1
|
Wang Z, Zhang J. Genetic and epigenetic bases of long-term adverse effects of childhood cancer therapy. Nat Rev Cancer 2025; 25:129-144. [PMID: 39511414 DOI: 10.1038/s41568-024-00768-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2024] [Indexed: 11/15/2024]
Abstract
Over the past decade, genome-scale molecular profiling of large childhood cancer survivorship cohorts has led to unprecedented advances in our understanding of the genetic and epigenetic bases of therapy-related adverse health outcomes in this vulnerable population. To facilitate the integration of knowledge generated from these studies into formulating next-generation precision care for survivors of childhood cancer, we summarize key findings of genetic and epigenetic association studies of long-term therapy-related adverse effects including subsequent neoplasms and cardiomyopathies among others. We also discuss therapy-related genotoxicities including clonal haematopoiesis and DNA methylation, which may underlie accelerated molecular ageing. Finally, we highlight enhanced risk prediction models for survivors of childhood cancer that incorporate both genetic factors and treatment exposures, aiming to achieve enhanced accuracy in predicting risks for this population. These new insights will hopefully inspire future studies that harness both expanding omics resources and evolving data science methodology to accelerate the translation of precision medicine for survivors of childhood cancer.
Collapse
Affiliation(s)
- Zhaoming Wang
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA.
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
2
|
Tan N, Luo H, Li W, Ling G, Wei Y, Wang W, Wang Y. The dual function of autophagy in doxorubicin-induced cardiotoxicity: Mechanism and natural products. Semin Cancer Biol 2025; 109:83-90. [PMID: 39827930 DOI: 10.1016/j.semcancer.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Doxorubicin (DOX) is an anthracycline antitumor drug discovered in 1969, which can care for leukemia, breast cancer, lymphoma, and sarcoma. However, cardiotoxicity induced by DOX seriously limits its clinical value. The etiopathogenesis and therapeutic strategies are not unified. Autophagy is a critical mechanism in the progression of DOX-induced cardiotoxicity (DIC), autophagy intervention is a potential therapeutic strategy for DIC. Natural product has been considered as a complementary and alternative approach to treat cardiovascular disease. In this review, we summarize the pathology of autophagy in DIC and the natural products for DIC therapy.
Collapse
Affiliation(s)
- Nannan Tan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; Anhui university of Chinese medicine, Hefei 230012, China
| | - Hanwen Luo
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Weili Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Guanjing Ling
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan Wei
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100029, China.
| | - Yong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
3
|
Jacquemyn X, Chinni BK, Barnes BT, Rao S, Kutty S, Manlhiot C. Unsupervised machine learning identifies distinct phenotypes in cardiac complications of pediatric patients treated with anthracyclines. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:74. [PMID: 39468669 PMCID: PMC11514752 DOI: 10.1186/s40959-024-00276-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 10/17/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Anthracyclines are essential in pediatric cancer treatment, but patients are at risk cancer therapy-related cardiac dysfunction (CTRCD). Standardized definitions by the International Cardio-Oncology Society (IC-OS) aim to enhance precision in risk assessment. OBJECTIVES Categorize distinct phenotypes among pediatric patients undergoing anthracycline chemotherapy using unsupervised machine learning. METHODS Pediatric cancer patients undergoing anthracycline chemotherapy at our institution were retrospectively included. Clinical and echocardiographic data at baseline, along with follow-up data, were collected from patient records. Unsupervised machine learning was performed, involving dimensionality reduction using principal component analysis and K-means clustering to identify different phenotypic clusters. Identified phenogroups were analyzed for associations with CTRCD, defined following contemporary IC-OS definitions, and hypertensive response. RESULTS A total of 187 patients (63.1% male, median age 15.5 years [10.4-18.7]) were included and received anthracycline chemotherapy with a median treatment duration of 0.66 years [0.35-1.92]. Median follow-up duration was 2.78 years [1.31-4.21]. Four phenogroups were identified with following distribution: Cluster 0 (32.6%, n = 61), Cluster 1 (13.9%, n = 26), Cluster 2 (24.6%, n = 46), and Cluster 3 (28.9%, n = 54). Cluster 0 showed the highest risk of moderate CTRCD (HR: 3.10 [95% CI: 1.18-8.16], P = 0.022) compared to other clusters. Cluster 3 demonstrated a protective effect against hypertensive response (HR: 0.30 [95% CI: 0.13- 0.67], P = 0.003) after excluding baseline hypertensive patients. Longitudinal assessments revealed differences in global longitudinal strain and systolic blood pressure among phenogroups. CONCLUSIONS Unsupervised machine learning identified distinct phenogroups among pediatric cancer patients undergoing anthracycline chemotherapy, offering potential for personalized risk assessment.
