of clinical research. NIA Clinical Research Operations & Management System (CROMS) is a comprehensive data management system to support the business functions, management, and oversight responsibilities of NIA grants that support the conduct of clinical research with human subjects. NIA investigators of grants, contracts, and cooperative agreements that are active as of July 1, 2021, including clinical trials funded as pilots, exploratory studies, or other projects through this Consortium, and support human subjects research as defined by the DHS HHS OHRP regulations at 45 CFR 46 will be required to interact with and use existing and future components of CROMS as required by NIA throughout the lifecycle of the grant, as described in NOT-AG-23-017. Data to be submitted to NIA CROMS includes those elements reported in the standard NIH requirement annual progress report (GPS 4.1.15.7). Details regarding the standard operating procedures for CROMS can be found on the NIA CROMS website.

When applicable, all NIA grantees must ensure:

1. The study’s Informed Consent Document (ICD) lists “The National Institutes of Health (NIH) and its authorized representatives” as one of the organizations that may look at or receive copies of information in participants’ study records. According to DHS HHS OHRP 45 CFR 46 §46.116, all ICDs must contain “A statement describing the extent, if any, to which confidentiality of records identifying the participant will be maintained.” If using the NIA informed consent template, please see Section 6: Statement of Confidentiality. 

2. An assigned NIH ClinicalTrials.gov identifier (NCT number) is reported in its respective CROMS study record within three months after assignment, and the reporting of final enrollment data to CROMS is consistent with final enrollment data reported in ClinicalTrials.gov. 

NIA’s Commitment to Inclusion in Research Involving Human Subjects 


NIA is committed to supporting and conducting research on aging that improves the health and well-being of all people. Therefore, NIA will prioritize the advancement of science that represents, in terms of race, ethnicity, sex, age, and comorbidity, the population affected by the condition being studied. Applicants should ensure as applicable that they 1) include proposed planned enrollment tables identifying the population(s) affected by the disease/condition, and 2) address the NIH Inclusion Policies for Research Involving Human Subjects and NIH-designated Populations with Health Disparities, as appropriate; as well as other populations that experience health disparities.

See Section VIII. Other Information for award authorities and regulations.

Investigators proposing NIH-defined clinical trials may refer to the Research Methods Resources website for information about developing statistical methods and study designs.

Section II. Award Information

Funding Instrument

Cooperative Agreement: A financial assistance mechanism used when there will be substantial Federal scientific or programmatic involvement. Substantial involvement means that, after award, NIH scientific or program staff will assist, guide, coordinate, or participate in project activities. See Section VI.2 for additional information about the substantial involvement for this NOFO.

Application Types Allowed
New
Resubmission
Revision

The OER Glossary and the How to Apply Application Guide provide details on these application types. Only those application types listed here are allowed for this NOFO.

Clinical Trial?

Optional: Accepting applications that either propose or do not propose clinical trial(s).

Funds Available and Anticipated Number of Awards

The number of awards is contingent upon NIH appropriations and the submission of a sufficient number of meritorious applications.

Award Budget

Application budgets are limited to $1,500,000 in direct costs per year and need to reflect the actual needs of the proposed project

Award Project Period

For Early Stage projects, the project period is limited to five years. 

For Late Stage projects, the project period is limited to four years.

NIH grants policies as described in the NIH Grants Policy Statement will apply to the applications submitted and awards made from this NOFO.

Section III. Eligibility Information

1. Eligible Applicants

Eligible Organizations

Higher Education Institutions

  • Public/State Controlled Institutions of Higher Education
  • Private Institutions of Higher Education

The following types of Higher Education Institutions are always encouraged to apply for NIH support as Public or Private Institutions of Higher Education:

  • Hispanic-serving Institutions
  • Historically Black Colleges and Universities (HBCUs)
  • Tribally Controlled Colleges and Universities (TCCUs)
  • Alaska Native and Native Hawaiian Serving Institutions
  • Asian American Native American Pacific Islander Serving Institutions (AANAPISIs)

Nonprofits Other Than Institutions of Higher Education

  • Nonprofits with 501(c)(3) IRS Status (Other than Institutions of Higher Education)
  • Nonprofits without 501(c)(3) IRS Status (Other than Institutions of Higher Education)

For-Profit Organizations

  • Small Businesses
  • For-Profit Organizations (Other than Small Businesses)

Local Governments

  • State Governments
  • County Governments
  • City or Township Governments
  • Special District Governments
  • Indian/Native American Tribal Governments (Federally Recognized)
  • Indian/Native American Tribal Governments (Other than Federally Recognized).

Federal Governments

  • Eligible Agencies of the Federal Government
  • U.S. Territory or Possession

Other

  • Independent School Districts
  • Public Housing Authorities/Indian Housing Authorities
  • Native American Tribal Organizations (other than Federally recognized tribal governments)
  • Faith-based or Community-based Organizations
  • Regional Organizations
Foreign Organizations

Non-domestic (non-U.S.) Entities (Foreign Organizations) are not eligible to apply.

Non-domestic (non-U.S.) components of U.S. Organizations are eligible to apply.

Foreign components, as defined in the NIH Grants Policy Statement, are allowed.

Required Registrations

Applicant Organizations

Applicant organizations must complete and maintain the following registrations as described in the How to Apply- Application Guide to be eligible to apply for or receive an award. All registrations must be completed prior to the application being submitted. Registration can take 6 weeks or more, so applicants should begin the registration process as soon as possible. Failure to complete registrations in advance of a due date is not a valid reason for a late submission, please reference the NIH Grants Policy Statement Section 2.3.9.2 Electronically Submitted Applications for additional information.

  • System for Award Management (SAM) – Applicants must complete and maintain an active registration, which requires renewal at least annually. The renewal process may require as much time as the initial registration. SAM registration includes the assignment of a Commercial and Government Entity (CAGE) Code for domestic organizations which have not already been assigned a CAGE Code.
    • NATO Commercial and Government Entity (NCAGE) Code – Foreign organizations must obtain an NCAGE code (in lieu of a CAGE code) in order to register in SAM.
    • Unique Entity Identifier (UEI) - A UEI is issued as part of the SAM.gov registration process. The same UEI must be used for all registrations, as well as on the grant application.
  • eRA Commons - Once the unique organization identifier is established, organizations can register with eRA Commons in tandem with completing their Grants.gov registrations; all registrations must be in place by time of submission. eRA Commons requires organizations to identify at least one Signing Official (SO) and at least one Program Director/Principal Investigator (PD/PI) account in order to submit an application.
  • Grants.gov – Applicants must have an active SAM registration in order to complete the Grants.gov registration.

