AIDS Imaging Research—Integrated Research Facility at Fort Detrick

The AIDS Imaging Research Section (AIRS) leverages preclinical and translational molecular imaging to study the pathogenesis of human immunodeficiency virus (HIV) infection using the simian/simian-human immunodeficiency virus (SIV/SHIV) nonhuman primate model.

Sinu P. John, Ph.D.

Section or Unit Name
Signaling Systems Section
Exclude from directory
Off
Section/Unit: Year Established
Section/Unit: Location
This Researcher/Clinician’s Person Page
Program Description

Our research focuses primarily on identification of cell intrinsic factors (protein coding and non-coding genes) associated with regulation of macrophage signaling. We use high throughput genome-wide techniques such as RNAi screening, CRISPR screening, RNA-seq, ATAC-seq, etc. to identify and characterize the genes and gene-regulatory mechanisms that modulate the immune response in macrophage cells. In addition, we study the role of various external factors (environmental pollutants, drugs, diet, etc.) that modulate the immune response in macrophages with an emphasis to develop therapeutic candidates for the treatment of infectious and immune diseases. We use both bacterial and several emerging viral models such as HIV, Influenza, SARS-CoV-2, etc. to study the impact of immune regulation by various intrinsic and external factors.

Selected Publications

John SP, Singh A, Sun J, Pierre MJ, Alsalih L, Lipsey C, Traore Z, Balcom-Luker S, Bradfield CJ, Song J, Markowitz TE, Smelkinson M, Ferrer M, Fraser IDC. Small-molecule screening identifies Syk kinase inhibition and rutaecarpine as modulators of macrophage training and SARS-CoV-2 infection. Cell Rep. 2022 Oct 4;41(1):111441.

John SP, Sun J, Carlson RJ, Cao B, Bradfield CJ, Song J, Smelkinson M, Fraser IDC. IFIT1 Exerts Opposing Regulatory Effects on the Inflammatory and Interferon Gene Programs in LPS-Activated Human Macrophages. Cell Rep. 2018 Oct 2;25(1):95-106.e6.

John SP, Chin CR, Perreira JM, Feeley EM, Aker AM, Savidis G, Smith SE, Elia AE, Everitt AR, Vora M, Pertel T, Elledge SJ, Kellam P, Brass AL. The CD225 domain of IFITM3 is required for both IFITM protein association and inhibition of influenza A virus and dengue virus replication. J Virol. 2013 Jul;87(14):7837-52.

Zhu J, Gaiha GD, John SP, Pertel T, Chin CR, Gao G, Qu H, Walker BD, Elledge SJ, Brass AL. Reactivation of latent HIV-1 by inhibition of BRD4. Cell Rep. 2012 Oct 25;2(4):807-16.

Everitt AR, Clare S, Pertel T, John SP, Wash RS, Smith SE, Chin CR, Feeley EM, Sims JS, Adams DJ, Wise HM, Kane L, Goulding D, Digard P, Anttila V, Baillie JK, Walsh TS, Hume DA, Palotie A, Xue Y, Colonna V, Tyler-Smith C, Dunning J, Gordon SB; GenISIS Investigators; MOSAIC Investigators; Smyth RL, Openshaw PJ, Dougan G, Brass AL, Kellam P. IFITM3 restricts the morbidity and mortality associated with influenza. Nature. 2012 Mar 25;484(7395):519-23.

Brass AL, Huang IC, Benita Y, John SP, Krishnan MN, Feeley EM, Ryan BJ, Weyer JL, van der Weyden L, Fikrig E, Adams DJ, Xavier RJ, Farzan M, Elledge SJ. The IFITM proteins mediate cellular resistance to influenza A H1N1 virus, West Nile virus, and dengue virus. Cell. 2009 Dec 24;139(7):1243-54.

Visit PubMed for a complete publication listing.

Major Areas of Research
  • Genes and epigenetic states modulating macrophage signaling and function
  • Identification and characterization of trained immunity stimuli
  • Applications of trained immunity in infectious and immune disease

Four Research Areas Comprise NIAID’s 2023 Omnibus Contract Solicitation

Funding News Edition:
See more articles in this edition

If you can carry out research that supports program objectives of NIAID’s Division of Microbiology and Infectious Diseases (DMID), consider submitting a proposal through the 2023 NIAID Omnibus Broad Agency Announcement (BAA) contract solicitation.

