Immunomonitoring of Mold Invasive Infections (IMMUNOFIL)

February 22, 2024 updated by: Assistance Publique - Hôpitaux de Paris
Mold invasive infections are associated with an important mortality despite optimization of the antifungal treatment. In a few case reports, immune checkpoints inhibitors, initially developed for neoplastic diseases, have shown a potential beneficial effect in such devastating infections by restoring an efficient immune response. The investigators propose a longitudinal monitoring of the adaptative immune response, notably immune checkpoint expression on T cells, during mold invasive infections to help identify the patients who could benefit from the adjunction of immunotherapy and the optimal timing of such strategy.

Study Overview

Status

Not yet recruiting

Detailed Description

Invasive fungal diseases (IFD) still cause substantial morbidity and mortality. New therapeutic approaches are therefore urgently needed, notably for patients not responding to conventional antifungal treatment. One strategy to prevent treatment failure is to improve the immune functions of immunocompromised hosts. Indeed, antifungals therapeutic efficacy is limited without the help of host immune reactivity (Stevens et al., 2000). As several studies have suggested that IFD are associated with an impaired Th1 host immune response, various cytokines have been evaluated in experimental and human fungal infections. In particular, adjunction of recombinant IFNγ has been proposed as a treatment option in patients with poor prognosis IFD with partial success. More recently, it has been shown that, when T cells are exposed to persistent antigens and/or inflammatory signals due to inefficient control of persisting infections, or in the context of tumors, a deterioration of their functions is observed, a state called "exhaustion". Molecular pathways involved in exhaustion have been partially deciphered, highlighting the importance of molecules called immune checkpoint such as PD-1 or CTLA-4. Furthermore, it was shown that blockade of these molecules can reverse this dysfunctional state. Immune checkpoint inhibitors (such as anti-PD1 antibodies) have become major weapons in oncology. In infectious diseases, and more particularly IFDs, data are much more limited. In animal models of several IFD, such as aspergillosis, cryptococcosis and histoplasmosis, repetitive administration of anti-PD-1 monoclonal antibodies significantly improved fungal clearance and survival of lethally infected animals. In humans, three case-reports, including one published by our team, have reported the efficacy of anti-PD-1 therapy combined with IFN-γ in the treatment of refractory IFDs. Therefore, by reversing T cell exhaustion, immune checkpoint blockade represents a therapeutic perspective for IFD treatment.

The use of immune checkpoint inhibitors may, however, entail severe, notably autoimmune off-target adverse effects and should be carefully balanced and monitored. The development of tools allowing identification of patients who could benefit from immunotherapy is of particular importance as well as assessment of the optimal timing of these innovative treatments. As such, a better understanding of the host immune response is one of the major approaches to developing new or improved antifungal strategies to control IFDs. Longitudinal data regarding the evolution of exhaustion markers expression in T cells of patients treated for an IFD are lacking.

The goal is to better characterize the adaptative immune response directed against molds, notably immune checkpoint expression, in order to identify the patients who could benefit from the adjunction of immunotherapy and the optimal timing of such strategy.

For this purpose, the investigators will include adult patients with mold IFD either at diagnosis or refractory to conventional therapy. They will measure activation and exhaustion markers on circulating T cells and monocytes by flow cytometry at three timepoints (enrollment, day 14 and week 6). Moreover, for patients with invasive aspergillosis or mucormycosis, they will evaluate the capacity of specific T cells to produce Th1, Th2 and Th 17 cytokines and to proliferate after specific antigenic stimulation, in the absence and in the presence of an anti-PD1 antibody in vitro (4 colors FLUOROSPOT) at two timepoint (enrollment and week 6). These data will provide a longitudinal assessment of the anti-fungal immune response. They will be correlated with the underlying diseases of the patients, the type of mold infection (aspergillosis, mucormycosis, fusariosis or scedosporiosis), the treatment received and the outcome. These results should help to better identify patients who could benefit from adjunctive anti PD-1 treatment and the optimal timing for such treatment.

Study Type

Observational

Enrollment (Estimated)

20

Contacts and Locations

This section provides the contact details for those conducting the study, and information on where this study is being conducted.