Program Directors/Principal Investigators (PD(s)/PI(s))

All PD(s)/PI(s) must have an eRA Commons account.  PD(s)/PI(s) should work with their organizational officials to either create a new account or to affiliate their existing account with the applicant organization in eRA Commons. If the PD/PI is also the organizational Signing Official, they must have two distinct eRA Commons accounts, one for each role. Obtaining an eRA Commons account can take up to 2 weeks.

Eligible Individuals (Program Director/Principal Investigator)

Any individual(s) with the skills, knowledge, and resources necessary to carry out the proposed research as the Program Director(s)/Principal Investigator(s) (PD(s)/PI(s)) is invited to work with their organization to develop an application for support. Individuals from diverse backgrounds, including underrepresented racial and ethnic groups, individuals with disabilities, and women are always encouraged to apply for NIH support. See, Reminder: Notice of NIH's Encouragement of Applications Supporting Individuals from Underrepresented Ethnic and Racial Groups as well as Individuals with Disabilities, NOT-OD-22-019.

For institutions/organizations proposing multiple PDs/PIs, visit the Multiple Program Director/Principal Investigator Policy and submission details in the Senior/Key Person Profile (Expanded) Component of the How to Apply-Application Guide.

2. Cost Sharing

This NOFO does not require cost sharing as defined in the NIH Grants Policy Statement Section 1.2 Definition of Terms.

3. Additional Information on Eligibility

Number of Applications

Applicant organizations may submit more than one application, provided that each application is scientifically distinct.

The NIH will not accept duplicate or highly overlapping applications under review at the same time, per NIH Grants Policy Statement Section 2.3.7.4 Submission of Resubmission Application. This means that the NIH will not accept:

  • A new (A0) application that is submitted before issuance of the summary statement from the review of an overlapping new (A0) or resubmission (A1) application.
  • A resubmission (A1) application that is submitted before issuance of the summary statement from the review of the previous new (A0) application.
  • An application that has substantial overlap with another application pending appeal of initial peer review (see NIH Grants Policy Statement 2.3.9.4 Similar, Essentially Identical, or Identical Applications).

Section IV. Application and Submission Information

1. Requesting an Application Package

The application forms package specific to this opportunity must be accessed through ASSIST, Grants.gov Workspace or an institutional system-to-system solution. Links to apply using ASSIST or Grants.gov Workspace are available in Part 1 of this NOFO. See your administrative office for instructions if you plan to use an institutional system-to-system solution.

2. Content and Form of Application Submission

It is critical that applicants follow the instructions in the Research (R) Instructions in the How to Apply - Application Guide except where instructed in this notice of funding opportunity to do otherwise. Conformance to the requirements in the Application Guide is required and strictly enforced. Applications that are out of compliance with these instructions may be delayed or not accepted for review.

Letter of Intent

Although a letter of intent is not required, is not binding, and does not enter into the review of a subsequent application, the information that it contains allows IC staff to estimate the potential review workload and plan the review.

By the date listed in Part 1. Overview Information, prospective applicants are asked to submit a letter of intent that includes the following information:

  • Descriptive title of proposed activity
  • Name(s), address(es), and telephone number(s) of the PD(s)/PI(s)
  • Names of other key personnel
  • Participating institution(s)
  • Number and title of this funding opportunity

The letter of intent should be sent to:

Lorenzo M. Refolo, Ph.D.
National Institute on Aging (NIA)
Telephone: 301-594-7576
Email: refolol@mail.nih.gov

Page Limitations

All page limitations described in the How to Apply- Application Guide and the Table of Page Limits must be followed.

Instructions for Application Submission

The following section supplements the instructions found in the How to Apply- Application Guide and should be used for preparing an application to this NOFO.

SF424(R&R) Cover

All instructions in the How to Apply- Application Guide must be followed.

SF424(R&R) Project/Performance Site Locations

All instructions in the How to Apply- Application Guide must be followed.

SF424(R&R) Other Project Information

All instructions in the How to Apply- Application Guide must be followed.

SF424(R&R) Senior/Key Person Profile

All instructions in the How to Apply- Application Guide must be followed.

R&R or Modular Budget

All instructions in the How to Apply- Application Guide must be followed.

R&R Subaward Budget

All instructions in the How to Apply-Application Guide must be followed.

PHS 398 Cover Page Supplement

All instructions in the How to Apply- Application Guide must be followed.

PHS 398 Research Plan

All instructions in the How to Apply- Application Guide must be followed, with the following additional instructions:

Specific Aims: The Specific Aims section should include aims delineated either for the Early or Late Stages of the project. For proposed Phase I clinical trials, define the aims of the study (e.g., safety, tolerability, PK/PD, target engagement and target modulation, dose-range).

Research Strategy:

Note: Applicants proposing human subjects and/or clinical trials must use the PHS Human Subjects and Clinical Trials Information Form to capture detailed study information. You may use the research strategy to discuss the overall approach but do not duplicate information collected in the PHS Human Subjects and Clinical Trials Information Form.

The Research Strategy section must include the following six sections. The sub-bullets provide details regarding what should be addressed in each section.  

  1. Clinical Impact (Significance):
    • Applicants should include a brief statement of the therapeutic hypothesis that includes: the projected patient reduction of symptoms, slowing disease progression, side effects, dose administration and regimen, and sustainability of effect.
    • Provide a Target Product Profile (TPP) in a table based on an FDA template that summarizes the minimal/ideal profile of the final marketed product and shows the ultimate goals of the proposed drug development effort, such as disease indication, patient population, delivery mode, treatment duration, treatment regimen, and standards for clinical efficacy.
    • Applicants requesting funding for a clinical trial should include a clinical protocol and management plan.
    • Indicate the target clinical population and specific stage of disease in which the treatment would be most efficacious.
    • Describe possible clinical trial endpoints. Indicate if biomarkers are available in animal models and humans to detect if the drug engages the target.
    • Describe the group clinical expertise used to determine the goals of the drug development program and the clinical trial.
    • Discuss how the proposed project relates to therapeutic development efforts underway in academia and industry.
       
