Cargando…

Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention

PURPOSE OF REVIEW: Cardiovascular toxicity is a leading cause of mortality among cancer survivors and has become increasingly prevalent due to improved cancer survival rates. In this review, we synthesize evidence illustrating how common cancer therapeutic agents, such as anthracyclines, human epide...

Descripción completa

Detalles Bibliográficos
Autores principales: Perry, Tatiana R., Roberts, Michelle L., Sunkara, Bipin, Maddula, Ragasnehith, McLeish, Tyson, Gomez, Jose, Lucas, Julliette, Rayan, David, Patel, Sahishnu, Liang, Mingyu, Bosnjak, Zeljko J., Brown, Sherry-Ann
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer US 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8088904/
https://www.ncbi.nlm.nih.gov/pubmed/33937943
http://dx.doi.org/10.1007/s11912-021-01066-2
_version_ 1783686935155310592
author Perry, Tatiana R.
Roberts, Michelle L.
Sunkara, Bipin
Maddula, Ragasnehith
McLeish, Tyson
Gomez, Jose
Lucas, Julliette
Rayan, David
Patel, Sahishnu
Liang, Mingyu
Bosnjak, Zeljko J.
Brown, Sherry-Ann
author_facet Perry, Tatiana R.
Roberts, Michelle L.
Sunkara, Bipin
Maddula, Ragasnehith
McLeish, Tyson
Gomez, Jose
Lucas, Julliette
Rayan, David
Patel, Sahishnu
Liang, Mingyu
Bosnjak, Zeljko J.
Brown, Sherry-Ann
author_sort Perry, Tatiana R.
collection PubMed
description PURPOSE OF REVIEW: Cardiovascular toxicity is a leading cause of mortality among cancer survivors and has become increasingly prevalent due to improved cancer survival rates. In this review, we synthesize evidence illustrating how common cancer therapeutic agents, such as anthracyclines, human epidermal growth factors receptors (HER2) monoclonal antibodies, and tyrosine kinase inhibitors (TKIs), have been evaluated in cardiomyocytes (CMs) derived from human-induced pluripotent stem cells (hiPSCs) to understand the underlying mechanisms of cardiovascular toxicity. We place this in the context of precision cardio-oncology, an emerging concept for personalizing the prevention and management of cardiovascular toxicities from cancer therapies, accounting for each individual patient’s unique factors. We outline steps that will need to be addressed by multidisciplinary teams of cardiologists and oncologists in partnership with regulators to implement future applications of hiPSCs in precision cardio-oncology. RECENT FINDINGS: Current prevention of cardiovascular toxicity involves routine screenings and management of modifiable risk factors for cancer patients, as well as the initiation of cardioprotective medications. Despite recent advancements in precision cardio-oncology, knowledge gaps remain and limit our ability to appropriately predict with precision which patients will develop cardiovascular toxicity. Investigations using patient-specific CMs facilitate pharmacological discovery, mechanistic toxicity studies, and the identification of cardioprotective pathways. Studies with hiPSCs demonstrate that patients with comorbidities have more frequent adverse responses, compared to their counterparts without cardiac disease. Further studies utilizing hiPSC modeling should be considered, to evaluate the impact and mitigation of known cardiovascular risk factors, including blood pressure, body mass index (BMI), smoking status, diabetes, and physical activity in their role in cardiovascular toxicity after cancer therapy. Future real-world applications will depend on understanding the current use of hiPSC modeling in order for oncologists and cardiologists together to inform their potential to improve our clinical collaborative practice in cardio-oncology. SUMMARY: When applying such in vitro characterization, it is hypothesized that a safety score can be assigned to each individual to determine who has a greater probability of developing cardiovascular toxicity. Using hiPSCs to create personalized models and ultimately evaluate the cardiovascular toxicity of individuals’ treatments may one day lead to more patient-specific treatment plans in precision cardio-oncology while reducing cardiovascular disease (CVD) morbidity and mortality.
format Online
Article
Text
id pubmed-8088904
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Springer US
record_format MEDLINE/PubMed
