Editorial: Recent advances in cardiotoxicity testing, volume II

Over the years, there has been a notable rise in the number of cardiovascular complications associated with various therapeutic interventions, particularly in the domain of cancer treatment (Abdul-Rahman et al., 2023; Pawar et al., 2024). Drug-induced cardiotoxicity poses a significant challenge, often leading to market withdrawals and detrimental health outcomes (Destere et al., 2023). This surge in cardiotoxicity cases underscores the necessity for robust preclinical screening strategies to mitigate risks before advancing to human clinical trials. Traditional screening methods heavily relied on animal models, which, albeit valuable, possess limitations in accurately reflecting human cardiac responses and high financial burden. Consequently, the quest for more reliable and cost-effective alternatives has led to a paradigm shift towards innovative in vitro models (Asnani et al., 2021). In recent years, significant strides have been made in leveraging human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs), organoids, and human heart slices as promising tools for cardiotoxicity assessment (Miller et al., 2020; Ou et al., 2020; Meki et al., 2021; Rothbauer et al., 2021; Miller et al., 2022; Zhu et al., 2024). These models offer a more physiologically relevant platform, exhibiting high sensitivity and specificity towards diverse therapeutic interventions while faithfully replicating human cardiac physiology and pathophysiology (Wang et al., 2008).

In this Research Topic, “Recent Advances in Cardiotoxicity Testing, Volume II,” we present a compilation of eight manuscripts comprising four review articles and four original research articles. Each manuscript delves into the latest breakthroughs in cardiotoxicity testing, encompassing a spectrum of innovative methodologies and insights. This collection serves as a comprehensive resource, shedding light on the cutting-edge strategies and advancements shaping the landscape of cardiotoxicity assessment.

(Altrocchi et al., 2023) described the development and validation of a novel assay utilizing human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to assess both acute and delayed drug-induced cardiotoxic effects of reference compounds with known cardiac outcomes. The assay, conducted on 48-well multielectrode array (MEA) plates, evaluated functional-electrophysiological parameters and viability over 4 days. Testing of various compounds, including tyrosine-kinase inhibitors, classic cardiac toxic drugs, ion-channel trafficking inhibitors, and compounds without known cardiotoxicity, revealed the assay’s ability to recapitulate different cardiotoxicities, such as prolongation of field potential, changes in beating rate, arrhythmic events, and impedance reduction. Additionally, fluorescence- and luminescence-based plate reader assays confirmed cytotoxic effects linked to changes observed in the MEA assay. The study underscores the importance of chronic drug evaluation in identifying cardiotoxic effects, emphasizing the potential of this hiPSC-CMs assay to contribute to early compound de-risking and optimize drug development processes.

Similarly (Arefin et al., 2023), investigated the stability and reproducibility of engineered heart tissues (EHTs) derived from human pluripotent stem cell (hPSC)-differentiated cardiomyocytes for cardiac contractility assays in drug development. Utilizing EHTs from different tissue casting batches and hPSC-cardiomyocyte lines, the researchers assessed contractile outputs using a video-optical assay and compared them with monolayer cultures. Drug-induced variations were tested with compounds known for their cardiac effects or cardiotoxicity, alongside analyses of intracellular calcium transients. Establishing quality control criteria based on excitation-contraction coupling, the study identified suitable EHTs for analysis. Results indicate comparable drug-induced contractile responses between EHTs and monolayers, with a close correlation between contractile output and calcium kinetics. The study emphasizes the importance of reproducibility in assessing drug-induced effects in EHTs, suggesting potential future applications with additional mechanistic criteria for specific contexts of use.

In another context (Advani et al., 2023), aimed at replicating the most common genetic associations of chemotherapy-related cardiotoxicity in the adjuvant NSABP B-31 clinical trial. Using genotyping data from 993 patients, TRPC6 rs77679196 and CBR3 rs1056892 (V244M) were found to be linked to doxorubicin-induced cardiac events in both NCCTG N9831 and NSABP B-31. However, other variants previously reported to be linked to trastuzumab-related decline in LVEF failed to replicate between these studies. In an effort to develop cardioprotective peptides (Zerihun and Qvit, 2023), used computational methods to develop peptides that inhibit mitochondrial fission 1 (Fis1)/mitochondrial dynamics 51 kDa (Mid51) protein-protein interactions to reduce the cellular damage that can lead to cardiotoxicity. They identified two peptides, CVP-241 and CVP-242, and characterized their binding dynamic both in silico and in vitro.