Collapse
Affiliation(s)
- Xander Jacquemyn
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
- Department of Cardiovascular Sciences, KU Leuven & Congenital and Structural Cardiology, UZ Leuven, Leuven, Belgium
| | - Bhargava K Chinni
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
| | - Benjamin T Barnes
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
| | - Sruti Rao
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
| | - Shelby Kutty
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA
| | - Cedric Manlhiot
- Department of Pediatrics, The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Johns Hopkins School of Medicine, Johns Hopkins University, Johns Hopkins Hospital, 600 N. Wolfe Street, 1389 Blalock, Baltimore, MD, 21287, USA.
| |
Collapse
|
4
|
Edwards LA, Yang C, Sharma S, Chen ZH, Gorantla L, Joshi SA, Longhi NJ, Worku N, Yang JS, Martinez Di Pietro B, Armenian S, Bhat A, Border W, Buddhe S, Blythe N, Stratton K, Leger KJ, Leisenring WM, Meacham LR, Nathan PC, Narasimhan S, Sachdeva R, Sadak K, Chow EJ, Boyle PM. Building a machine learning-assisted echocardiography prediction tool for children at risk for cancer therapy-related cardiomyopathy. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:66. [PMID: 39385257 PMCID: PMC11462765 DOI: 10.1186/s40959-024-00268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Despite routine echocardiographic surveillance for childhood cancer survivors, the ability to predict cardiomyopathy risk in individual patients is limited. We explored the feasibility and optimal processes for machine learning-enhanced cardiomyopathy prediction in survivors using serial echocardiograms from five centers. METHODS We designed a series of deep convolutional neural networks (DCNNs) for prediction of cardiomyopathy (shortening fraction ≤ 28% or ejection fraction ≤ 50% on two occasions) for at-risk survivors ≥ 1-year post initial cancer therapy. We built DCNNs with four subsets of echocardiographic data differing in timing relative to case (survivor who developed cardiomyopathy) index diagnosis and two input formats (montages) with differing image selections. We used holdout subsets in a 10-fold cross-validation framework and standard metrics to assess model performance (e.g., F1-score, area under the precision-recall curve [AUPRC]). Performance of the input formats was compared using a combined 5 × 2 cross-validation F-test. RESULTS The dataset included 542 pairs of montages: 171 montage pairs from 45 cases at time of cardiomyopathy diagnosis or pre-diagnosis and 371 pairs from 70 at-risk survivors who didn't develop cardiomyopathy during follow-up (non-case). The DCNN trained to distinguish between non-case and time of cardiomyopathy diagnosis or pre-diagnosis case montages achieved an AUROC of 0.89 ± 0.02, AUPRC 0.83 ± 0.03, and F1-score: 0.76 ± 0.04. When limited to smaller subsets of case data (e.g., ≥ 1 or 2 years pre-diagnosis), performance worsened. Model input format did not impact performance accuracy across models. CONCLUSIONS This methodology is a promising first step toward development of a DCNN capable of accurately differentiating pre-diagnosis versus non-case echocardiograms to predict survivors more likely to develop cardiomyopathy.
