Human Engineered Heart Tissue Patches Remuscularize the Injured Heart in a Dose-Dependent Manner

Standard

Human Engineered Heart Tissue Patches Remuscularize the Injured Heart in a Dose-Dependent Manner. / Querdel, Eva; Reinsch, Marina; Castro, Liesa; Köse, Deniz; Bähr, Andrea; Reich, Svenja; Geertz, Birgit; Ulmer, Bärbel; Schulze, Mirja; Lemoine, Marc D; Krause, Tobias; Lemme, Marta; Sani, Jascha; Shibamiya, Aya; Stüdemann, Tim; Köhne, Maria; Bibra, Constantin von; Hornaschewitz, Nadja; Pecha, Simon; Nejahsie, Yusuf; Mannhardt, Ingra; Christ, Torsten; Reichenspurner, Hermann; Hansen, Arne; Klymiuk, Nikolai; Krane, M; Kupatt, C; Eschenhagen, Thomas; Weinberger, Florian.

In: CIRCULATION, Vol. 143, No. 20, 18.05.2021, p. 1991-2006.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Querdel, E, Reinsch, M, Castro, L, Köse, D, Bähr, A, Reich, S, Geertz, B, Ulmer, B, Schulze, M, Lemoine, MD, Krause, T, Lemme, M, Sani, J, Shibamiya, A, Stüdemann, T, Köhne, M, Bibra, CV, Hornaschewitz, N, Pecha, S, Nejahsie, Y, Mannhardt, I, Christ, T, Reichenspurner, H, Hansen, A, Klymiuk, N, Krane, M, Kupatt, C, Eschenhagen, T & Weinberger, F 2021, 'Human Engineered Heart Tissue Patches Remuscularize the Injured Heart in a Dose-Dependent Manner', CIRCULATION, vol. 143, no. 20, pp. 1991-2006. https://doi.org/10.1161/CIRCULATIONAHA.120.047904

APA

Querdel, E., Reinsch, M., Castro, L., Köse, D., Bähr, A., Reich, S., Geertz, B., Ulmer, B., Schulze, M., Lemoine, M. D., Krause, T., Lemme, M., Sani, J., Shibamiya, A., Stüdemann, T., Köhne, M., Bibra, C. V., Hornaschewitz, N., Pecha, S., ... Weinberger, F. (2021). Human Engineered Heart Tissue Patches Remuscularize the Injured Heart in a Dose-Dependent Manner. CIRCULATION, 143(20), 1991-2006. https://doi.org/10.1161/CIRCULATIONAHA.120.047904

Vancouver

Bibtex

@article{8adf46c7270b49588166835d70f70b89,
title = "Human Engineered Heart Tissue Patches Remuscularize the Injured Heart in a Dose-Dependent Manner",
abstract = "BACKGROUND: Human engineered heart tissue (EHT) transplantation represents a potential regenerative strategy for patients with heart failure and has been successful in preclinical models. Clinical application requires upscaling, adaptation to good manufacturing practices, and determination of the effective dose.METHODS: Cardiomyocytes were differentiated from 3 different human induced pluripotent stem cell lines including one reprogrammed under good manufacturing practice conditions. Protocols for human induced pluripotent stem cell expansion, cardiomyocyte differentiation, and EHT generation were adapted to substances available in good manufacturing practice quality. EHT geometry was modified to generate patches suitable for transplantation in a small-animal model and perspectively humans. Repair efficacy was evaluated at 3 doses in a cryo-injury guinea pig model. Human-scale patches were epicardially transplanted onto healthy hearts in pigs to assess technical feasibility.RESULTS: We created mesh-structured tissue patches for transplantation in guinea pigs (1.5×2.5 cm, 9-15×106 cardiomyocytes) and pigs (5×7 cm, 450×106 cardiomyocytes). EHT patches coherently beat in culture and developed high force (mean 4.6 mN). Cardiomyocytes matured, aligned along the force lines, and demonstrated advanced sarcomeric structure and action potential characteristics closely resembling human ventricular tissue. EHT patches containing ≈4.5, 8.5, 12×106, or no cells were transplanted 7 days after cryo-injury (n=18-19 per group). EHT transplantation resulted in a dose-dependent remuscularization (graft size: 0%-12% of the scar). Only high-dose patches improved left ventricular function (+8% absolute, +24% relative increase). The grafts showed time-dependent cardiomyocyte proliferation. Although standard EHT patches did not withstand transplantation in pigs, the human-scale patch enabled successful patch transplantation.CONCLUSIONS: EHT patch transplantation resulted in a partial remuscularization of the injured heart and improved left ventricular function in a dose-dependent manner in a guinea pig injury model. Human-scale patches were successfully transplanted in pigs in a proof-of-principle study.",
author = "Eva Querdel and Marina Reinsch and Liesa Castro and Deniz K{\"o}se and Andrea B{\"a}hr and Svenja Reich and Birgit Geertz and B{\"a}rbel Ulmer and Mirja Schulze and Lemoine, {Marc D} and Tobias Krause and Marta Lemme and Jascha Sani and Aya Shibamiya and Tim St{\"u}demann and Maria K{\"o}hne and Bibra, {Constantin von} and Nadja Hornaschewitz and Simon Pecha and Yusuf Nejahsie and Ingra Mannhardt and Torsten Christ and Hermann Reichenspurner and Arne Hansen and Nikolai Klymiuk and M Krane and C Kupatt and Thomas Eschenhagen and Florian Weinberger",
year = "2021",
month = may,
day = "18",
doi = "10.1161/CIRCULATIONAHA.120.047904",
language = "English",
volume = "143",
pages = "1991--2006",
journal = "CIRCULATION",
issn = "0009-7322",
publisher = "Lippincott Williams and Wilkins",
number = "20",