  2. Biological Rationale and Profile of the Drug Candidate (Significance):
    • Applicants should summarize the evidence that validates the drug target from cellular or animal models and clinical studies, provide a brief summary of the rationale for the selection of the target, the level of agreement in the field regarding the target's role in disease pathogenesis and clinical relevance of the target, including the optimal disease stage (e.g., asymptomatic, MCI, mild, moderate, or severe AD) to pharmacologically engage the target.
    • Provide the evidence that altering target activity as proposed will give desirable outcomes for the proposed stage of AD.
    • Applicants should discuss the disease-relevance of in vitro or in vivo models that are proposed or that have been used and whether the endpoints measured, and levels of activity observed are likely to be clinically relevant.
    • Applicants should discuss plans to incorporate any translatable, clinically relevant biomarkers into their preclinical development program.
    • Applicants should discuss how target engagement and/or pharmacodynamic markers will be used to detect activity at the putative target in preclinical models and in patients, if available or proposed for development.
    • All studies using animal models presented to justify the choice of therapeutic target, drug candidate or, to measure efficacy must be in compliance with NIH guidance on rigor and reproducibility. In particular, investigators proposing animal model studies are expected to follow the general ARRIVE guidelines for rigorous animal research and the best practices guidelines for AD preclinical efficacy studies.These studies should include a power analysis and associated assumptions for the determination of sample size, statistical handling of the data (such as criteria for data inclusion or exclusion), procedures used for blinding and randomization, and whether studies were balanced for sex and were replicated.
    • Provide a Drug Candidate Profile Table that summarizes the drug-like properties, pharmacological and ADMET activities of the drug candidate proposed for optimization or as the development candidate. Note any potential liabilities.
    • For Early Stage projects, show that the drug candidate proposed as the starting point for optimization alters the activity of the putative target as intended and/or produces desired outcomes in disease models, with sufficient detail to allow reviewers to evaluate the rigor of the experimental design. Explain the choice of models, assays, and endpoints for these studies.
    • For Late Stage projects, show that the proposed development candidate has clinically relevant in vivo activity, when delivered by the clinically intended route of administration, at exposure levels that can likely be achieved clinically with the proposed human dosing regimen.
    • For Late Stage projects, show the data that demonstrate the relationship between drug candidate exposure and activity and explain how these data support the clinical dosing regimen proposed in the TPP.
       
  3. Testing Strategy (Approach):
    • Specify whether the project is proposed for entry at the Early or Late Stage.
    • For small molecule projects, present evidence that the drug candidate(s) meet the stage specific entry criteria outlined in Section I. Notice of Funding Opportunity Description of this NOFO for the proposed project stage (Early or Late).
    • For biologics projects, present evidence that the drug candidate(s) meet the stage specific entry criteria outlined in Section I. Notice of Funding Opportunity Description of this NOFO for the proposed project stage (Early or Late).
    • Explain the rationale for the choice of assays, assay design, and advancement criteria, and clarify how these relate to the desired drug properties presented in the TPP.
    • Show assay validation data or present plans to optimize and validate assays.
    • Include a figure showing the Testing Funnel that shows how proposed assays will be ordered and grouped into testing tiers.
    • Include a table of proposed activities that provides the following information: activity (e.g., medicinal chemistry, optimization of biologic candidate, assays, PK, etc.), throughput for testing chemical analogs or biologic derivatives, and advancement criteria through the testing funnel.
    • Clearly indicate which activities will be conducted by the PD/PI and associated personnel and which activities will be conducted by contractors. Include experimental designs and justification for all studies that will be conducted by the PD/PI and associated personnel. Activities that will be conducted by CROs should be specifically described in terms that will include expected deliverables. Use of foreign CROs must be justified.
    • Include a Gantt chart that lays out each step in the critical path of the project (e.g., optimization of drug-like properties, optimization of bioactivity, pharmacology profiling, toxicology profiling, efficacy in animal models, GMP synthesis, formulations development, and IND-enabling studies, Phase I clinical trial).
       
  4. Milestones:
    • Because drug discovery and development are inherently high risk, it is expected that there may be significant attrition as projects progress. Therefore, the application must include a Table of go/no-go milestones with quantitative success criteria for each year of funding.
       
  5. Innovation
    • If the drug candidate targets the same molecule, pathway, or cellular process that has previously been tested in clinical trials for AD, explain why the proposed approach would be expected to provide a benefit over previously tested approaches.
    • If similar drugs have been tested in clinical trials for AD, explain why the proposed drug candidate would be expected to give significantly better clinical outcomes.
    • If the drug candidate is an improvement over an earlier generation of drug(s) that has not been marketed, discuss the advantages of the proposed new drug candidate over those previously tested. Include results from previous clinical trials with related agents.
    • Comment on the novelty of proposed approach, target, pathway, assays, or models.
       
  6. Intellectual Property (IP)
    • Applicants are encouraged to prepare this section in consultation with their institutions' technology transfer officials.
    • Applicants must describe any constraints of which they are aware that could impede their use of compounds, assays, or models for research purposes and/or commercial development (e.g., certain restrictions under transfer or sharing agreements, applicants' previous or present intellectual property filings and publications, compounds with similar structures that are under patent and/or on the market, etc.) and how these issues would be addressed. If the applicant's institution has filed pertinent patents, the applicant must indicate filing dates, the type of patent, and application status.
    • For a multiple-PD/PI, multiple-institution application, applicants must describe the infrastructure of each institution for bringing the technologies to practical application and for coordinating these efforts (e.g., licensing, managing intellectual property) among the institutions consistent with achieving the goals of the program. Applicants must clarify how IP will be shared or otherwise managed if there are multiple PDs/PIs and institutions involved.


Information Regarding Clinical Trials

  • Applicants are strongly advised to discuss plans with NIH program staff prior to submitting their application to determine whether a clinical trial is feasible within the proposed funding timeframe.
  • Applicants must submit supporting trial and regulatory documents to NIH for administrative review prior to the commencement of the trial.

