Repeat Immunoadsorption Post Covid ME/CFS

November 21, 2022 updated by: Carmen Scheibenbogen, Charite University, Berlin, Germany

Observational Study of Repeat Immunoadsorption (RIA) in Post Covid ME/CFS Patients With Elevated ß2 Adrenergic Receptor Autoantibodies

The evidence for an autoimmune etiology in postinfectious myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is growing. The investigators observed in a not yet published study that in ME/CFS triggered by COVID, similar to ME/CFS after other infections, there is a close correlation of ß2 adrenergic receptor (ß2R) autoantibodies with symptom severity.

Immunoadsorption (IA) to remove autoantibodies has been used successfully in many autoantibody-mediated diseases. The investigators have already performed two proof of concept studies of IA in postinfectious ME/CFS with elevated ß2R antibodies, which resulted in improvement in most patients. This observational study aims to assess symptom outcome and functional ability in 20 patients with Post-COVID Syndrome (PCS) meeting ME/CFS diagnostic criteria with elevated ß2R antibodies undergoing antibody depletion by IA. The study will be conducted as a non-interventional observational study. IA with Miltenyi's TheraSorb® column in PCS will be performed in the approved use. Patients who have symptom improvement after the 1st IA will receive two additional IAs at 3 and 6 months, which will also be documented.

The results of this observational study will provide the basis for a randomized controlled clinical trial (RCT) combining IA with B-cell depletion therapy preferentially with Obinutuzumab.

Study Overview

Status

Recruiting

Conditions

Intervention / Treatment

Detailed Description

Studies indicate an overlap between post-COVID syndrome (PCS) and ME/CFS (Kedor et al., 2022). Post-infectious ME/CFS (ICD-10 code G93.3) is a complex and severely disabling disease with no approved treatment and thus, a very high medical need. Following an acute infection, patients suffer from severe central and muscle fatigue, chronic pain, cognitive impairment, and immune and autonomic dysfunction. The key symptom of ME/CFS is exertion intolerance with minor exertion resulting in symptom aggravation, also called post-exertional malaise (PEM), lasting until the next day up to weeks.

Various pathogens are known to induce post-infectious ME/CFS, including the Epstein-Barr virus and SARS-CoV-2 (Sotzny, 2018). The investigators' ongoing Charité PA-COVID study (Berlin prospective COVID-19 patient cohort) and other reports show that SARS-CoV-2 triggers ME/CFS in a subset of mostly younger patients without preexisting comorbidities (Kedor et al., 2022). Due to the rapid increase of COVID-19 incidence worldwide, the prevalence of post-COVID-19 ME/CFS will likely be a substantial problem for health care and society.

The evidence for an autoimmune etiology in post-infectious ME/CFS is growing (reviewed in Sotzny et al., 2018). Also, COVID-19 is a disease with a high risk for autoimmunity and developing ME/CFS due to the infection of many different cell types and the severity of immune activation. There is evidence from several studies from the investigators and other groups that natural regulatory autoantibodies targeting G protein-coupled receptors (GPCR) are involved in the pathogenesis of various autoimmune diseases. The investigators and others described enhanced levels and dysfunction of ß2R autoantibodies in ME/CFS and their correlation with the severity of key clinical symptoms (Freitag et al., 2021). The investigators' working hypothesis is that GPCR-specific autoantibodies with altered binding affinity or epitope specificity lead to immune dysregulation and autonomous dysfunction, and play a significant role in the pathomechanism of ME/CFS (Wirth et al., 2020). The investigators observed -- in a not yet published study -- a close correlation of ß2R antibodies with symptom severity in ME/CFS after COVID, similar to ME/CFS after other infections (Freitag et al., 2021). Two small proof-of-concept studies with immunoadsorption (IA) in patients with ME/CFS after other infections have shown improvements in symptoms in most patients (Scheibenbogen et al., 2018; Toelle et al., 2020).

In this observational non-controlled trial, PCS ME/CFS patients who receive IA will be evaluated for clinical efficacy, a decrease in autoantibodies, and a change of biomarkers. IA with Miltenyi's TheraSorb® column will be performed in the approved use. Patients who have symptom improvement after the 1st IA will receive two additional IAs at 3 and 6 months, which will also be documented. Symptom severity will be assessed by online questionnaires and at repeated time points.

