search
Back to results

Iron Substitution With Ferric Carboxymaltose as Treatment Strategy for Heart Failure Patients With Preserved Ejection Fraction (IRON-HFpEF)

Primary Purpose

Heart Failure With Normal Ejection Fraction, Iron-deficiency

Status
Withdrawn
Phase
Phase 4
Locations
Study Type
Interventional
Intervention
Ferric carboxymaltose
Placebo
Sponsored by
Cantonal Hosptal, Baselland
About
Eligibility
Locations
Arms
Outcomes
Full info

About this trial

This is an interventional treatment trial for Heart Failure With Normal Ejection Fraction focused on measuring Exercise tolerance, iron substitution

Eligibility Criteria

18 Years - undefined (Adult, Older Adult)All SexesDoes not accept healthy volunteers

Inclusion Criteria:

  • Informed consent as documented by signature
  • NYHA functional classes II-III
  • Signs and symptoms of chronic HF, such as:
  • Dyspnea
  • Paroxysmal nocturnal dyspnea
  • Reduced exercise tolerance
  • Fatigue
  • Extended recovery after exercising
  • Peripheral edema (lower leg, ankle)
  • EF (ejection fraction) >50%
  • Structural or functional changes in echocardiography:
  • Left atrial volume index (LAVI) >34 ml/m2 OR
  • Left ventricular mass index (LVMI) >115 g/m2 (men), >95 g/m2 (women) OR
  • E/E' (ratio between mitral peak velocity of early filling (E) to early diastolic mitral annular velocity (E')) >13 AND mean E' septal and lateral wall <9 cm/s
  • NT-proBNP >125 pg/ml
  • At least 4 weeks on stable medical treatment or without signs and symptoms of cardiac decompensation
  • Iron deficiency defined as:
  • Ferritin <100 ng/ml OR
  • Ferritin <300 ng/ml with a transferrin saturation (TSAT) <20%

Exclusion Criteria:

  • Age <18 years
  • Pregnancy or lactation
  • Life-expectancy <6 months
  • Planned cardiac interventions in the following 6 months
  • Unstable angina pectoris
  • Uncontrolled brady- or tachyarrhythmia
  • Severe uncorrected valvular heart disease
  • Paroxysmal atrial fibrillation
  • Clinically significant concomitant disease states (e.g. hypertension grades 2-3 (>160/100 mmHg), severe renal failure (GFR <30 ml/min/1.73m2), hepatic dysfunction (ALT or AST >3x upper limit of normal, chronic obstructive pulmonary disease (COPD) grades III-IV)
  • On-going cancer treatment
  • Significant musculoskeletal disease limiting exercise tolerance
  • Active infection
  • Immunosuppressive medical therapy
  • Earlier hypersensitivity to parenteral iron preparation
  • Anemia and iron deficiency due to active and/or chronic bleeding
  • Blood transfusion within the previous 30 days
  • Red cell, folate and vitamin B12 deficiency
  • Known or suspected non-compliance, drug or alcohol abuse
  • Inability to follow the procedures of the study, e.g. due to insufficient language skills, psychological disorders, dementia, etc.
  • Participation in another intervention study
  • Enrolment of the investigators, their family members, and other persons involved in the study procedures
  • Hemoglobin < 120 ng/ml in male patients or < 110 ng/ml in female patients

Sites / Locations

    Arms of the Study

    Arm 1

    Arm 2

    Arm Type

    Experimental

    Placebo Comparator

    Arm Label

    Iron substitution

    Placebo

    Arm Description

    Iron deficiency status will be assessed at the baseline visit (Day 0) as well as after 6 weeks of iron substitution (Week 6). The study drug will be given as FCM solution (Ferinject®, Vifor Pharma AG, Villars-sur-Glâne, Switzerland) by intravenous injection. Infusions of 10 or 20 mL (which is the amount of FCM that is equivalent to 500 or 1000 mg of iron, respectively) will be administered in ≥6 minutes diluted in ≈100 mL of sterile 0.9% sodium chloride solution (NaCl) for 10 mL, or in ≥15 minutes diluted in ≈200 mL for 20 mL. Dosing will be based on screening Hb level and weight, rather than on ferritin and TSAT results. On Day 0 (baseline visit), patients with Hb ≤14 g/dL, both <70 kg and >70 kg will receive 1000 mg FCM (20 mL), whereas patients with Hb >14g/dL will receive 500 mg FCM (10 mL).