Eligibility Criteria for Research Involving Human Subjects

Without duplicating information in the Human Subjects and Clinical Trails form, applications in response to this NOFO must provide rationale that eligibility criteria encourage diversity across multiple factors such as age, race, ethnicity, sex, gender, education, socioeconomic status, geographic location, comorbidities, cognitive status, and other populations experiencing health disparities to diversify and increase participation in clinical trials. The goal is for clinical trial participants to adequately reflect the diversity of the real-world population, so that researchers can determine whether the variables being studied affect women or members of any racial and ethnic population group in accordance with the NIH Revitalization Act of 1993.

Study teams must demonstrate that they have considered inclusive practices including proposed planned enrollment tables representative of the population affected by the disease/condition. Where applicable, study teams should also demonstrate that they have critically evaluated which eligibility criteria from an earlier phase trial should be carried forward into a later phase trial. The eligibility criteria section must:

  • Describe how the study results generalize to the wider patient population with this disease/condition.
  • Explain how the rationale for selected eligibility criteria justify the level of restriction in the study compared to clinical practice.
  • Provide evidence that the eligibility criteria support the proposed research and encourage diversity across multiple sociodemographic factors.

Recruitment and Retention Plan for Research Involving Human Subjects

Without duplicating information in the Human Subjects and Clinical Trails form, applications in response to this NOFO must propose innovative and proactive recruitment strategies for involving historically underrepresented populations to ensure more diverse and inclusive representation as applicable and justified by the scientific goals. An emphasis on diversity across multiple factors such as age, race, ethnicity, sex, gender, education, socioeconomic status, geographic locations, comorbidities, cognitive status, and other populations experiencing health disparities is encouraged. Recruitment and Retention Plans should demonstrate an understanding of the participant burden involved in research participation and strategies for minimizing this burden, as well as leveraging community partners and outreach efforts. The Recruitment and Retention Plan must:

  • Describe potential barriers to participation and plans to minimize these barriers.
  • Describe a detailed plan for the recruitment of underrepresented populations and a plan to leverage existing relationships with and/or conduct outreach to diverse community groups.
  • Describe staff training to be sensitive to cultural, racial, and ethnic differences. 

Resource Sharing Plan: Individuals are required to comply with the instructions for the Resource Sharing Plans as provided in the How to Apply- Application Guide.

Other Plan(s): Note: Effective for due dates on or after January 25, 2023, the Data Management and Sharing Plan will be attached in the Other Plan(s) attachment in FORMS-H application forms packages.

All instructions in the How to Apply-Application Guide must be followed, with the following additional instructions:

  • All applicants planning research (funded or conducted in whole or in part by NIH) that results in the generation of scientific data are required to comply with the instructions for the Data Management and Sharing Plan. All applications, regardless of the amount of direct costs requested for any one year, must address a Data Management and Sharing Plan.
  • If patent protection is being sought, investigators should explain how data will be shared after filing for patent protection to allow for both further research and the development of commercial products to advance forward, consistent with achieving the goals of the program.

Appendix: Only limited Appendix materials are allowed. Follow all instructions for the Appendix as described in the How to Apply- Application Guide.

  • No publications or other material, with the exception of blank questionnaires or blank surveys, may be included in the Appendix.

PHS Human Subjects and Clinical Trials Information

When involving human subjects research, clinical research, and/or NIH-defined clinical trials (and when applicable, clinical trials research experience) follow all instructions for the PHS Human Subjects and Clinical Trials Information form in the How to Apply- Application Guide, with the following additional instructions:

If you answered “Yes” to the question “Are Human Subjects Involved?” on the R&R Other Project Information form, you must include at least one human subjects study record using the Study Record: PHS Human Subjects and Clinical Trials Information form or Delayed Onset Study record.

Study Record: PHS Human Subjects and Clinical Trials Information

All instructions in the How to Apply- Application Guide must be followed.

Section 4 - Protocol Synopsis

4.1. Study Design

4.1a. Detailed Description

Include determination of dose levels.

4.1c. Interventions

For "Intervention Description", include route of administration.

4.2. Outcome Measures

At least one outcome measure should include PK assessments, with attention to demonstration of CNS penetration (if appropriate) and target engagement or modulation.

Delayed Onset Study

Note: Delayed onset does NOT apply to a study that can be described but will not start immediately (i.e., delayed start). All instructions in the How to Apply- Application Guide must be followed.

PHS Assignment Request Form

All instructions in the How to Apply- Application Guide must be followed.

3. Unique Entity Identifier and System for Award Management (SAM)

See Part 2. Section III.1 for information regarding the requirement for obtaining a unique entity identifier and for completing and maintaining active registrations in System for Award Management (SAM), NATO Commercial and Government Entity (NCAGE) Code (if applicable), eRA Commons, and Grants.gov

4. Submission Dates and Times

Part I. contains information about Key Dates and times. Applicants are encouraged to submit applications before the due date to ensure they have time to make any application corrections that might be necessary for successful submission. When a submission date falls on a weekend or Federal holiday, the application deadline is automatically extended to the next business day.

Organizations must submit applications to Grants.gov (the online portal to find and apply for grants across all Federal agencies). Applicants must then complete the submission process by tracking the status of the application in the eRA Commons, NIH’s electronic system for grants administration. NIH and Grants.gov systems check the application against many of the application instructions upon submission. Errors must be corrected and a changed/corrected application must be submitted to Grants.gov on or before the application due date and time.  If a Changed/Corrected application is submitted after the deadline, the application will be considered late. Applications that miss the due date and time are subjected to the NIH Grants Policy Statement Section 2.3.9.2 Electronically Submitted Applications.

Applicants are responsible for viewing their application before the due date in the eRA Commons to ensure accurate and successful submission.

Information on the submission process and a definition of on-time submission are provided in the How to Apply-Application Guide.

5. Intergovernmental Review (E.O. 12372)

This initiative is not subject to intergovernmental review.

6. Funding Restrictions

All NIH awards are subject to the terms and conditions, cost principles, and other considerations described in the NIH Grants Policy Statement.

Pre-award costs are allowable only as described in the NIH Grants Policy Statement Section 7.9.1 Selected Items of Cost.

7. Other Submission Requirements and Information

Applications must be submitted electronically following the instructions described in the How to Apply Application Guide. Paper applications will not be accepted.

Applicants must complete all required registrations before the application due date. Section III. Eligibility Information contains information about registration.