Study Contact

Study Contact Backup

Study Locations

      • Paris, France, 75015
        • Hôpital Necker Enfants Malades
        • Contact:

Participation Criteria

Researchers look for people who fit a certain description, called eligibility criteria. Some examples of these criteria are a person's general health condition or prior treatments.

Eligibility Criteria

Ages Eligible for Study

  • Adult
  • Older Adult

Accepts Healthy Volunteers

No

Sampling Method

Non-Probability Sample

Study Population

Tertiary care centers

Description

Inclusion Criteria:

  • Age > 18 years
  • Mold invasive fungal infection: Aspergillus, Mucorales, Fusarium, Scedosporium
  • Proven or probable according to 2019 EORTC/MGS criteria modified by the adjunction of diabetes mellitus in the host criteria and Mucorales PCR in the microbiological criteria
  • Within 14 days of IFD diagnosis or at a refractory state defined by the 2009 MGS/EORT failure criteria (clinical, radiological, or microbiological failure) of a first-line antifungal treatment leading to a change of therapy by the attending physician of the patient

Exclusion Criteria:

  • Lymphocyte count < 0.5 G/L
  • Bacterial infection in the last 14 days
  • Previous treatment with anti-PD1 antibodies

Study Plan

This section provides details of the study plan, including how the study is designed and what the study is measuring.

How is the study designed?

Design Details

Cohorts and Interventions

Group / Cohort
Cohort of patients with mold invasive fungal infection
Patients >18 y, with a diagnosis of proven or probable mold invasive fungal infection (Aspergillus, Mucorales, Fusarium or Scedosporium), according to modified 2019 EORTC/MGS criteria, at diagnosis or at a refractory state after a first-line antifungal treatment

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Immune checkpoint expression on T cells
Time Frame: Day 0, day 14, week 6
percentage of circulating T cells expressing immune checkpoint molecules as well as the intracellular transcription factor TCF1 and mean fluorescence intensity (MFI) of this expression in each cell sub-population
Day 0, day 14, week 6

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Aspergillus/Mucorales FLUOROSPOT
Time Frame: Day 0, week 6
Qualitative and quantitative measure of specific T cells IFN-γ/IL-2/IL-5/IL-17 production by FLUOROSPOT after antigenic stimulation in absence and presence of an anti-PD1 antibody in patients with invasive aspergillosis or mucormycosis
Day 0, week 6
T cell proliferation
Time Frame: Day 0, week 6
T cell proliferation test after antigenic stimulation
Day 0, week 6
Immune checkpoint expression on monocytes
Time Frame: Day 0, day 14, week 6
Percentage of circulating monocytes expressing immune checkpoint molecules
Day 0, day 14, week 6

Collaborators and Investigators

This is where you will find people and organizations involved with this study.

Investigators

  • Study Chair: Fanny LANTERNIER, MD, PhD, APHP

Study record dates

These dates track the progress of study record and summary results submissions to ClinicalTrials.gov. Study records and reported results are reviewed by the National Library of Medicine (NLM) to make sure they meet specific quality control standards before being posted on the public website.

Study Major Dates

Study Start (Estimated)

March 1, 2024

Primary Completion (Estimated)

December 1, 2025

Study Completion (Estimated)

March 1, 2026

Study Registration Dates

First Submitted

February 22, 2024

First Submitted That Met QC Criteria

February 22, 2024

First Posted (Actual)

February 29, 2024

Study Record Updates

Last Update Posted (Actual)

February 29, 2024

Last Update Submitted That Met QC Criteria

February 22, 2024

Last Verified

February 1, 2024

More Information

Terms related to this study

Plan for Individual participant data (IPD)

Plan to Share Individual Participant Data (IPD)?

NO

Drug and device information, study documents

Studies a U.S. FDA-regulated drug product

No

Studies a U.S. FDA-regulated device product

No

This information was retrieved directly from the website clinicaltrials.gov without any changes. If you have any requests to change, remove or update your study details, please contact register@clinicaltrials.gov. As soon as a change is implemented on clinicaltrials.gov, this will be updated automatically on our website as well.

Clinical Trials on Invasive Fungal Disease

3
Subscribe