spelling pubmed-80889042021-05-03 Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention Perry, Tatiana R. Roberts, Michelle L. Sunkara, Bipin Maddula, Ragasnehith McLeish, Tyson Gomez, Jose Lucas, Julliette Rayan, David Patel, Sahishnu Liang, Mingyu Bosnjak, Zeljko J. Brown, Sherry-Ann Curr Oncol Rep Cardio-oncology (EH Yang, Section Editor) PURPOSE OF REVIEW: Cardiovascular toxicity is a leading cause of mortality among cancer survivors and has become increasingly prevalent due to improved cancer survival rates. In this review, we synthesize evidence illustrating how common cancer therapeutic agents, such as anthracyclines, human epidermal growth factors receptors (HER2) monoclonal antibodies, and tyrosine kinase inhibitors (TKIs), have been evaluated in cardiomyocytes (CMs) derived from human-induced pluripotent stem cells (hiPSCs) to understand the underlying mechanisms of cardiovascular toxicity. We place this in the context of precision cardio-oncology, an emerging concept for personalizing the prevention and management of cardiovascular toxicities from cancer therapies, accounting for each individual patient’s unique factors. We outline steps that will need to be addressed by multidisciplinary teams of cardiologists and oncologists in partnership with regulators to implement future applications of hiPSCs in precision cardio-oncology. RECENT FINDINGS: Current prevention of cardiovascular toxicity involves routine screenings and management of modifiable risk factors for cancer patients, as well as the initiation of cardioprotective medications. Despite recent advancements in precision cardio-oncology, knowledge gaps remain and limit our ability to appropriately predict with precision which patients will develop cardiovascular toxicity. Investigations using patient-specific CMs facilitate pharmacological discovery, mechanistic toxicity studies, and the identification of cardioprotective pathways. Studies with hiPSCs demonstrate that patients with comorbidities have more frequent adverse responses, compared to their counterparts without cardiac disease. Further studies utilizing hiPSC modeling should be considered, to evaluate the impact and mitigation of known cardiovascular risk factors, including blood pressure, body mass index (BMI), smoking status, diabetes, and physical activity in their role in cardiovascular toxicity after cancer therapy. Future real-world applications will depend on understanding the current use of hiPSC modeling in order for oncologists and cardiologists together to inform their potential to improve our clinical collaborative practice in cardio-oncology. SUMMARY: When applying such in vitro characterization, it is hypothesized that a safety score can be assigned to each individual to determine who has a greater probability of developing cardiovascular toxicity. Using hiPSCs to create personalized models and ultimately evaluate the cardiovascular toxicity of individuals’ treatments may one day lead to more patient-specific treatment plans in precision cardio-oncology while reducing cardiovascular disease (CVD) morbidity and mortality. Springer US 2021-05-03 2021 /pmc/articles/PMC8088904/ /pubmed/33937943 http://dx.doi.org/10.1007/s11912-021-01066-2 Text en © The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2021 This article is made available via the PMC Open Access Subset for unrestricted research re-use and secondary analysis in any form or by any means with acknowledgement of the original source. These permissions are granted for the duration of the World Health Organization (WHO) declaration of COVID-19 as a global pandemic.
spellingShingle Cardio-oncology (EH Yang, Section Editor)
Perry, Tatiana R.
Roberts, Michelle L.
Sunkara, Bipin
Maddula, Ragasnehith
McLeish, Tyson
Gomez, Jose
Lucas, Julliette
Rayan, David
Patel, Sahishnu
Liang, Mingyu
Bosnjak, Zeljko J.
Brown, Sherry-Ann
Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention
title Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention
title_full Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention
title_fullStr Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention
title_full_unstemmed Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention
title_short Modeling Precision Cardio-Oncology: Using Human-Induced Pluripotent Stem Cells for Risk Stratification and Prevention
title_sort modeling precision cardio-oncology: using human-induced pluripotent stem cells for risk stratification and prevention
topic Cardio-oncology (EH Yang, Section Editor)
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8088904/
https://www.ncbi.nlm.nih.gov/pubmed/33937943
http://dx.doi.org/10.1007/s11912-021-01066-2
work_keys_str_mv AT perrytatianar modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT robertsmichellel modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT sunkarabipin modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT maddularagasnehith modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT mcleishtyson modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT gomezjose modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT lucasjulliette modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT rayandavid modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT patelsahishnu modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT liangmingyu modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT bosnjakzeljkoj modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention
AT brownsherryann modelingprecisioncardiooncologyusinghumaninducedpluripotentstemcellsforriskstratificationandprevention