}

RIS

TY - JOUR

T1 - Human Engineered Heart Tissue Patches Remuscularize the Injured Heart in a Dose-Dependent Manner

AU - Querdel, Eva

AU - Reinsch, Marina

AU - Castro, Liesa

AU - Köse, Deniz

AU - Bähr, Andrea

AU - Reich, Svenja

AU - Geertz, Birgit

AU - Ulmer, Bärbel

AU - Schulze, Mirja

AU - Lemoine, Marc D

AU - Krause, Tobias

AU - Lemme, Marta

AU - Sani, Jascha

AU - Shibamiya, Aya

AU - Stüdemann, Tim

AU - Köhne, Maria

AU - Bibra, Constantin von

AU - Hornaschewitz, Nadja

AU - Pecha, Simon

AU - Nejahsie, Yusuf

AU - Mannhardt, Ingra

AU - Christ, Torsten

AU - Reichenspurner, Hermann

AU - Hansen, Arne

AU - Klymiuk, Nikolai

AU - Krane, M

AU - Kupatt, C

AU - Eschenhagen, Thomas

AU - Weinberger, Florian

PY - 2021/5/18

Y1 - 2021/5/18

N2 - BACKGROUND: Human engineered heart tissue (EHT) transplantation represents a potential regenerative strategy for patients with heart failure and has been successful in preclinical models. Clinical application requires upscaling, adaptation to good manufacturing practices, and determination of the effective dose.METHODS: Cardiomyocytes were differentiated from 3 different human induced pluripotent stem cell lines including one reprogrammed under good manufacturing practice conditions. Protocols for human induced pluripotent stem cell expansion, cardiomyocyte differentiation, and EHT generation were adapted to substances available in good manufacturing practice quality. EHT geometry was modified to generate patches suitable for transplantation in a small-animal model and perspectively humans. Repair efficacy was evaluated at 3 doses in a cryo-injury guinea pig model. Human-scale patches were epicardially transplanted onto healthy hearts in pigs to assess technical feasibility.RESULTS: We created mesh-structured tissue patches for transplantation in guinea pigs (1.5×2.5 cm, 9-15×106 cardiomyocytes) and pigs (5×7 cm, 450×106 cardiomyocytes). EHT patches coherently beat in culture and developed high force (mean 4.6 mN). Cardiomyocytes matured, aligned along the force lines, and demonstrated advanced sarcomeric structure and action potential characteristics closely resembling human ventricular tissue. EHT patches containing ≈4.5, 8.5, 12×106, or no cells were transplanted 7 days after cryo-injury (n=18-19 per group). EHT transplantation resulted in a dose-dependent remuscularization (graft size: 0%-12% of the scar). Only high-dose patches improved left ventricular function (+8% absolute, +24% relative increase). The grafts showed time-dependent cardiomyocyte proliferation. Although standard EHT patches did not withstand transplantation in pigs, the human-scale patch enabled successful patch transplantation.CONCLUSIONS: EHT patch transplantation resulted in a partial remuscularization of the injured heart and improved left ventricular function in a dose-dependent manner in a guinea pig injury model. Human-scale patches were successfully transplanted in pigs in a proof-of-principle study.

AB - BACKGROUND: Human engineered heart tissue (EHT) transplantation represents a potential regenerative strategy for patients with heart failure and has been successful in preclinical models. Clinical application requires upscaling, adaptation to good manufacturing practices, and determination of the effective dose.METHODS: Cardiomyocytes were differentiated from 3 different human induced pluripotent stem cell lines including one reprogrammed under good manufacturing practice conditions. Protocols for human induced pluripotent stem cell expansion, cardiomyocyte differentiation, and EHT generation were adapted to substances available in good manufacturing practice quality. EHT geometry was modified to generate patches suitable for transplantation in a small-animal model and perspectively humans. Repair efficacy was evaluated at 3 doses in a cryo-injury guinea pig model. Human-scale patches were epicardially transplanted onto healthy hearts in pigs to assess technical feasibility.RESULTS: We created mesh-structured tissue patches for transplantation in guinea pigs (1.5×2.5 cm, 9-15×106 cardiomyocytes) and pigs (5×7 cm, 450×106 cardiomyocytes). EHT patches coherently beat in culture and developed high force (mean 4.6 mN). Cardiomyocytes matured, aligned along the force lines, and demonstrated advanced sarcomeric structure and action potential characteristics closely resembling human ventricular tissue. EHT patches containing ≈4.5, 8.5, 12×106, or no cells were transplanted 7 days after cryo-injury (n=18-19 per group). EHT transplantation resulted in a dose-dependent remuscularization (graft size: 0%-12% of the scar). Only high-dose patches improved left ventricular function (+8% absolute, +24% relative increase). The grafts showed time-dependent cardiomyocyte proliferation. Although standard EHT patches did not withstand transplantation in pigs, the human-scale patch enabled successful patch transplantation.CONCLUSIONS: EHT patch transplantation resulted in a partial remuscularization of the injured heart and improved left ventricular function in a dose-dependent manner in a guinea pig injury model. Human-scale patches were successfully transplanted in pigs in a proof-of-principle study.

U2 - 10.1161/CIRCULATIONAHA.120.047904

DO - 10.1161/CIRCULATIONAHA.120.047904

M3 - SCORING: Journal article

C2 - 33648345

VL - 143

SP - 1991

EP - 2006

JO - CIRCULATION

JF - CIRCULATION

SN - 0009-7322

IS - 20

ER -