Novel receptor-derived cyclopeptides to treat heart failure caused by anti-β1-adrenoceptor antibodies in a human-analogous rat model

Standard

Novel receptor-derived cyclopeptides to treat heart failure caused by anti-β1-adrenoceptor antibodies in a human-analogous rat model. / Boivin, Valérie; Beyersdorf, Niklas; Palm, Dieter; Nikolaev, Viacheslav O; Schlipp, Angela; Müller, Justus; Schmidt, Doris; Kocoski, Vladimir; Kerkau, Thomas; Hünig, Thomas; Ertl, Georg; Lohse, Martin J; Jahns, Roland.

in: PLOS ONE, Jahrgang 10, Nr. 2, 20.02.2015, S. e0117589.

Publikationen: SCORING: Beitrag in Fachzeitschrift/ZeitungSCORING: ZeitschriftenaufsatzForschungBegutachtung

Harvard

Boivin, V, Beyersdorf, N, Palm, D, Nikolaev, VO, Schlipp, A, Müller, J, Schmidt, D, Kocoski, V, Kerkau, T, Hünig, T, Ertl, G, Lohse, MJ & Jahns, R 2015, 'Novel receptor-derived cyclopeptides to treat heart failure caused by anti-β1-adrenoceptor antibodies in a human-analogous rat model', PLOS ONE, Jg. 10, Nr. 2, S. e0117589. https://doi.org/10.1371/journal.pone.0117589

APA

Boivin, V., Beyersdorf, N., Palm, D., Nikolaev, V. O., Schlipp, A., Müller, J., Schmidt, D., Kocoski, V., Kerkau, T., Hünig, T., Ertl, G., Lohse, M. J., & Jahns, R. (2015). Novel receptor-derived cyclopeptides to treat heart failure caused by anti-β1-adrenoceptor antibodies in a human-analogous rat model. PLOS ONE, 10(2), e0117589. https://doi.org/10.1371/journal.pone.0117589

Vancouver

Bibtex

@article{87f66d1f06f448baa159b889b38d2c21,
title = "Novel receptor-derived cyclopeptides to treat heart failure caused by anti-β1-adrenoceptor antibodies in a human-analogous rat model",
abstract = "Despite recent therapeutic advances the prognosis of heart failure remains poor. Recent research suggests that heart failure is a heterogeneous syndrome and that many patients have stimulating auto-antibodies directed against the second extracellular loop of the β1 adrenergic receptor (β1EC2). In a human-analogous rat model such antibodies cause myocyte damage and heart failure. Here we used this model to test a novel antibody-directed strategy aiming to prevent and/or treat antibody-induced cardiomyopathy. To generate heart failure, we immunised n = 76/114 rats with a fusion protein containing the human β1EC2 (amino-acids 195-225) every 4 weeks; n = 38/114 rats were control-injected with 0.9% NaCl. Intravenous application of a novel cyclic peptide mimicking β1EC2 (β1EC2-CP, 1.0 mg/kg every 4 weeks) or administration of the β1-blocker bisoprolol (15 mg/kg/day orally) was initiated either 6 weeks (cardiac function still normal, prevention-study, n = 24 (16 treated vs. 8 untreated)) or 8.5 months after the 1st immunisation (onset of cardiomyopathy, therapy-study, n = 52 (40 treated vs. 12 untreated)); n = 8/52 rats from the therapy-study received β1EC2-CP/bisoprolol co-treatment. We found that β1EC2-CP prevented and (alone or as add-on drug) treated antibody-induced cardiac damage in the rat, and that its efficacy was superior to mono-treatment with bisoprolol, a standard drug in heart failure. While bisoprolol mono-therapy was able to stop disease-progression, β1EC2-CP mono-therapy -or as an add-on to bisoprolol- almost fully reversed antibody-induced cardiac damage. The cyclo¬peptide acted both by scavenging free anti-β1EC2-antibodies and by targeting β1EC2-specific memory B-cells involved in antibody-production. Our model provides the basis for the clinical translation of a novel double-acting therapeutic strategy that scavenges harmful anti-β1EC2-antibodies and also selectively depletes memory B-cells involved in the production of such antibodies. Treatment with immuno-modulating cyclopeptides alone or as an add-on to β1-blockade represents a promising new therapeutic option in immune-mediated heart failure.",
keywords = "Animals, Antibodies, B-Lymphocytes, Bisoprolol, CD4-Positive T-Lymphocytes, Disease Models, Animal, Disease Progression, Echocardiography, Heart Failure, Humans, Male, Myocardium, Peptides, Cyclic, Rats, Rats, Inbred Lew, Receptors, Adrenergic, beta-1",
author = "Val{\'e}rie Boivin and Niklas Beyersdorf and Dieter Palm and Nikolaev, {Viacheslav O} and Angela Schlipp and Justus M{\"u}ller and Doris Schmidt and Vladimir Kocoski and Thomas Kerkau and Thomas H{\"u}nig and Georg Ertl and Lohse, {Martin J} and Roland Jahns",
year = "2015",
month = feb,
day = "20",
doi = "10.1371/journal.pone.0117589",
language = "English",
volume = "10",
pages = "e0117589",
journal = "PLOS ONE",
issn = "1932-6203",
publisher = "Public Library of Science",
number = "2",

}

RIS

TY - JOUR

T1 - Novel receptor-derived cyclopeptides to treat heart failure caused by anti-β1-adrenoceptor antibodies in a human-analogous rat model