DMID will use this BAA to advance the research and development of promising candidate vaccines, therapeutics, and diagnostics for biodefense, emerging infectious diseases, and pandemic preparedness.

As we summarize below, the 2023 BAA covers four distinct research areas. You may respond to one, any combination of, or all the research areas. For each research area, be sure to present separate, detailed technical and business proposals designed to meet the objectives described.

For all four research areas:

Research Area 001: Development of Vaccine Candidates for Biodefense and Emerging Infectious Diseases

  • Description: Supports advancing vaccine candidates, technologies, and platforms that could be deployed against agents that include category A, B, and C NIAID Emerging Infectious Diseases and Pathogens.
  • Number of Awards, Total Costs: For fiscal year (FY) 2024, NIAID estimates it will award up to $12.8 million total for the non-severable base period of five or six cost-reimbursement type contracts across research areas 001, 002, and 004.
  • Due Date: April 11, 2023, by 3 p.m. Eastern Time.

Research Area 002: Development of Therapeutic Candidates for Biodefense, Antimicrobial Resistant (AMR) Infections, and Emerging Infectious Diseases

  • Description: Supports the development of promising new therapeutics to address infections and diseases caused by NIAID Emerging Infectious Diseases and Pathogens (including category A, B, and C priority pathogens) and selected bacterial and fungal pathogens identified in the CDC 2019 Antibiotic Resistance Threats in the United States report.
  • Number of Awards, Total Costs: For FY 2024, NIAID estimates it will award up to $12.8 million total for the non-severable base period of five or six cost-reimbursement type contracts across research areas 001, 002, and 004.
  • Due Date: April 11, 2023, by 3 p.m. Eastern Time.

Research Area 003: The Antiviral Program for Pandemics (APP): Development of Antivirals for RNA Viral Families of Pandemic Potential

  • Description: Supports the development of antivirals as described in the Antiviral Program for Pandemics. NIAID encourages proposals to develop safe and effective antivirals to combat SARS-CoV-2, the virus that causes COVID-19, as well as to build sustainable platforms for targeted drug discovery and development of a robust pipeline of antivirals against viruses with pandemic potential.
  • Number of Awards, Total Costs: For FY 2024, NIAID estimates it will award up to $3.5 million total for the non-severable base period of one cost-reimbursement type contract.
  • Due Date: March 13, 2023, by 3 p.m. Eastern Time.

Research Area 004: Development of In Vitro Diagnostics for Biodefense, AMR Infections, and Emerging Infectious Diseases

  • Description: Supports the development of promising diagnostics technologies for detection of signatures from biothreat pathogens and pathogens causing emerging, reemerging, and AMR infectious diseases, and for pandemic preparedness.
  • Number of Awards, Total Costs: For FY 2024, NIAID estimates it will award up to $12.8 million total for the non-severable base period of five or six cost-reimbursement type contracts across research areas 001, 002, and 004.
  • Due Date: April 11, 2023, by 3 p.m. Eastern Time.

Find complete details in the solicitation itself as well as any amendments issued since this article was published.

Contact Us

Email us at deaweb@niaid.nih.gov for help navigating NIAID’s grant and contract policies and procedures.

New Solicitation Focuses on Development of Cellular Immunology Core

Funding News Edition:
See more articles in this edition

NIAID supports basic research on microbiology and immunology that may lead to the development of vaccines, therapeutics, and medical diagnostics for the prevention, treatment, and diagnosis of infectious and immune-mediated diseases.

NIAID’s Division of AIDS is seeking a partner to operate the Cellular Immunology Core Laboratory, which will conduct, analyze, develop, optimize, and validate cellular immunologic assays for HIV, Simian Immunodeficiency Virus, Mycobacterium tuberculosis, and other pathogens, to be performed on fresh and frozen preclinical samples.