In this Research Topic (Xiao et al., 2023), provided a bibliometric and knowledge-Map analysis from 2010 to 2022 to spot the light on the hotspots and emerging trends of cardiotoxicity (Lee et al., 2024). provided a perspective regarding the use of iPSC-cardiomyocytes in the preclinical prediction of candidate pharmaceutical toxicity. They explored compared and contrasted the electrophysiology, calcium handling, cellular signaling, contractile machinery, and metabolism between iPSC-CMs and adult CMs. As well they highlighted the methodologies when using iPSC-CMs to ensure a more representative phenotype of the adult human CM. On the other hand (Wu et al., 2023), reviewed the pathogenic mechanism of Immune checkpoint inhibitors-induced cardiotoxicity and evaluated the commonly used cardio-protective drugs. Finally (Zhang et al., 2023), reviewed the gut microbiota product, trimethylamine oxide (TMAO), and its link to cardio pathology, renal pathology, and cardio-renal syndrome.

As this field of research is evolving rapidly, the present volume of the Research Topic provides an update on the mechanisms of chemotherapy-induced cardiotoxicities, genetic variants related to them, possible modifiers and targets including peptides and metabolites, and testing platforms.

Author contributions

A-BS: Writing–original draft, Writing–review and editing. TM: Writing–original draft, Writing–review and editing.

Funding

The author(s) declare that financial support was received for the research, authorship, and/or publication of this article. This work was supported by the National Institutes of Health R01HL147921, P30GM127607, R15HL168688, R01HL166280 to TM.

Conflict of interest

TM holds equities in Tenaya Therapeutics.

The remaining author declares that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

The author(s) declared that they were an editorial board member of Frontiers, at the time of submission.

References

Abdul-Rahman, T., Dunham, A., Huang, H., Bukhari, S. M. A., Mehta, A., Awuah, W. A., et al. (2023). Chemotherapy induced cardiotoxicity: a state of the art review on general mechanisms, prevention, treatment and recent advances in novel therapeutics. Curr. Problems Cardiol. 48 (4), 101591. doi:10.1016/j.cpcardiol.2023.101591

CrossRef Full Text | Google Scholar

Advani, P. P., Ruddy, K. J., Herrmann, J., Ray, J. C., Craver, E. C., Yothers, G., et al. (2023). Replication of genetic associations of chemotherapy-related cardiotoxicity in the adjuvant NSABP B-31 clinical trial. Front. Oncol. 13, 1139347. doi:10.3389/fonc.2023.1139347

PubMed Abstract | CrossRef Full Text | Google Scholar

Altrocchi, C., Van Ammel, K., Steemans, M., Kreir, M., Tekle, F., Teisman, A., et al. (2023). Evaluation of chronic drug-induced electrophysiological and cytotoxic effects using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Front. Pharmacol. 14, 1229960. doi:10.3389/fphar.2023.1229960

PubMed Abstract | CrossRef Full Text | Google Scholar

Arefin, A., Mendoza, M., Dame, K., Garcia, M. I., Strauss, D. G., and Ribeiro, A. J. S. (2023). Reproducibility of drug-induced effects on the contractility of an engineered heart tissue derived from human pluripotent stem cells. Front. Pharmacol. 14, 1212092. doi:10.3389/fphar.2023.1212092

PubMed Abstract | CrossRef Full Text | Google Scholar

Asnani, A., Moslehi, J. J., Adhikari, B. B., Baik, A. H., Beyer, A. M., de Boer, R. A., et al. (2021). Preclinical models of cancer therapy–associated cardiovascular toxicity: a scientific statement from the American heart association. Circulation Res. 129 (1), e21–e34. doi:10.1161/RES.0000000000000473

PubMed Abstract | CrossRef Full Text | Google Scholar

Destere, A., Merino, D., Lavrut, T., Rocher, F., Viard, D., Drici, M.-D., et al. (2023). Drug-induced cardiac toxicity and adverse drug reactions, a narrative review. Therapies.