    Patients in the control group will receive a placebo solution administered as normal saline (0.9% weight/volume (w/v) NaCl) by intravenous injection as per the instructions for active treatment.

    Outcomes

    Primary Outcome Measures

    Change in peak oxygen uptake (VO2peak)
    Change of VO2peak will be measured by spiroergometry at the baseline and post-intervention visit.

    Secondary Outcome Measures

    Change in ventriculo-arterial coupling (VAC)
    VAC is defined as the ratio of arterial elastance (Ea) and end-systolic elastance (Ees) and will be approximated echocardiographically based on the method described by Antonini-Canterin et al.
    Change in arteriovenous oxygen difference (Da-vO2)
    Da-vO2 will be calculated using the Fick Principle
    Change in pulse wave velocity (PWV)
    PWV will be measured at the same time as the echocardiographic parameters with the VaSera VS-2000 Vascular Screening System (Fukuda Denshi Co. Ltd, Tokyo, Japan) and evaluated by experienced blinded members of the study team.
    Change in New York Heart Association (NYHA) functional class
    NYHA functional class will be determined according to the New York Heart Association classification.
    Change in habitual physical activity
    Habitual physical activity will be measured by an accelerometer over a period of 14 consecutive days for 24 hours per day following baseline and post-intervention visits. Patients will wear a waterproof micro-electromechanical triaxial activity bracelet on the non-dominant wrist (GeneActiv, Activinsights Ltd, Kimbolton, Cambridgeshire, UK) to assess physical activity intensity (light, moderate, vigorous) and periods of inactivity, sleep and wake.
    Change in body composition measured by body mass index (BMI)
    BMI will be calculated from measured height in meters and weight in kilograms. Weight and height will be combined to report BMI in kg/m^2.
    Change in baseline body composition measured by waist to hip ratio (WHR)
    WHR will be calculated from measured waist circumference (WC) and hip circumference (HC) in centimetres. WC will be divided by HC to report WHR.
    Change in total hemoglobin mass (tHb-mass)
    tHB-mass will be measured with the carbon monoxide (CO)-rebreathing method
    Change in quality of life (QoL) by the 36-Item Short Form Health Survey (SF-36)
    The SF-36 consists of 36 items, which are formatted as binary questions or as semantic 6-point differential scales. It refers to the past 4 weeks and includes 9 content areas concerning vitality, general health perception, physical functioning, social functioning, role limitations (emotional/physical problems), pain, mental health and health change.
    Change in baseline quality of life (QoL) by the Kansas City Cardiomyopathy Questionnaire (KCCQ) at 12 weeks.
    The KCCQ consists of 15 items concerning overall symptoms, emotional, social and mental status within the past 2 weeks.
    Change in quality of life (QoL) by the Minnesota Living with Heart Failure Questionnaire (MLWHFQ) at 12 weeks.
    The MLWHFQ refers to the past 4 weeks and includes 21 questions on a 6-point scale with a maximum of 105 points (<24 good QoL, >45 poor QoL).

    Full Information

    First Posted
    July 5, 2019
    Last Updated
    July 25, 2022
    Sponsor
    Cantonal Hosptal, Baselland
    Collaborators
    Clinical Trial Unit, University Hospital Basel, Switzerland
    search

    1. Study Identification

    Unique Protocol Identification Number
    NCT05477498
    Brief Title
    Iron Substitution With Ferric Carboxymaltose as Treatment Strategy for Heart Failure Patients With Preserved Ejection Fraction
    Acronym
    IRON-HFpEF
    Official Title
    Iron Substitution With Ferric Carboxymaltose as Treatment Strategy for Heart Failure Patients With Preserved Ejection Fraction: A Prospective, Double-blind, Randomized, Placebo-controlled Trial
    Study Type
    Interventional

    2. Study Status

    Record Verification Date
    July 2022
    Overall Recruitment Status
    Withdrawn
    Why Stopped
    Principal Investigator left institution
    Study Start Date
    December 1, 2021 (Anticipated)
    Primary Completion Date
    August 31, 2022 (Anticipated)
    Study Completion Date
    December 31, 2022 (Anticipated)