Collapse
Affiliation(s)
- Lindsay A Edwards
- Department of Pediatrics, Division of Cardiology, Duke University Medical Center, DUMC Box 3090, Durham, NC, 27710, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
| | | | - Surbhi Sharma
- Department of Bioengineering, University of Washington, 3720 15th Ave NE N361, UW Mailbox 355061, Seattle, WA, 98195, USA
| | - Zih-Hua Chen
- Department of Bioengineering, University of Washington, 3720 15th Ave NE N361, UW Mailbox 355061, Seattle, WA, 98195, USA
| | - Lahari Gorantla
- Department of Bioengineering, University of Washington, 3720 15th Ave NE N361, UW Mailbox 355061, Seattle, WA, 98195, USA
| | - Sanika A Joshi
- Department of Bioengineering, University of Washington, 3720 15th Ave NE N361, UW Mailbox 355061, Seattle, WA, 98195, USA
| | - Nicolas J Longhi
- Department of Bioengineering, University of Washington, 3720 15th Ave NE N361, UW Mailbox 355061, Seattle, WA, 98195, USA
| | - Nahom Worku
- Department of Bioengineering, University of Washington, 3720 15th Ave NE N361, UW Mailbox 355061, Seattle, WA, 98195, USA
| | - Jamie S Yang
- Department of Bioengineering, University of Washington, 3720 15th Ave NE N361, UW Mailbox 355061, Seattle, WA, 98195, USA
| | | | - Saro Armenian
- Departments of Pediatrics and Population Sciences, City of Hope, Duarte, CA, USA
| | - Aarti Bhat
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Seattle Children's Hospital, Seattle, WA, USA
| | - William Border
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sujatha Buddhe
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Division of Pediatric Cardiology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Nancy Blythe
- Clinical Research and Public Health Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kayla Stratton
- Clinical Research and Public Health Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Kasey J Leger
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Seattle Children's Hospital, Seattle, WA, USA
| | - Wendy M Leisenring
- Clinical Research and Public Health Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lillian R Meacham
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Paul C Nathan
- Department of Pediatrics, University of Toronto, The Hospital for Sick Children, Toronto, ON, Canada
| | - Shanti Narasimhan
- Department of Pediatrics, University of Minnesota, Masonic Children's Hospital, Minneapolis, MN, USA
| | - Ritu Sachdeva
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Karim Sadak
- Department of Pediatrics, University of Minnesota, Masonic Children's Hospital, Minneapolis, MN, USA
| | - Eric J Chow
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Seattle Children's Hospital, Seattle, WA, USA
- Clinical Research and Public Health Sciences Divisions, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Patrick M Boyle
- Department of Bioengineering, University of Washington, 3720 15th Ave NE N361, UW Mailbox 355061, Seattle, WA, 98195, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
5
|
Wyatt KD, Alexander N, Hills GD, Liang WH, Kadauke S, Volchenboum SL, Mian A, Phillips CA. Making sense of artificial intelligence and large language models-including ChatGPT-in pediatric hematology/oncology. Pediatr Blood Cancer 2024; 71:e31143. [PMID: 38924670 DOI: 10.1002/pbc.31143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024]
Abstract
ChatGPT and other artificial intelligence (AI) systems have captivated the attention of healthcare providers and researchers for their potential to improve care processes and outcomes. While these technologies hold promise to automate processes, increase efficiency, and reduce cognitive burden, their use also carries risks. In this commentary, we review basic concepts of AI, outline some of the capabilities and limitations of currently available tools, discuss current and future applications in pediatric hematology/oncology, and provide an evaluation and implementation framework that can be used by pediatric hematologist/oncologists considering the use of AI in clinical practice.