For assistance with your electronic application or for more information on the electronic submission process, visit How to Apply – Application Guide. If you encounter a system issue beyond your control that threatens your ability to complete the submission process on-time, you must follow the Dealing with System Issues guidance. For assistance with application submission, contact the Application Submission Contacts in Section VII.

Important reminders:

All PD(s)/PI(s) must include their eRA Commons ID in the Credential field of the Senior/Key Person Profile form. Failure to register in the Commons and to include a valid PD/PI Commons ID in the credential field will prevent the successful submission of an electronic application to NIH. See Section III of this NOFO for information on registration requirements.

The applicant organization must ensure that the unique entity identifier provided on the application is the same identifier used in the organization’s profile in the eRA Commons and for the System for Award Management. Additional information may be found in the How to Apply Application Guide.

See more tips for avoiding common errors.

Upon receipt, applications will be evaluated for completeness and compliance with application instructions by the Center for Scientific Review and responsiveness by NIA, NIH. Applications that are incomplete, non-compliant and/or nonresponsive will not be reviewed.

 

Post Submission Materials

Applicants are required to follow the instructions for post-submission materials, as described in the policy

Section V. Application Review Information

1. Criteria

Only the review criteria described below will be considered in the review process. Applications submitted to the NIH in support of the NIH mission are evaluated for scientific and technical merit through the NIH peer review system.

For this particular NOFO, note the following:

  • Projects should not be penalized if the mechanism of action of the compound is unknown. While this may add to the risk, the increased risk may be counterbalanced by increased novelty.
  • Evaluation of the approach should focus on the biological rationale, the rigor and transparency of supporting preclinical data, the potential for identifying a compound/biologic with drug-like properties, potential patient benefit, competitive landscape (novelty), and strengths/weaknesses of studies to be conducted by the PD/PI.
  • Work to be conducted by CROs, specifically expected deliverables for each type of CRO, is subject to review.
  • Applications that propose entry at the Late Stage, but that reviewers consider better suited for the Early Stage or vice versa should nevertheless be evaluated and scored based on the entry criteria and expectations for the proposed entry stage. Reviewers can note in their comments that the project may be more appropriate for entry at a different stage.
  • All studies using animal models presented to justify the choice of therapeutic target or drug candidate must be in compliance with NIH guidance on rigor and reproducibility. In particular, investigators proposing animal model studies are expected to follow the general ARRIVE guidelines for rigorous animal research and the best practices guidelines for AD preclinical efficacy studies. These studies should include a power analysis and associated assumptions for the determination of sample size, statistical handling of the data (such as criteria for data inclusion or exclusion), procedures used for blinding and randomization, and whether studies were balanced for sex and were replicated.
Overall Impact

Reviewers will provide an overall impact score to reflect their assessment of the likelihood for the project to exert a sustained, powerful influence on the research field(s) involved, in consideration of the following scored review criteria and additional review criteria (as applicable for the project proposed). An application does not need to be strong in all categories to be judged likely to have a major scientific impact.

Scored Review Criteria

Reviewers will evaluate Factors 1, 2 and 3 in the determination of scientific merit, and in providing an overall impact score. In addition, Factors 1 and 2 will each receive a separate criterion score. 

 

Significance

  • Evaluate the importance of the proposed research in the context of current scientific challenges and opportunities, either for advancing knowledge within the field, or more broadly. Assess whether the application addresses an important gap in knowledge in the field, would solve a critical problem, or create a valuable conceptual or technical advance.
  • Evaluate the rationale for undertaking the study, the rigor of the scientific background for the work (e.g., prior literature and/or preliminary data) and whether the scientific background justifies the proposed study.

Innovation

  • Evaluate the extent to which innovation influences the importance of undertaking the proposed research. Note that while technical or conceptual innovation can influence the importance of the proposed research, a project that is not applying novel concepts or approaches may be of critical importance for the field.
  • Evaluate whether the proposed work applies novel concepts, methods or technologies or uses existing concepts, methods, technologies in novel ways, to enhance the overall impact of the project.

Specific to this NOFO:

  • Evaluate whether the proposed drug candidate offers a significant advantage over other treatments under development (in any drug class).
  • Evaluate the likelihood that completion of the research objectives will lead to an AD therapy (i.e., is there a clear path into the clinic).
  • Evaluate the strength of the data supporting the choice of therapeutic target and compound/biologic for the proposed stage of disease.
 

Approach

  • Evaluate the scientific quality of the proposed work. Evaluate the likelihood that compelling, reproducible findings will result (rigor) and assess whether the proposed studies can be done well and within the timeframes proposed (feasibility).

Rigor:

  • Evaluate the potential to produce unbiased, reproducible, robust data.
  • Evaluate the rigor of experimental design and whether appropriate controls are in place.
  • Evaluate whether the sample size is sufficient and well-justified.
  • Assess the quality of the plans for analysis, interpretation, and reporting of results.
  • Evaluate whether the investigators presented adequate plans to address relevant biological variables, such as sex or age, in the design, analysis, and reporting.
  • For applications involving human subjects or vertebrate animals, also evaluate:
    • the rigor of the intervention or study manipulation (if applicable to the study design).
    • whether outcome variables are justified.
    • whether the results will be generalizable or, in the case of a rare disease/special group, relevant to the particular subgroup.
    • whether the sample is appropriate and sufficiently diverse to address the proposed question(s).
  • For applications involving human subjects, including clinical trials, assess the adequacy of inclusion plans as appropriate for the scientific goals of the research. Considerations of appropriateness may include disease/condition/behavior incidence, prevalence, or population burden, population representation, and/or current state of the science.

Feasibility:

  • Evaluate whether the proposed approach is sound and achievable, including plans to address problems or new challenges that emerge in the work. For proposed studies in which feasibility may be less certain, evaluate whether the uncertainty is balanced by the potential for major advances.
  • For applications involving human subjects, including clinical trials, evaluate the adequacy and feasibility of the plan to recruit and retain an appropriately diverse population of participants. Additionally, evaluate the likelihood of successfully achieving the proposed enrollment based on age, racial, ethnic, and sex/gender categories.
  • For clinical trial applications, evaluate whether the study timeline and milestones are feasible.

Specific to this NOFO:

Eligibility Criteria for Research Involving Human Subjects:

  • Evaluate how well the eligibility criteria fosters inclusive practices for the population affected by the disease/condition.
  •  Assess to what extent is justification provided for eligibility restrictions.
  •  Evaluate how well barriers to participation are addressed.
  •  Assess to what extent is evidence provided that the eligibility criteria support the proposed research and encourage diversity across multiple sociodemographic factors.