A new request for proposals (RFP) seeks contractors who can do the following:

  • Conduct validated immunological assays using good laboratory practice (GLP)-like processes
  • Perform data analyses
  • Receive, store, catalog, track, and maintain an inventory of the specimens for evaluation
  • Manage, report, and deposit study data
  • Perform project management activities related to contract activities
  • Conduct initial and final transition activities, as needed

The contractor shall use current state-of-the-art technologies, including, at a minimum, ELISPOT, intracellular cytokine staining, flow cytometry, cell sorting, tetramer staining, and other assays to accomplish the technical objectives, and shall incorporate new and optimized technologies for assay development into contract activities when appropriate. Assays shall incorporate appropriate positive and negative controls using reference panels to define background and dynamic range and shall demonstrate reproducibility and consistency.

Additionally, the contractor shall also use state-of-the-art methods to analyze the data generated by the conduct of the assays, which may require different data analysis methods/platforms as appropriate for each type of assay. Data analyses may require generating publication quality figures, when directed by the contracting officer’s representative. Lastly, an independent Quality Assurance Program, not associated with the contract, shall conduct regular inspections to review facilities, equipment, personnel, methods, practices, and records.

Details and Due Date

Read the RFP Cellular Immunology Core Laboratory for complete details.

NIAID anticipates awarding one cost reimbursement, level-of-effort (term) type contract for a 1-year base period plus six 1-year options for a total possible performance period of 7 years, beginning on March 1, 2024.

Proposals are due on March 17, 2023, by 3 p.m. Eastern Time. Submit applications online through NIH’s electronic Contract Proposal Submission (eCPS) site.

Send inquiries to Kimberly Dormer, NIAID’s contract specialist for this opportunity, at kimberly.dormer@nih.gov.

Contact Us

Email us at deaweb@niaid.nih.gov for help navigating NIAID’s grant and contract policies and procedures.

Fernanda D. Young, M.D.

Section or Unit Name
Food Allergy Research Section
Exclude from directory
Off
Section/Unit: Year Established
Section/Unit: Location
This Researcher/Clinician’s Person Page
Parent Lab/Program
Program Description

Food allergy is a significant health problem in the United States, affecting children and adults, both immune competent and immune compromised. The overall goal of the Food Allergy Research Section (FARS) is to understand the genetic, immunologic, and biochemical pathways that lead to the development of food allergy and how they can be modified for therapeutic benefit. We aim to achieve this goal using a multifaceted approach with studies involving both patients and animal models of their diseases.

Our team in particular is studying why only some patients who have detectable IgE to specific foods experience an allergic reaction when they eat the food, and we are working to identify immunologic markers that can predict the severity and persistence of food allergy.

By achieving a greater understanding of the key environmental, immunologic, and biochemical pathways that drive the development of food allergy, we will be able to develop novel interventions that are based on a advanced understanding of disease pathogenesis.

Selected Publications

Tsao LR, Young FD, Otani IM, Castells MC. Hypersensitivity Reactions to Platinum Agents and Taxanes. Clin Rev Allergy Immunol. 2022 Jun;62(3):432-448.

Hubbard TP, Billings G, Dörr T, Sit B, Warr AR, Kuehl CJ, Kim M, Delgado F, Mekalanos JJ, Lewnard JA, Waldor MK. A live vaccine rapidly protects against cholera in an infant rabbit model. Sci Transl Med. 2018 Jun 13;10(445):eaap8423.

Dörr T, Delgado F, Umans BD, Gerding MA, Davis BM, Waldor MK. A Transposon Screen Identifies Genetic Determinants of Vibrio cholerae Resistance to High-Molecular-Weight Antibiotics. Antimicrob Agents Chemother. 2016 Jul 22;60(8):4757-63.

Dörr T, Alvarez L, Delgado F, Davis BM, Cava F, Waldor MK. A cell wall damage response mediated by a sensor kinase/response regulator pair enables beta-lactam tolerance. Proc Natl Acad Sci U S A. 2016 Jan 12;113(2):404-9.

Malen R, Knerr S, Delgado F, Fullerton SM, Thompson B. Rural Mexican-Americans' perceptions of family health history, genetics, and disease risk: implications for disparities-focused research dissemination. J Community Genet. 2016 Jan;7(1):91-6.