    3. Sponsor/Collaborators

    Responsible Party, by Official Title
    Principal Investigator
    Name of the Sponsor
    Cantonal Hosptal, Baselland
    Collaborators
    Clinical Trial Unit, University Hospital Basel, Switzerland

    4. Oversight

    Studies a U.S. FDA-regulated Drug Product
    No
    Studies a U.S. FDA-regulated Device Product
    No
    Data Monitoring Committee
    Yes

    5. Study Description

    Brief Summary
    This study aims to investigate the effects of treatment with intravenous ferric carboxymaltose on exercise tolerance measured as VO2peak in patients with HFpEF and iron deficiency, compared to placebo.
    Detailed Description
    Iron deficiency is a common comorbidity associated with chronic heart failure (HF) in both, patients with preserved (HFpEF) and reduced ejection fraction (HFrEF), which has unfavorable clinical and prognostic effects. Previous studies have confirmed that HF patients with iron deficiency have a lower exercise tolerance than those without iron deficiency. In iron deficient patients with HFrEF, treatment with intravenous ferric carboxymaltose (FCM) improved symptoms, exercise tolerance and quality of life (QoL). Since the latest guidelines published by the European Society of Cardiology (ESC) in 2016, iron substitution is an official class IIa recommendation in HFrEF, while it has not yet been endorsed in the treatment guidelines for HFpEF. To date, no evidence is available on iron supplementation in HFpEF. Therefore, a clear rationale exists for examining the effects of correcting iron deficiency in this high-risk and steadily growing patient group. The proposed study will be a single-centre, prospective, double-blind, randomized, placebo-controlled trial in a primary care setting including 86 patients with stable HFpEF and iron deficiency. Participants will undergo three study visits: a baseline visit, a status control visit, and a post-intervention visit. At the baseline visit, measurements of exercise tolerance (using spiroergometry), laboratory parameters and disease-specific biomarkers (using blood samples), tHb-mass (using the carbon monoxide rebreathing method), cardiac and arterial vessel structure and function (using electrocardiogram, echocardiography and PVW), QoL (using 3 validated questionnaires), body composition (using BMI and WHR), and habitual physical activity (using a wrist-worn accelerometer) will be performed. Then, patients randomized to the treatment group will receive FCM (Vifor Pharma AG, Villars-sur-Glâne, Switzerland), whereas those in the control group will receive placebo. At week 6, iron deficiency status will be re-evaluated in all patients and, if necessary, another application of FCM or placebo will be administered, respectively. After the 12-week treatment period, the study measurements will be repeated in all patients (post-intervention visit) to investigate the effects of the intervention.

    6. Conditions and Keywords

    Primary Disease or Condition Being Studied in the Trial, or the Focus of the Study
    Heart Failure With Normal Ejection Fraction, Iron-deficiency
    Keywords
    Exercise tolerance, iron substitution

    7. Study Design

    Primary Purpose
    Treatment
    Study Phase
    Phase 4
    Interventional Study Model
    Parallel Assignment
    Model Description
    Intervention group: Active treatment with ferric carboxymaltose, given as diluted solution by intravenous injection Control group: Placebo, administered as normal saline 0.9%
    Masking
    ParticipantCare ProviderInvestigatorOutcomes Assessor
    Masking Description
    Each administration of the study drug will be carried out after completion of all applicable study related assessments. FCM is a dark brown solution and cannot be easily masked from placebo (0.9% saline). Therefore, unblinded study personnel (at least one study nurse) who will not be involved in any study procedures for efficacy or safety will be responsible for preparing the infusion and packing bag and tube in an opaque wrapping. Preparation and wrapping will take place in a different room to maintain subject blinding. Administration of the study drug will take place by blinded study personnel. The results of the central laboratory on iron deficiency status and Hb will be sent only to the unblinded study personnel, who will be responsible for evaluating these parameters for subsequent dosing and/or other intervention, if applicable.
    Allocation
    Randomized
    Enrollment
    0 (Actual)