AU - Boivin, Valérie

AU - Beyersdorf, Niklas

AU - Palm, Dieter

AU - Nikolaev, Viacheslav O

AU - Schlipp, Angela

AU - Müller, Justus

AU - Schmidt, Doris

AU - Kocoski, Vladimir

AU - Kerkau, Thomas

AU - Hünig, Thomas

AU - Ertl, Georg

AU - Lohse, Martin J

AU - Jahns, Roland

PY - 2015/2/20

Y1 - 2015/2/20

N2 - Despite recent therapeutic advances the prognosis of heart failure remains poor. Recent research suggests that heart failure is a heterogeneous syndrome and that many patients have stimulating auto-antibodies directed against the second extracellular loop of the β1 adrenergic receptor (β1EC2). In a human-analogous rat model such antibodies cause myocyte damage and heart failure. Here we used this model to test a novel antibody-directed strategy aiming to prevent and/or treat antibody-induced cardiomyopathy. To generate heart failure, we immunised n = 76/114 rats with a fusion protein containing the human β1EC2 (amino-acids 195-225) every 4 weeks; n = 38/114 rats were control-injected with 0.9% NaCl. Intravenous application of a novel cyclic peptide mimicking β1EC2 (β1EC2-CP, 1.0 mg/kg every 4 weeks) or administration of the β1-blocker bisoprolol (15 mg/kg/day orally) was initiated either 6 weeks (cardiac function still normal, prevention-study, n = 24 (16 treated vs. 8 untreated)) or 8.5 months after the 1st immunisation (onset of cardiomyopathy, therapy-study, n = 52 (40 treated vs. 12 untreated)); n = 8/52 rats from the therapy-study received β1EC2-CP/bisoprolol co-treatment. We found that β1EC2-CP prevented and (alone or as add-on drug) treated antibody-induced cardiac damage in the rat, and that its efficacy was superior to mono-treatment with bisoprolol, a standard drug in heart failure. While bisoprolol mono-therapy was able to stop disease-progression, β1EC2-CP mono-therapy -or as an add-on to bisoprolol- almost fully reversed antibody-induced cardiac damage. The cyclo¬peptide acted both by scavenging free anti-β1EC2-antibodies and by targeting β1EC2-specific memory B-cells involved in antibody-production. Our model provides the basis for the clinical translation of a novel double-acting therapeutic strategy that scavenges harmful anti-β1EC2-antibodies and also selectively depletes memory B-cells involved in the production of such antibodies. Treatment with immuno-modulating cyclopeptides alone or as an add-on to β1-blockade represents a promising new therapeutic option in immune-mediated heart failure.

AB - Despite recent therapeutic advances the prognosis of heart failure remains poor. Recent research suggests that heart failure is a heterogeneous syndrome and that many patients have stimulating auto-antibodies directed against the second extracellular loop of the β1 adrenergic receptor (β1EC2). In a human-analogous rat model such antibodies cause myocyte damage and heart failure. Here we used this model to test a novel antibody-directed strategy aiming to prevent and/or treat antibody-induced cardiomyopathy. To generate heart failure, we immunised n = 76/114 rats with a fusion protein containing the human β1EC2 (amino-acids 195-225) every 4 weeks; n = 38/114 rats were control-injected with 0.9% NaCl. Intravenous application of a novel cyclic peptide mimicking β1EC2 (β1EC2-CP, 1.0 mg/kg every 4 weeks) or administration of the β1-blocker bisoprolol (15 mg/kg/day orally) was initiated either 6 weeks (cardiac function still normal, prevention-study, n = 24 (16 treated vs. 8 untreated)) or 8.5 months after the 1st immunisation (onset of cardiomyopathy, therapy-study, n = 52 (40 treated vs. 12 untreated)); n = 8/52 rats from the therapy-study received β1EC2-CP/bisoprolol co-treatment. We found that β1EC2-CP prevented and (alone or as add-on drug) treated antibody-induced cardiac damage in the rat, and that its efficacy was superior to mono-treatment with bisoprolol, a standard drug in heart failure. While bisoprolol mono-therapy was able to stop disease-progression, β1EC2-CP mono-therapy -or as an add-on to bisoprolol- almost fully reversed antibody-induced cardiac damage. The cyclo¬peptide acted both by scavenging free anti-β1EC2-antibodies and by targeting β1EC2-specific memory B-cells involved in antibody-production. Our model provides the basis for the clinical translation of a novel double-acting therapeutic strategy that scavenges harmful anti-β1EC2-antibodies and also selectively depletes memory B-cells involved in the production of such antibodies. Treatment with immuno-modulating cyclopeptides alone or as an add-on to β1-blockade represents a promising new therapeutic option in immune-mediated heart failure.

KW - Animals

KW - Antibodies

KW - B-Lymphocytes

KW - Bisoprolol

KW - CD4-Positive T-Lymphocytes

KW - Disease Models, Animal

KW - Disease Progression

KW - Echocardiography

KW - Heart Failure

KW - Humans

KW - Male

KW - Myocardium

KW - Peptides, Cyclic

KW - Rats

KW - Rats, Inbred Lew

KW - Receptors, Adrenergic, beta-1

U2 - 10.1371/journal.pone.0117589

DO - 10.1371/journal.pone.0117589

M3 - SCORING: Journal article

C2 - 25700031

VL - 10

SP - e0117589

JO - PLOS ONE

JF - PLOS ONE

SN - 1932-6203

IS - 2

ER -