Delgado F, Tabor HK, Chow PM, Conta JH, Feldman KW, Tsuchiya KD, Beck AE. Single-nucleotide polymorphism arrays and unexpected consanguinity: considerations for clinicians when returning results to families. Genet Med. 2015 May;17(5):400-4.

Visit PubMed for a complete publication listing.

Additional Information
Major Areas of Research

Dr. Young’s work places an emphasis on clinical research to identify contributory factors to:

  • food allergy and allergic disease
  • transplant associated food allergy
  • drug allergy and drug reactions

Fernanda D. Young, M.D.

Contact: For contact information, search the NIH Enterprise Directory.

Specialty(s): Allergy and Immunology, Internal Medicine, Pediatrics
Provides direct clinical care to patients at NIH Clinical Center

Education:

M.D., University of Washington School of Medicine, Seattle, WA

Languages Spoken: Spanish
Fern Young Headshot

Assessment of PrEP Eligibility Tool Among the General Population in Uganda

NIAID Now |

To reduce the spread of HIV infection, the World Health Organization (WHO) recommends pre-exposure prophylaxis (PrEP) in populations with an annual HIV infection incidence greater than 3%. Given that the annual HIV incidence in Uganda is estimated at 0.40% (0.46% among females and 0.35% among males), there are people who may have a higher HIV risk and may benefit from PrEP but are not targeted for services. To further reduce the spread of HIV in Uganda, researchers used results from three survey rounds of HIV-negative participants within the Rakai Community Cohort Study (RCCS) to estimate the prevalence of high-risk individuals eligible for PrEP within the general population and the incidence of HIV infection associated with eligibility. 

In this study, a subset of questions from Uganda’s PrEP eligibility tool that are routinely asked within the RCCS surveys were used to determine PrEP eligibility. Eligibility was defined as reporting at least one of the following HIV risk behaviors in the past 12 months: sexual intercourse with more than one partner of unknown HIV status; nonmarital sex act without a condom; sex engagement in exchange for money, goods, or services; or experiencing genital ulcers. HIV incidence was estimated by analyzing seroconversion from HIV-negative to HIV-positive in participants who contributed to at least two of the survey rounds.

Overall, 29% of participants in the analysis met the eligibility criteria. Of these, 22% reported one HIV risk, 6% reported two HIV risks, and 1% reported three HIV risks. The results showed that PrEP eligible participants had twice the risk of acquiring HIV than their non-eligible counterparts. Furthermore, risk increased threefold in uncircumcised males but not circumcised males. Additionally, men who reported higher prevalence of risky behaviors had lower increase in HIV incidence compared to women, likely due to circumcision status and higher antiretroviral therapy coverage in HIV-infected females, leading to a decrease in transmission to men. The findings of this study support the use of PrEP eligibility screening in general populations with HIV incidence lower than 3% to reduce HIV acquisition even further in these populations.

Reference: Ssempijja et al. High Rates of Pre-exposure Prophylaxis Eligibility and Associated HIV Incidence in a Population With a Generalized HIV Epidemic in Rakai, Uganda, JAIDS Journal of Acquired Immune Deficiency Syndromes, July 1, 2022 - Volume 90 - Issue 3 - p 291-299.

Contact Information

Contact the NIAID Media Team.

301-402-1663
niaidnews@niaid.nih.gov

Search NIAID Blog

Opportunity to Establish Consortium for Design of TB Drug Regimens

Funding News Edition:
See more articles in this edition

Tuberculosis (TB) is currently a leading cause of death by an infectious disease and, according to the World Health Organization, 9.9 million people became ill with TB and 1.5 million died of TB in 2020. Annually, approximately 3.3 percent of new cases and 18 percent of previously treated cases are rifampin-resistant TB. Around one-quarter of the world’s population has latent TB, and CDC estimates that as many as 13 million latently infected people live in the United States.

To address this pandemic, NIAID’s 2018 Strategic Plan for Tuberculosis Research supports research to improve treatment for all forms of TB in all populations and age groups, including research to develop less toxic regimens of shorter duration for safe and effective treatment.