Patients will be recruited for IA from the investigators' ongoing observational study with a follow-up of at least six months without disease improvement (Kedor et al., 2022). Patients have received a comprehensive diagnostic assessment to exclude other diseases, central nervous system, or organ comorbidity. ß2R autoantibodies in serum are determined by ELISA (Celltrend). Within four weeks prior to IA, patients will be assessed for eligibility for study participation by clinical investigation, laboratory analysis, and symptom questionnaires. The first ten patients will be treated end of August - mid of October and analyzed for efficacy until the beginning of December 2022 with a grant from the Weidenhammer-Zoebele foundation. The subsequent ten patients will be treated and analyzed with funding from the Federal Ministry of Education and Research(BMBF). These ten patients will also receive vessel diagnostics before (within four weeks) and four weeks after the first IA. In all 20 patients, blood will be collected the week before and four weeks after the first IA. All visits and treatments will take place in outpatient clinics.

Antibody depletion in PCS using IA has yet to be investigated in a clinical trial. The results of this observational study will provide the basis for a RCT using IA and, in responders, consecutive B-cell depleting therapy with an anti-CD20 monoclonal antibody versus placebo.

Study Type

Observational

Enrollment (Anticipated)

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 Locations

      • Berlin, Germany, 10117
        • Recruiting
        • Charité - Universitätsmedizin Berlin
        • 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

16 years to 63 years (Adult, Older Adult)

Accepts Healthy Volunteers

No

Genders Eligible for Study

All

Sampling Method

Non-Probability Sample

Study Population

Patients presenting at the investigators' outpatient clinic who meet the CCC for ME/CFS (PEM for at least 14h or longer) and who are found to have elevated ß2R autoantibodies will be offered an IA and participation in this observational study. Also, PCS patients who do not meet CCC criteria due to shorter PEM (< 14h) but meet Systemic Exertion Intolerance Disease criteria may receive IA and be enrolled in the study.

Description

Inclusion Criteria:

  • Consenting patients aged 18-65 years with a diagnosis of Post-COVID ME/CFS according to the Canadian Consensus Criteria (CCC) or fulfilling CCC with exertion intolerance with symptom worsening (post exertional malaise = PEM) duration of less than 14 hours (thus fulfilling Institute of Medicine criteria)
  • Evidence of COVID infection at disease onset (PCR) or N-IgG
  • Detection of autoantibodies (elevated ß2 receptor adrenergic autoantibodies)
  • Immunoadsorption with the TheraSorb® column for 5 days
  • Health insurance

Exclusion Criteria:

  • Lack of willingness to store pseudonymized disease data as part of the study
  • Pregnancy
  • Other illnesses that do not allow the diagnosis of PCS to be made with certainty (e.g., heart failure, lung disease, severe depression, cancer)
  • Acute infection (COVID, HIV, hepatitis)
  • Severe fatigue disease with bedriddenness

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

  • Observational Models: Cohort
  • Time Perspectives: Prospective

What is the study measuring?

Primary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Improvement in Physical Function (PF) as measured by the Short Form 36 Health Survey Questionnaire (SF-36)
Time Frame: 4 weeks after first IA
The Short Form 36 Health Survey (SF-36) is an established and widely used health-related quality of life measure. The Physical Function (PF) domain asks patients to report limitations on ten mobility activities, such as walking specified distances, carrying groceries, and bathing or dressing. Scores are weighted and transformed into a scale ranging from 0 (greatest possible health restrictions, i.e., severe disability) to 100 (no health restrictions). An intra-patient change of 10 points in SF-36-PF from baseline to week four is considered clinically meaningful.
4 weeks after first IA