Recruitment and Retention Plan for Research Involving Human Subjects:

  • Evaluate how well the recruitment and retention plan demonstrates efforts to engage underrepresented populations in the clinical trial, as applicable and justified by the scientific goals.
  •  Assess to what extent will the efforts described to increase community engagement reduce identified barriers and sustain the engagement of underrepresented populations.
  •  Evaluate how well are plans to train staff to be sensitive to cultural, racial, and ethnic differences are described.
 

Investigator(s)

Evaluate whether the investigator(s) have demonstrated background, training, and expertise, as appropriate for their career stage, to conduct the proposed work. For Multiple Principal Investigator (MPI) applications, assess the quality of the leadership plan to facilitate coordination and collaboration.

Environment

Evaluate whether the institutional resources are appropriate to ensure the successful execution of the proposed work.

Additional Review Criteria

As applicable for the project proposed, reviewers will consider the following additional items while determining scientific and technical merit, but will not give criterion scores for these items, and should consider them in providing an overall impact score.

 

For research that involves human subjects but does not involve one of the categories of research that are exempt under 45 CFR Part 46, evaluate the justification for involvement of human subjects and the proposed protections from research risk relating to their participation according to the following five review criteria: 1) risk to subjects; 2) adequacy of protection against risks; 3) potential benefits to the subjects and others; 4) importance of the knowledge to be gained; and 5) data and safety monitoring for clinical trials.

For research that involves human subjects and meets the criteria for one or more of the categories of research that are exempt under 45 CFR Part 46, evaluate: 1) the justification for the exemption; 2) human subjects involvement and characteristics; and 3) sources of materials. For additional information on review of the Human Subjects section, please refer to the Guidelines for the Review of Human Subjects.

 

When the proposed research includes Vertebrate Animals, evaluate the involvement of live vertebrate animals according to the following criteria: (1) description of proposed procedures involving animals, including species, strains, ages, sex, and total number to be used; (2) justifications for the use of animals versus alternative models and for the appropriateness of the species proposed; (3) interventions to minimize discomfort, distress, pain and injury; and (4) justification for euthanasia method if NOT consistent with the AVMA Guidelines for the Euthanasia of Animals. For additional information on review of the Vertebrate Animals section, please refer to the Worksheet for Review of the Vertebrate Animals Section.

 

When the proposed research includes Biohazards, evaluate whether specific materials or procedures that will be used are significantly hazardous to research personnel and/or the environment, and whether adequate protection is proposed.

 

As applicable, evaluate the full application as now presented.

 

As applicable, evaluate the progress made in the last funding period.

 

As applicable, evaluate the appropriateness of the proposed expansion of the scope of the project.

 

Assess whether the proposed project milestones and associated quantitative success criteria are adequate and feasible.

Additional Review Considerations

As applicable for the project proposed, reviewers will consider each of the following items, but will not give scores for these items, and should not consider them in providing an overall impact score.

 

For projects involving key biological and/or chemical resources, evaluate the brief plans proposed for identifying and ensuring the validity of those resources.

Not Applicable

 

Evaluate whether the budget and the requested period of support are fully justified and reasonable in relation to the proposed research.

 
  • Evaluate whether the applicant demonstrated that there are no constraints, of which they are aware, that could impede their use of drug candidates including small molecules or biologics, assays, or models for research purposes and/or commercial development.
  • Evaluate whether the applicant demonstrated that the proposed drug candidate is unlikely to be blocked or impeded by intellectual property constraints.
  • Evaluate the applicant described pertinent patent applications that have been filed, including the filing date(s), type of patent and application status.
  • If multiple institutions are proposed, evaluate how intellectual property will be shared or otherwise managed to avoid encumbering the IP, consistent with achieving the goals of the program. 

2. Review and Selection Process

Applications will be evaluated for scientific and technical merit by (an) appropriate Scientific Review Group(s) convened by National Institute on Aging (NIA), in accordance with NIH peer review policy and procedures, using the stated review criteria. Assignment to a Scientific Review Group will be shown in the eRA Commons.

As part of the scientific peer review, all applications will receive a written critique.

Applications may undergo a selection process in which only those applications deemed to have the highest scientific and technical merit (generally the top half of applications under review) will be discussed and assigned an overall impact score.

Applications will be assigned on the basis of established PHS referral guidelines to the appropriate NIH Institute or Center. Applications will compete for available funds with all other recommended applications. Following initial peer review, recommended applications will receive a second level of review by the appropriate national Advisory Council or Board. The following will be considered in making funding decisions:

  • Scientific and technical merit of the proposed project as determined by scientific peer review.
  • Availability of funds.
  • Relevance of the proposed project to program priorities.

3. Anticipated Announcement and Award Dates

After the peer review of the application is completed, the PD/PI will be able to access his or her Summary Statement (written critique) via the eRA Commons. Refer to Part 1 for dates for peer review, advisory council review, and earliest start date.

Information regarding the disposition of applications is available in the NIH Grants Policy Statement Section 2.4.4 Disposition of Applications.

Section VI. Award Administration Information

1. Award Notices

If the application is under consideration for funding, NIH will request "just-in-time" information from the applicant as described in the NIH Grants Policy Statement.

A formal notification in the form of a Notice of Award (NoA) will be provided to the applicant organization for successful applications. The NoA signed by the grants management officer is the authorizing document and will be sent via email to the recipient's business official.

Recipients must comply with any funding restrictions described in Section IV.6. Funding Restrictions. Selection of an application for award is not an authorization to begin performance. Any costs incurred before receipt of the NoA are at the recipient's risk. These costs may be reimbursed only to the extent considered allowable pre-award costs.

Any application awarded in response to this NOFO will be subject to terms and conditions found on the Award Conditions and Information for NIH Grants website.  This includes any recent legislation and policy applicable to awards that is highlighted on this website.

Individual awards are based on the application submitted to, and as approved by, the NIH and are subject to the IC-specific terms and conditions identified in the NoA.