In recent years, the global scientific community has focused on applying cutting-edge science and technologies to understanding the physiology and pathogenesis of Mycobacterium tuberculosis (Mtb) and translating the knowledge gained into targeted therapy. However, limited pharmaceutical industry engagement in antimicrobial drug development has slowed the translation of these discoveries into effective treatment regimens.

NIAID invites applications to establish a Consortium of TB preclinical and clinical experts to systematically refine preclinical models by analyses of relevant preclinical and clinical data through the funding opportunity announcement (FOA) Consortium for Design of TB Drug Regimens (UM1, Clinical Trial Not Allowed).

Consortium Research Objectives

The Consortium will establish a collaborative, multidisciplinary research platform to systematically evaluate new candidate agents in combination. These agents may originate from academic, public-private partnerships, or pharmaceutical industry research efforts, and the Consortium will evaluate them for their potential for incorporation into new TB drug regimens across the human lifespan, including young children.

By generating adequate and comparable preclinical data on regimen efficacy, pharmacometrics, toxicities, drug-drug interactions, and other factors, the Consortium will increase efficiency, accelerate development of new combinations, and identify the most promising regimens for future clinical testing to effect shorter, relapse-free cures of pulmonary TB in adults, children, and persons being treated for HIV.

The TB Drug Regimen Consortium will take the following actions:

  • Provide an open platform to unite preclinical and clinical TB experts (U.S. and international) in the systematic analysis of available preclinical and clinical efficacy and safety data to identify preclinical research projects, scientific needs/gaps, and set an agenda of cooperative goals and research priorities.
  • Optimize preclinical models with back-translation of clinical data from recent trials into animal and other model development.
  • Facilitate communication, coordination, and planning of preclinical research to inform NIH-supported clinical trial networks (both adult and pediatric focused) and other sponsors/investigators by review and discussion of all available information on new agents/combinations and ongoing and anticipated research.
  • Incorporate pediatric-focused research to stimulate preclinical, translational activities that can enable accelerated clinical testing and earlier introduction of new TB drugs and regimens in pediatric populations, with an emphasis on young children (aged 6 years or younger).
  • Perform preclinical evaluations of mechanisms of action of individual and combination drug candidates.
  • Perform preclinical animal model studies (with emphasis on new drug combination efficacy comparisons) identified as needed by the Consortium leadership executive committee after review and approval by NIAID.
  • Provide compiled data and analyses on evaluated combination regimens with priority ranking assessment for consideration by clinical trial networks to guide optimized and coordinated choices of new regimens for phase 2 and 3 trials.
  • Assess evolving clinical research challenges and opportunities for potential inclusion of special populations, new technologies (including biomarkers), and the broader spectrum of TB disease into the Consortium research agenda.

The Consortium will use a strategic planning process for its scientific and translational activities. In establishing and refining research for treatment of TB diseases, the Consortium will actively engage major sponsors, researchers, and communities within and outside of the Consortium. The Consortium Scientific Leadership Group Executive Committee (SLG Ex Com) will ensure that the research is coordinated and complementary to other TB drug research enterprises.

TB Consortium Organizational Structure

The Consortium’s structure will facilitate coordinated and efficient TB drug regimen development. It will include a Scientific Leadership Group (SLG) including a Pediatric Focus Committee, led by SLG Ex Com, a Preclinical Laboratory Group (PLG), a Data Science and Modelling Group (DSMG), and an Administrative Group (AG). These functional areas will be integrally connected to all aspects of the research agenda for the treatment of TB diseases, focusing primarily on pulmonary TB in both adults and children.

Below are some characteristics of the TB Consortium. For full descriptions of each required committee, refer to the FOA linked at the beginning of the article.

SLG Executive Committee (SLG Ex Com) will be responsible for final decisions on prioritized research to be performed, ensuring progress, and providing administrative oversight. This group includes the Consortium program directors and principal investigators (PDs/PIs), the leaders of Preclinical Laboratories and the Data Science and Modelling Group, the chair of the Pediatric Focus Committee, NIAID clinical trial network representatives, and NIAID scientific representatives.