    8. Arms, Groups, and Interventions

    Arm Title
    Iron substitution
    Arm Type
    Experimental
    Arm Description
    Iron deficiency status will be assessed at the baseline visit (Day 0) as well as after 6 weeks of iron substitution (Week 6). The study drug will be given as FCM solution (Ferinject®, Vifor Pharma AG, Villars-sur-Glâne, Switzerland) by intravenous injection. Infusions of 10 or 20 mL (which is the amount of FCM that is equivalent to 500 or 1000 mg of iron, respectively) will be administered in ≥6 minutes diluted in ≈100 mL of sterile 0.9% sodium chloride solution (NaCl) for 10 mL, or in ≥15 minutes diluted in ≈200 mL for 20 mL. Dosing will be based on screening Hb level and weight, rather than on ferritin and TSAT results. On Day 0 (baseline visit), patients with Hb ≤14 g/dL, both <70 kg and >70 kg will receive 1000 mg FCM (20 mL), whereas patients with Hb >14g/dL will receive 500 mg FCM (10 mL).
    Arm Title
    Placebo
    Arm Type
    Placebo Comparator
    Arm Description
    Patients in the control group will receive a placebo solution administered as normal saline (0.9% weight/volume (w/v) NaCl) by intravenous injection as per the instructions for active treatment.
    Intervention Type
    Drug
    Intervention Name(s)
    Ferric carboxymaltose
    Other Intervention Name(s)
    Ferinject®
    Intervention Description
    Application of FCM solution (Ferinject®, Vifor Pharma AG, Villars-sur-Glâne, Switzerland) by intravenous injection.
    Intervention Type
    Drug
    Intervention Name(s)
    Placebo
    Other Intervention Name(s)
    NaCl 0.9%
    Intervention Description
    Application of placebo solution administered as normal saline (0.9% weight/volume (w/v) NaCl) by intravenous injection as per the instructions for active treatment.
    Primary Outcome Measure Information:
    Title
    Change in peak oxygen uptake (VO2peak)
    Description
    Change of VO2peak will be measured by spiroergometry at the baseline and post-intervention visit.
    Time Frame
    12 weeks
    Secondary Outcome Measure Information:
    Title
    Change in ventriculo-arterial coupling (VAC)
    Description
    VAC is defined as the ratio of arterial elastance (Ea) and end-systolic elastance (Ees) and will be approximated echocardiographically based on the method described by Antonini-Canterin et al.
    Time Frame
    12 weeks
    Title
    Change in arteriovenous oxygen difference (Da-vO2)
    Description
    Da-vO2 will be calculated using the Fick Principle
    Time Frame
    12 weeks
    Title
    Change in pulse wave velocity (PWV)
    Description
    PWV will be measured at the same time as the echocardiographic parameters with the VaSera VS-2000 Vascular Screening System (Fukuda Denshi Co. Ltd, Tokyo, Japan) and evaluated by experienced blinded members of the study team.
    Time Frame
    12 weeks
    Title
    Change in New York Heart Association (NYHA) functional class
    Description
    NYHA functional class will be determined according to the New York Heart Association classification.
    Time Frame
    12 weeks
    Title
    Change in habitual physical activity
    Description
    Habitual physical activity will be measured by an accelerometer over a period of 14 consecutive days for 24 hours per day following baseline and post-intervention visits. Patients will wear a waterproof micro-electromechanical triaxial activity bracelet on the non-dominant wrist (GeneActiv, Activinsights Ltd, Kimbolton, Cambridgeshire, UK) to assess physical activity intensity (light, moderate, vigorous) and periods of inactivity, sleep and wake.
    Time Frame
    12 weeks
    Title
    Change in body composition measured by body mass index (BMI)
    Description
    BMI will be calculated from measured height in meters and weight in kilograms. Weight and height will be combined to report BMI in kg/m^2.
    Time Frame
    12 weeks
    Title
    Change in baseline body composition measured by waist to hip ratio (WHR)
    Description
    WHR will be calculated from measured waist circumference (WC) and hip circumference (HC) in centimetres. WC will be divided by HC to report WHR.
    Time Frame
    12 weeks
    Title
    Change in total hemoglobin mass (tHb-mass)
    Description
    tHB-mass will be measured with the carbon monoxide (CO)-rebreathing method
    Time Frame
    12 weeks
    Title
    Change in quality of life (QoL) by the 36-Item Short Form Health Survey (SF-36)
    Description
    The SF-36 consists of 36 items, which are formatted as binary questions or as semantic 6-point differential scales. It refers to the past 4 weeks and includes 9 content areas concerning vitality, general health perception, physical functioning, social functioning, role limitations (emotional/physical problems), pain, mental health and health change.
    Time Frame
    12 weeks
    Title
    Change in baseline quality of life (QoL) by the Kansas City Cardiomyopathy Questionnaire (KCCQ) at 12 weeks.
    Description
    The KCCQ consists of 15 items concerning overall symptoms, emotional, social and mental status within the past 2 weeks.
    Time Frame
    12 weeks
    Title
    Change in quality of life (QoL) by the Minnesota Living with Heart Failure Questionnaire (MLWHFQ) at 12 weeks.
    Description
    The MLWHFQ refers to the past 4 weeks and includes 21 questions on a 6-point scale with a maximum of 105 points (<24 good QoL, >45 poor QoL).
    Time Frame
    12 weeks