ClinicalTrials.gov: If an award provides for one or more clinical trials. By law (Title VIII, Section 801 of Public Law 110-85), the "responsible party" must register and submit results information for certain “applicable clinical trials” on the ClinicalTrials.gov Protocol Registration and Results System Information Website (https://register.clinicaltrials.gov). NIH expects registration and results reporting of all trials whether required under the law or not. For more information, see https://grants.nih.gov/policy/clinical-trials/reporting/index.htm

Institutional Review Board or Independent Ethics Committee Approval: Recipient institutions must ensure that all protocols are reviewed by their IRB or IEC. To help ensure the safety of participants enrolled in NIH-funded studies, the recipient must provide NIH copies of documents related to all major changes in the status of ongoing protocols.

Data and Safety Monitoring Requirements: The NIH policy for data and safety monitoring requires oversight and monitoring of all NIH-conducted or -supported human biomedical and behavioral intervention studies (clinical trials) to ensure the safety of participants and the validity and integrity of the data. Further information concerning these requirements is found at http://grants.nih.gov/grants/policy/hs/data_safety.htm and in the application instructions (SF424 (R&R) and PHS 398).

Investigational New Drug or Investigational Device Exemption Requirements: Consistent with federal regulations, clinical research projects involving the use of investigational therapeutics, vaccines, or other medical interventions (including licensed products and devices for a purpose other than that for which they were licensed) in humans under a research protocol must be performed under a Food and Drug Administration (FDA) investigational new drug (IND) or investigational device exemption (IDE).

2. Administrative and National Policy Requirements

All NIH grant and cooperative agreement awards include the NIH Grants Policy Statement as part of the NoA. For these terms of award, see the NIH Grants Policy Statement Part II: Terms and Conditions of NIH Grant Awards, Subpart A: General and Part II: Terms and Conditions of NIH Grant Awards, Subpart B: Terms and Conditions for Specific Types of Grants, Recipients, and Activities, including of note, but not limited to:

If a recipient is successful and receives a Notice of Award, in accepting the award, the recipient agrees that any activities under the award are subject to all provisions currently in effect or implemented during the period of the award, other Department regulations and policies in effect at the time of the award, and applicable statutory provisions.

If a recipient receives an award, the recipient must follow all applicable nondiscrimination laws. The recipient agrees to this when registering in SAM.gov. The recipient must also submit an Assurance of Compliance (HHS-690). To learn more, see the HHS Office for Civil Rights website.

HHS recognizes that NIH research projects are often limited in scope for many reasons that are nondiscriminatory, such as the principal investigator’s scientific interest, funding limitations, recruitment requirements, and other considerations. Thus, criteria in research protocols that target or exclude certain populations are warranted where nondiscriminatory justifications establish that such criteria are appropriate with respect to the health or safety of the subjects, the scientific study design, or the purpose of the research. For additional guidance regarding how the provisions apply to NIH grant programs, please contact the Scientific/Research Contact that is identified in Section VII under Agency Contacts of this NOFO.

In accordance with the statutory provisions contained in Section 872 of the Duncan Hunter National Defense Authorization Act of Fiscal Year 2009 (Public Law 110-417), NIH awards will be subject to the Federal Awardee Performance and Integrity Information System (FAPIIS) requirements. FAPIIS requires Federal award making officials to review and consider information about an applicant in the designated integrity and performance system (currently FAPIIS) prior to making an award. An applicant, at its option, may review information in the designated integrity and performance systems accessible through FAPIIS and comment on any information about itself that a federal agency previously entered and is currently in FAPIIS. The Federal awarding agency will consider any comments by the applicant, in addition to other information in FAPIIS, in making a judgement about the applicant’s integrity, business ethics, and record of performance under Federal awards when completing the review of risk posed by applicants as described in 2 CFR Part 200.206 “Federal awarding agency review of risk posed by applicants.” This provision will apply to all NIH grants and cooperative agreements except fellowships.

Cooperative Agreement Terms and Conditions of Award

The following special terms of award are in addition to, and not in lieu of, otherwise applicable U.S. Office of Management and Budget (OMB) administrative guidelines, U.S. Department of Health and Human Services (HHS) grant administration regulations at 2 CFR Part 200, and other HHS, PHS, and NIH grant administration policies.

The administrative and funding instrument used for this program will be the cooperative agreement, an "assistance" mechanism (rather than an "acquisition" mechanism), in which substantial NIH programmatic involvement with the recipients is anticipated during the performance of the activities. Under the cooperative agreement, the NIH purpose is to support and stimulate the recipients' activities by involvement in and otherwise working jointly with the recipients in a partnership role; it is not to assume direction, prime responsibility, or a dominant role in the activities. Consistent with this concept, the dominant role and prime responsibility resides with the recipients for the project as a whole, although specific tasks and activities may be shared among the recipients and NIH as defined below.

The PD(s)/PI(s) will have the primary responsibility for:

  • Determining experimental approaches, designing protocols, conducting experiments, and analyzing and interpreting research data for studies funded through this NOFO. The PD/PI is ultimately responsible for the project and will make the final decisions regarding all project plans, provided that they can be executed within NIH-approved budgets and according to NIA/NIH grant and contract policies.
  • Collaborating with NIA Program Staff in establishing a Go/No-Go milestone plan (typically a minimum of one milestone per funding year) at the start of the project and updated as needed.
  • Submitting mid-year and annual milestone progress reports in a standard format for evaluation by NIA Program Staff.
  • Adhering to NIA/NIH policies, including those regarding data release, intellectual property, and publications.
  • Implementing all scientific and policy decisions approved by the NIA/NIH.
  • Working closely with the PDs/PIs' institution's technology transfer officials to ensure that royalty agreements, patent filings, and all other necessary intellectual property arrangements are completed in a timely manner.
  •  Submitting an IND with documentation for one of the following: 1) acceptance of clinical protocol by FDA; 2) elapse of the 30-day post filing waiting period without comment from the FDA; 3) completion of protocol changes or amendments requested by FDA.
  • Submitting a data and safety monitoring plan (DSMP) in accordance with the NIA policies to NIA for review and notification of NIA approval.
  • Registering Phase I trial on clinical trials.gov.

Recipients will retain custody of and have primary rights to the data and software developed under these awards, subject to Government rights of access consistent with current HHS, PHS, and NIH policies. 