Pediatric Focus Committee (PFC) will ensure that pediatric populations benefit from research by the Consortium, including suggesting additional research needed to extend the impact of Consortium activities towards pediatric populations (especially related to pediatric pulmonary TB). The PFC will formulate plans to initiate relevant key pediatric translational and preclinical activities early during clinical testing of TB drug/combination drug regimens in adults, including those prioritized by SLG or by the Report of the meeting to review the WHO’s Paediatric Antituberculosis Drug Optimization priority list.

Preclinical Laboratory Group (PLG) consists of Preclinical Laboratories (PLs) and will be responsible for designing and implementing in vitro and in vivo studies of efficacy, mechanisms of action, safety, and pharmacometrics of drugs/combinations approved by the SLG Ex Com. At least two PLs will operate mouse models of TB pulmonary disease with pharmacology/pharmacodynamic capabilities for each drug tested and will have experience in successfully informing clinical trial design. Additional PLs may focus on other small animal models of disease, lesion-centric evaluations and rankings, novel quantitative measures of mycobacterial burden/response to therapy, biomarkers, drug effects on mycobacterial metabolism, or other areas supportive of drug regimen evaluation.

Data Science and Modelling Group (DSMG) will summarize and model data for the Consortium to review. The DSMG will also be responsible for collecting, organizing, validating, and analyzing existing preclinical and clinical data from completed studies and trials, from across the published literature, and from clinical trial meta data provided by associated networks/sponsors of adult and pediatric studies.

In conjunction with the Administrative Group, the DSMG will compile reports for the SLG (e.g., assembling dossiers on candidate drug’s history, chemistry, manufacturing and controls, preclinical efficacy and toxicity data, and clinical trial data across the human lifespan, including children). The DSMG will maintain a public chemical structure-based database of known drugs and new candidate compounds with links to public chemical databases and PubMed abstracts.

Administrative Group (AG) provides administrative support, organizes meetings, compiles summaries, facilitates communications, and prepares reports. The AG will coordinate research and data sharing agreements and organize experimental results in formats suitable for regulatory filings, as well as develop and prepare effective legal agreements between participating parties for maximizing disclosure of data to protect intellectual property.

After award, NIAID will form an External Advisory Board to evaluate and comment on scientific progress to the SLG Ex Com of the Consortium.

Note: The FOA lists several research areas that NIAID considers nonresponsive and if you include any of them in your application, NIAID will not review it. For the full list of nonresponsive research areas, go to the FOA linked at the beginning of the article.

Administrative Details and Due Date

Application budgets are not limited but need to reflect the actual needs of the proposed project.

Your proposed project period cannot exceed 5 years. All applications are due February 7, 2023, by 5 p.m. local time of the applicant organization.

Direct any inquiries to Dr. Barbara Laughon at 240-627-3302 or blaughon@mail.nih.gov. Direct questions specific to the pediatric component to Dr. Tania Lombo at 301-761-7612 or tania.lombo@nih.gov.

For matters concerning peer review, contact Dr. Yong Gao at 240-669-5048 or yong.gao@nih.gov.

Contact Us

Email us at deaweb@niaid.nih.gov for help navigating NIAID’s grant and contract policies and procedures.

First FDA-Approved Treatment for Eosinophilic Esophagitis Has Roots in NIAID-Funded Research

NIAID Now |

The first Food and Drug Administration approval of a treatment for eosinophilic esophagitis, announced in May 2022, marked a vital achievement not only for people with the disease, but also for scientists including a NIAID grantee whose research helped lay a foundation for this milestone.

Eosinophilic esophagitis, or EoE, is a chronic disease characterized by an overabundance of a specific type of white blood cell, an eosinophil, in the esophagus. The disease is driven by allergic inflammation due to food. This inflammation damages the esophagus and prevents it from working properly. For people with EoE, swallowing even small amounts of food can be a painful and worrisome choking experience. EoE is the most common reason people must go to emergency departments for a healthcare provider to remove food stuck in their esophagus. People with EoE are often left to contend with the frustration and anxiety of a sometimes-lengthy list of foods to avoid, a poor quality of life, and a higher risk of depression. In severe cases, a feeding tube may be the only option to ensure proper caloric intake and adequate nutrition. About 160,000 people in the United States are living with EoE.