Google Scholar

Lee, T. Y. T., Coles, J. G., and Maynes, J. T. (2024). iPSC-cardiomyocytes in the preclinical prediction of candidate pharmaceutical toxicity. Front. Pharmacol. 15, 1308217. doi:10.3389/fphar.2024.1308217

PubMed Abstract | CrossRef Full Text | Google Scholar

Miller, J. M., Meki, M. H., Elnakib, A., Ou, Q., Abouleisa, R. R. E., Tang, X.-L., et al. (2022). Biomimetic cardiac tissue culture model (CTCM) to emulate cardiac physiology and pathophysiology ex vivo. Commun. Biol. 5 (1), 934. doi:10.1038/s42003-022-03919-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Miller, J. M., Meki, M. H., Ou, Q., George, S. A., Gams, A., Abouleisa, R. R. E., et al. (2020). Heart slice culture system reliably demonstrates clinical drug-related cardiotoxicity. Toxicol. Appl. Pharmacol. 406, 115213. doi:10.1016/j.taap.2020.115213

PubMed Abstract | CrossRef Full Text | Google Scholar

Ou, Q., Abouleisa, R. R. E., Tang, X.-L., Juhardeen, H. R., Meki, M. H., Miller, J. M., et al. (2020). Slicing and culturing pig hearts under physiological conditions. JoVE J. Vis. Exp. (157), e60913. doi:10.3791/60913

PubMed Abstract | CrossRef Full Text | Google Scholar

Pawar, V., Wankhede, Y., Kaur, S., Pawar, B., Vasdev, N., Gupta, T., et al. (2024). Drug-induced cardiotoxicity. Public Health Toxicol. Issues Drug Res. 2, 253–282. doi:10.1016/b978-0-443-15842-1.00024-7

CrossRef Full Text | Google Scholar

Rothbauer, M., Bachmann, B. E. M., Eilenberger, C., Kratz, S. R. A., Spitz, S., Höll, G., et al. (2021). A decade of organs-on-a-chip emulating human physiology at the microscale: a critical status report on progress in toxicology and pharmacology. Micromachines 12 (5), 470. doi:10.3390/mi12050470

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, D., Patel, C., Cui, C., and Yan, G.-X. (2008). Preclinical assessment of drug-induced proarrhythmias: role of the arterially perfused rabbit left ventricular wedge preparation. Pharmacol. Ther. 119 (2), 141–151. doi:10.1016/j.pharmthera.2008.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, Y., Xu, Y., Xu, L., Peng, H., Gao, A., Wang, J., et al. (2023). Effects of Shenmai injection against chronic heart failure: a meta-analysis and systematic review of preclinical and clinical studies. Front. Pharmacol. 14, 1338975. doi:10.3389/fphar.2023.1338975

PubMed Abstract | CrossRef Full Text | Google Scholar

Xiao, D., Li, J., Liu, Y., Wang, T., Niu, C., Zhuang, R., et al. (2023). Emerging trends and hotspots evolution in cardiotoxicity: a bibliometric and knowledge-Map analysis from 2010 to 2022. Front. Cardiovasc. Med. 10, 1089916. doi:10.3389/fcvm.2023.1089916

PubMed Abstract | CrossRef Full Text | Google Scholar

Zerihun, M., and Qvit, N. (2023). Selective inhibitors targeting Fis1/Mid51 protein-protein interactions protect against hypoxia-induced damage in cardiomyocytes. Front. Pharmacol. 14, 1275370. doi:10.3389/fphar.2023.1275370

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, J., Zhu, P., Li, S., Gao, Y., and Xing, Y. (2023). From heart failure and kidney dysfunction to cardiorenal syndrome: TMAO may be a bridge. Front. Pharmacol. 14, 1291922. doi:10.3389/fphar.2023.1291922

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, Y., Yang, S., Zhang, T., Ge, Y., Wan, X., and Liang, G. (2024). Cardiac organoids: a 3D technology for disease modeling and drug screening. Curr. Med. Chem. 31. doi:10.2174/0929867331666230727104911

CrossRef Full Text | Google Scholar

留言 (0)

沒有登入
gif