NIH staff have substantial programmatic involvement that is above and beyond the normal stewardship role in awards, as described below:

  • An NIA Program Officer will be assigned to the project and will be responsible for the normal scientific and programmatic stewardship of the award. The NIA Program Officer will be named in the award notice and will be the primary contact with the PI/PD. The Program Officer will be responsible for assessing the progress of the project towards accomplishment of milestones, and for recommending the level of continued funding.
  • An NIA Project Scientist will be assigned to the project with substantial scientific and programmatic involvement during the conduct of this activity through technical assistance, advice and coordination that is above and beyond the normal stewardship role in awards. The NIA Project Scientist will provide additional expertise that is needed for proper scientific management of the award. This includes assisting the PI/PD in the development of a project milestone plan at the outset of the project, approving the final milestone language for incorporation into the award notice, enhancing the project's progress by providing access to various NIH resources, when appropriate, providing technical assistance, advice, and coordination to the project, although the dominant role and responsibilities for the activities funded by this NOFO reside with the PI/PD.
  • PLEASE NOTE: If a funded project does not make sufficient progress toward the agreed upon milestones at any stage, funding for the project may be discontinued.

Areas of Joint Responsibility include:

  • None; all responsibilities are divided between recipients and NIH staff as described above.

Dispute Resolution:

Any disagreements that may arise in scientific or programmatic matters (within the scope of the award) between recipients and NIH may be brought to Dispute Resolution. A Dispute Resolution Panel composed of three members will be convened: a designee of the Steering Committee chosen without NIH staff voting, one NIH designee, and a third designee with expertise in the relevant area who is chosen by the other two; in the case of individual disagreement, the first member may be chosen by the individual recipient. This special dispute resolution procedure does not alter the recipient's right to appeal an adverse action that is otherwise appealable in accordance with PHS regulation 42 CFR Part 50, Subpart D and HHS regulation 45 CFR Part 16.

3. Data Management and Sharing

Consistent with the 2023 NIH Policy for Data Management and Sharing, when data management and sharing is applicable to the award, recipients will be required to adhere to the Data Management and Sharing requirements as outlined in the NIH Grants Policy Statement. Upon the approval of a Data Management and Sharing Plan, it is required for recipients to implement the plan as described.

If patent protection is being sought, investigators should explain how data will be shared after filing for patent protection to allow for both further research and the development of commercial products to advance forward, consistent with achieving the goals of the program.

4. Reporting

When multiple years are involved, recipients will be required to submit the Research Performance Progress Report (RPPR) annually and financial statements as required in the NIH Grants Policy Statement.

A final RPPR, invention statement, and the expenditure data portion of the Federal Financial Report are required for closeout of an award, as described in the NIH Grants Policy Statement. NIH NOFOs outline intended research goals and objectives. Post award, NIH will review and measure performance based on the details and outcomes that are shared within the RPPR, as described at 2 CFR Part 200.301.

The Federal Funding Accountability and Transparency Act of 2006, as amended (FFATA), includes a requirement for recipients of Federal grants to report information about first-tier subawards and executive compensation under Federal assistance awards issued in FY2011 or later.  All recipients of applicable NIH grants and cooperative agreements are required to report to the Federal Subaward Reporting System (FSRS) available at www.fsrs.gov on all subawards over the threshold.  See the NIH Grants Policy Statement for additional information on this reporting requirement.

In accordance with the regulatory requirements provided 2 CFR Part 200.113 and Appendix XII to 2 CFR Part 200, recipients that have currently active Federal grants, cooperative agreements, and procurement contracts from all Federal awarding agencies with a cumulative total value greater than $10,000,000 for any period of time during the period of performance of a Federal award, must report and maintain the currency of information reported in the System for Award Management (SAM) about civil, criminal, and administrative proceedings in connection with the award or performance of a Federal award that reached final disposition within the most recent five-year period.  The recipient must also make semiannual disclosures regarding such proceedings. Proceedings information will be made publicly available in the designated integrity and performance system (currently FAPIIS).  This is a statutory requirement under section 872 of Public Law 110-417, as amended (41 U.S.C. 2313).  As required by section 3010 of Public Law 111-212, all information posted in the designated integrity and performance system on or after April 15, 2011, except past performance reviews required for Federal procurement contracts, will be publicly available.  Full reporting requirements and procedures are found in Appendix XII to 2 CFR Part 200 – Award Term and Condition for Recipient Integrity and Performance Matters.

Section VII. Agency Contacts

We encourage inquiries concerning this funding opportunity and welcome the opportunity to answer questions from potential applicants.

Application Submission Contacts

eRA Service Desk (Questions regarding ASSIST, eRA Commons, application errors and warnings, documenting system problems that threaten submission by the due date, and post-submission issues)

Finding Help Online: https://www.era.nih.gov/need-help (preferred method of contact)
Telephone: 301-402-7469 or 866-504-9552 (Toll Free)

General Grants Information (Questions regarding application instructions, application processes, and NIH grant resources)
Email: GrantsInfo@nih.gov (preferred method of contact)
Telephone: 301-637-3015

Grants.gov Customer Support (Questions regarding Grants.gov registration and Workspace)
Contact Center Telephone: 800-518-4726
Email: support@grants.gov

Scientific/Research Contact(s)

Lorenzo M. Refolo, Ph.D.
National Institute on Aging (NIA)
Telephone: 301-594-7576
Email: refolol@nia.nih.gov

Peer Review Contact(s)

Ramesh Vemuri, Ph.D.
National Institute on Aging (NIA)
Telephone: 301-402-7700
Email: ramesh.vemuri@nih.gov

Financial/Grants Management Contact(s)

Jeni Smits
National Institute on Aging (NIA)
Telephone: 301-496-1472
Email: jeni.smits@nih.gov

Section VIII. Other Information

Recently issued trans-NIH policy notices may affect your application submission. A full list of policy notices published by NIH is provided in the NIH Guide for Grants and Contracts. All awards are subject to the terms and conditions, cost principles, and other considerations described in the NIH Grants Policy Statement.

Authority and Regulations

Awards are made under the authorization of Sections 301 and 405 of the Public Health Service Act as amended (42 USC 241 and 284) and under Federal Regulations 42 CFR Part 52 and 2 CFR Part 200.

NIH Office of Extramural Research Logo
Department of Health and Human Services (HHS) - Home Page
Department of Health
and Human Services (HHS)
USA.gov - Government Made Easy
NIH... Turning Discovery Into Health®