NIAID funding enabled Marc E. Rothenberg, M.D., Ph.D., at Cincinnati Children’s in Ohio to conduct basic and preclinical research starting in 1999 that uncovered the molecular cause of EoE. This finding suggested the type of drug needed to treat the disorder.

Before 2001, some in the medical community thought EoE was a form of acid reflux disease. Dr. Rothenberg’s lab published a paper that year establishing that EoE is actually an allergic disorder. The paper showed that mice exposed to an inhaled respiratory allergen developed EoE. This helped focus subsequent studies of EoE on the role of allergic inflammation.

A few years later, two seminal papers from Dr. Rothenberg’s lab identified the molecular changes taking place in the esophagus of people with EoE and showed that a cell-signaling molecule called IL-13 was responsible for many of those changes. IL-13 and another cell-signaling molecule, IL-4, together drive allergic inflammation in many diseases.

The first of these two key papers reported the results of an analysis of gene expression in cells on the lining of the esophagus in people with and without EoE. The study identified 574 genes that were copied into RNA “transcripts”—the instructions for making proteins—in greater or smaller numbers in people with EoE than in healthy people. This EoE transcript signature, or transcriptome, served as a key reference for subsequent studies of the disorder.

The second paper demonstrated in 2007 that many of the EoE-associated genes identified in the earlier paper are directly activated by IL-13 in cells lining the esophagus, implicating this molecule as a major regulator of the biological pathways involved in EoE. This finding suggested that IL-13-blocking drugs might effectively treat the disease. The study further showed that 98% of the EoE transcriptome reverted to normal levels of gene expression in people whose EoE was successfully treated with a class of steroid hormones called glucocorticoids. This indicated that the EoE transcriptome changes in response to changes in signs and symptoms of the disease. Dr. Rothenberg and colleagues therefore proposed using the EoE transcriptome to monitor the efficacy and mechanism of action of IL-13-blocking drugs at the molecular level.

Several years later, Dr. Rothenberg designed and led the first clinical trial to test the efficacy of an anti-IL-13 monoclonal antibody for treating EoE. The investigators found that the antibody lowered levels of eosinophils in the esophagus and returned the expression of 29 key genes in the EoE transcriptome to normal levels in most treated participants. This and related molecular analyses from the trial, funded by Novartis Pharma AG of Basel, Switzerland, further supported Dr. Rothenberg’s theory that EoE was driven by IL-13.

These findings contributed to a body of foundational EoE research developed by a multitude of scientists and physicians. This scientific foundation, among many other factors, led Regeneron Pharmaceuticals Inc. of Tarrytown, New York, and Sanofi of Paris to begin testing one of their drugs for the treatment of EoE. The drug, a monoclonal antibody called dupilumab, works by blocking both IL-13 and IL-4.

Ultimately, Regeneron and Sanofi conducted a Phase 3 clinical trial showing dupilumab substantially improved the signs and symptoms of EoE compared to a placebo. Based on these results, FDA approved dupilumab for the treatment of the disease on May 20, 2022, making it the first medicine specifically indicated to treat EoE in the United States.

NIAID-funded basic and translational research continues to contribute to the development of preventive and therapeutic strategies for dozens of allergic, immunologic, and infectious diseases.

References:

A Mishra, et al. An etiological role for aeroallergens and eosinophils in experimental esophagitis. The Journal of Clinical Investigation DOI: 10.1172/JCI10224 (2001).

C Blanchard, et al. Eotaxin-3 and a uniquely conserved gene-expression profile in eosinophilic esophagitis. The Journal of Clinical Investigation DOI: 10.1172/JCI26679 (2006).

C Blanchard, et al. IL-13 involvement in eosinophilic esophagitis: Transcriptome analysis and reversibility with glucocorticoids. Journal of Allergy and Clinical Immunology DOI: 10.1016/j.jaci.2007.10.024 (2007).

ME Rothenberg, et al. Intravenous anti–IL-13 mAb QAX576 for the treatment of eosinophilic esophagitis. Journal of Allergy and Clinical Immunology DOI: 10.1016/j.jaci.2014.07.049 (2015).

Contact Information

Contact the NIAID Media Team.

301-402-1663
niaidnews@niaid.nih.gov

Search NIAID Blog

John Misasi, M.D.