    10. Eligibility

    Sex
    All
    Minimum Age & Unit of Time
    18 Years
    Accepts Healthy Volunteers
    No
    Eligibility Criteria
    Inclusion Criteria: Informed consent as documented by signature NYHA functional classes II-III Signs and symptoms of chronic HF, such as: Dyspnea Paroxysmal nocturnal dyspnea Reduced exercise tolerance Fatigue Extended recovery after exercising Peripheral edema (lower leg, ankle) EF (ejection fraction) >50% Structural or functional changes in echocardiography: Left atrial volume index (LAVI) >34 ml/m2 OR Left ventricular mass index (LVMI) >115 g/m2 (men), >95 g/m2 (women) OR E/E' (ratio between mitral peak velocity of early filling (E) to early diastolic mitral annular velocity (E')) >13 AND mean E' septal and lateral wall <9 cm/s NT-proBNP >125 pg/ml At least 4 weeks on stable medical treatment or without signs and symptoms of cardiac decompensation Iron deficiency defined as: Ferritin <100 ng/ml OR Ferritin <300 ng/ml with a transferrin saturation (TSAT) <20% Exclusion Criteria: Age <18 years Pregnancy or lactation Life-expectancy <6 months Planned cardiac interventions in the following 6 months Unstable angina pectoris Uncontrolled brady- or tachyarrhythmia Severe uncorrected valvular heart disease Paroxysmal atrial fibrillation Clinically significant concomitant disease states (e.g. hypertension grades 2-3 (>160/100 mmHg), severe renal failure (GFR <30 ml/min/1.73m2), hepatic dysfunction (ALT or AST >3x upper limit of normal, chronic obstructive pulmonary disease (COPD) grades III-IV) On-going cancer treatment Significant musculoskeletal disease limiting exercise tolerance Active infection Immunosuppressive medical therapy Earlier hypersensitivity to parenteral iron preparation Anemia and iron deficiency due to active and/or chronic bleeding Blood transfusion within the previous 30 days Red cell, folate and vitamin B12 deficiency Known or suspected non-compliance, drug or alcohol abuse Inability to follow the procedures of the study, e.g. due to insufficient language skills, psychological disorders, dementia, etc. Participation in another intervention study Enrolment of the investigators, their family members, and other persons involved in the study procedures Hemoglobin < 120 ng/ml in male patients or < 110 ng/ml in female patients
    Overall Study Officials:
    First Name & Middle Initial & Last Name & Degree
    Thomas Dieterle, MD
    Organizational Affiliation
    University Department of Internal Medicine, Cantonal Hospital Baselland
    Official's Role
    Principal Investigator

    12. IPD Sharing Statement

    Plan to Share IPD
    Yes
    IPD Sharing Plan Description
    Patient-level anonymised datasets can be requested after completion of all planned analyses and publications from the study centre (anticipated by mid 2022). Public access to the study protocol will be granted by publishing it in a scientific journal.

    Learn more about this trial

    Iron Substitution With Ferric Carboxymaltose as Treatment Strategy for Heart Failure Patients With Preserved Ejection Fraction

    We'll reach out to this number within 24 hrs