Secondary Outcome Measures

Outcome Measure
Measure Description
Time Frame
Efficacy of repeat immunoadsorptions (IA) on Physical Function (PF) as measured by the Short Form 36 Health Survey Questionnaire (SF-36)
Time Frame: 3 - 6 months after first IA
Patients who have meaningful improvements in PF after the first IA, will receive two additional IAs at 3 and 6 months. Enhanced efficacy of repeat IAs on PF as measured by the SF-36 will be documented and assessed.
3 - 6 months after first IA
Response duration of repeat immunoadsorptions (IA) on Physical Function (PF) as measured by the Short Form 36 Health Survey Questionnaire (SF-36)
Time Frame: 3 - 6 months after first IA
Additive effect of repeat IA to increase response duration after each IA cycle. Response duration assessed by SF-36 PF (> 10 points).
3 - 6 months after first IA
Improvement in ability to work in daily hours
Time Frame: 12 months after last IA
Number of patients with an intra-patient change in his/her/their ability to work in daily hours from 6 months before first IA to 12 months after last IA.
12 months after last IA
Improvement in physical and mental fatigue as measured by the Chalder Fatigue Scale
Time Frame: 4 weeks after first IA; 6 and 12 months after last IA
The Chalder Fatigue Scale measures the extent and severity of tiredness and has been used in multiple randomized trials of behavioral interventions in patients with ME/CFS. Each of the 11 items is answered on a 4-point scale with an overall score ranging from 0 (asymptomatic) to 33 (maximum symptomology). Intra-patient change in physical and mental fatigue from baseline to follow-up points will be documented as indexed by the Chalder Fatigue Scale.
4 weeks after first IA; 6 and 12 months after last IA
Improvement in fatigue as measured by the Fatigue Severity Score (FSS)
Time Frame: 4 weeks after first IA; 6 and 12 months after last IA
The Fatigue Severity Scale (FSS) is a 9-item scale that measures the severity of fatigue and its effect on a person's activities and lifestyle. Answers are scored on a seven-point scale (1 = strongly disagree; 7 = strongly agree). Thus, the minimum score is 9 (no fatigue), and the highest is 63 (heavy fatigue). Intra-patient change in fatigue severity from baseline to follow-up points will be documented as indexed by the FSS.
4 weeks after first IA; 6 and 12 months after last IA
Improvement in symptoms of myalgic encephalomyelitis (ME)/ chronic fatigue syndrome (CFS) as measured by Canadian Consensus Criteria (CCC) Symptom Score
Time Frame: 4 weeks after first IA; 6 and 12 months after last IA
The Canadian Consensus Criteria (CCC) Symptom Score quantifies ME/CFS symptoms. Its score ranges from 1 (no symptoms) to 10 (extreme symptoms). Intra-patient change in ME/CFS symptoms from baseline to follow-up points will be documented as indexed by the CCC Symptom Score.
4 weeks after first IA; 6 and 12 months after last IA
Improvement in symptoms of autonomic dysfunction as measured by the Composite Autonomic Symptom Score (COMPASS-31)
Time Frame: 4 weeks after first IA; 6 and 12 months after last IA
The Composite Autonomic Symptom Score (COMPASS-31) is a refined, internally consistent, and markedly abbreviated quantitative measure of autonomic symptoms. It is based on the original Autonomic Symptom Profile (ASP) and COMPASS, applies a much-simplified scoring algorithm, and is suitable for widespread use in autonomic research and practice. It evaluates six domains of autonomic function: orthostatic intolerance, vasomotor, secretomotor, gastrointestinal, bladder, and pupillomotor domains. The score ranges from 0 (no symptoms) to 100 (strong autonomic dysfunction). Intra-patient change in autonomic dysfunction from baseline to follow-up points will be documented as indexed by the COMPASS-31.
4 weeks after first IA; 6 and 12 months after last IA
Improvement in hand grip strength
Time Frame: 4 weeks after first IA; 6 and 12 months after last IA
Intra-patient change in hand grip strength from baseline to follow-up points assessed using the Handgrip-Muscle-Fatigue Test
4 weeks after first IA; 6 and 12 months after last IA
Improvement of vascular regulation as indexed by the reactive hyperemia index (RHI) assessed using Endopat.
Time Frame: 6 and 12 months after last IA
Intra-patient change in endothelial cell function from baseline to follow-up points assessed using Endopat
6 and 12 months after last IA
Improvement in symptoms of orthostatic intolerance assessed using the Passive Standing Test
Time Frame: 4 weeks after first IA; 6 and 12 months after last IA
Intra-patient change in orthostatic intolerance from baseline to follow-up points assessed using the Passive Standing Test.
4 weeks after first IA; 6 and 12 months after last IA
Decrease in ß2R autoantibodies and soluble biomarkers of disease
Time Frame: 4 weeks after first IA; 6 and 12 months after last IA
Intra-patient change in ß2R autoantibodies and soluble biomarkers of disease from baseline to follow-up points assessed
4 weeks after first IA; 6 and 12 months after last IA
Verification of tolerability
Time Frame: 4 weeks after last IA
Number of patients tolerating the IA treatment.
4 weeks after last IA

Collaborators and Investigators

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

Publications and helpful links

The person responsible for entering information about the study voluntarily provides these publications. These may be about anything related to the study.

General Publications

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 (Actual)

August 28, 2022

Primary Completion (Anticipated)

August 31, 2023

Study Completion (Anticipated)

January 30, 2025

Study Registration Dates

First Submitted

November 14, 2022

First Submitted That Met QC Criteria

November 21, 2022

First Posted (Actual)

November 29, 2022

Study Record Updates

Last Update Posted (Actual)

November 29, 2022

Last Update Submitted That Met QC Criteria

November 21, 2022

Last Verified

November 1, 2022

More Information

Terms related to this study

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 Post-COVID ME/CFS

Clinical Trials on IA with TheraSorb ® column

3
Subscribe