Collapse
Affiliation(s)
- Kirk D Wyatt
- Department of Pediatric Hematology/Oncology, Roger Maris Cancer Center, Fargo, North Dakota, USA
- Data for the Common Good, University of Chicago, Chicago, Illinois, USA
| | - Natasha Alexander
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Gerard D Hills
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Regenstrief Institute, Indianapolis, Indiana, USA
- Riley Children's Health at Indiana University Health, Indianapolis, Indiana, USA
| | - Wayne H Liang
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - Stephan Kadauke
- Department of Pathology and Lab Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Cell and Gene Therapy Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Samuel L Volchenboum
- Data for the Common Good, University of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, University of Chicago, Chicago, Illinois, USA
| | - Amir Mian
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Dell Children's Hospital, Austin, Texas, USA
- Dell Medical School, University of Texas at Austin, Austin, Texas, USA
| | - Charles A Phillips
- Department of Pediatrics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Liu X, Ge S, Zhang A. Pediatric Cardio-Oncology: Screening, Risk Stratification, and Prevention of Cardiotoxicity Associated with Anthracyclines. CHILDREN (BASEL, SWITZERLAND) 2024; 11:884. [PMID: 39062333 PMCID: PMC11276082 DOI: 10.3390/children11070884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Anthracyclines have significantly improved the survival of children with malignant tumors, but the associated cardiotoxicity, an effect now under the purview of pediatric cardio-oncology, due to its cumulative and irreversible effects on the heart, limits their clinical application. A systematic screening and risk stratification approach provides the opportunity for early identification and intervention to mitigate, reverse, or prevent myocardial injury, remodeling, and dysfunction associated with anthracyclines. This review summarizes the risk factors, surveillance indexes, and preventive strategies of anthracycline-related cardiotoxicity to improve the safety and efficacy of anthracyclines.
Collapse
Affiliation(s)
- Xiaomeng Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Shuping Ge
- Department of Pediatric and Adult Congenital Cardiology, Geisinger Clinic, Danville, PA 17822, USA
| | - Aijun Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
7
|
Boen HM, Alaerts M, Goovaerts I, Saenen JB, Franssen C, Vorlat A, Vermeulen T, Heidbuchel H, Van Laer L, Loeys B, Van Craenenbroeck EM. Variants in structural cardiac genes in patients with cancer therapy-related cardiac dysfunction after anthracycline chemotherapy: a case control study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:26. [PMID: 38689299 PMCID: PMC11059765 DOI: 10.1186/s40959-024-00231-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Variants in cardiomyopathy genes have been identified in patients with cancer therapy-related cardiac dysfunction (CTRCD), suggesting a genetic predisposition for the development of CTRCD. The diagnostic yield of genetic testing in a CTRCD population compared to a cardiomyopathy patient cohort is not yet known and information on which genes should be assessed in this population is lacking. METHODS We retrospectively included 46 cancer patients with a history of anthracycline induced CTRCD (defined as a decrease in left ventricular ejection fraction (LVEF) to < 50% and a ≥ 10% reduction from baseline by echocardiography). Genetic testing was performed for 59 established cardiomyopathy genes. Only variants of uncertain significance and (likely) pathogenic variants were included. Diagnostic yield of genetic testing was compared with a matched cohort of patients with dilated cardiomyopathy (DCM, n = 46) and a matched cohort of patients without cardiac disease (n = 111). RESULTS Average LVEF at time of CTRCD diagnosis was 30.1 ± 11.0%. Patients were 52.9 ± 14.6 years old at time of diagnosis and 30 (65.2%) were female. Most patients were treated for breast cancer or lymphoma, with a median doxorubicin equivalent dose of 300 mg/m2 [112.5-540.0]. A genetic variant, either pathogenic, likely pathogenic or of uncertain significance, was identified in 29/46 (63.0%) of patients with CTRCD, which is similar to the DCM cohort (34/46, 73.9%, p = 0.262), but significantly higher than in the negative control cohort (47/111, 39.6%, p = 0.018). Variants in TTN were the most prevalent in the CTRCD cohort (43% of all variants). All (likely) pathogenic variants identified in the CTRCD cohort were truncating variants in TTN. There were no significant differences in severity of CTRCD and in recovery rate in variant-harbouring individuals versus non-variant harbouring individuals. CONCLUSIONS In this case-control study, cancer patients with anthracycline-induced CTRCD have an increased burden of genetic variants in cardiomyopathy genes, similar to a DCM cohort. If validated in larger prospective studies, integration of genetic data in risk prediction models for CTRCD may guide cancer treatment. Moreover, genetic results have important clinical impact, both for the patient in the setting of precision medicine, as for the family members that will receive genetic counselling.
Collapse
Affiliation(s)
- Hanne M Boen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium.
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium.
| | - Maaike Alaerts
- Centrum of Medical Genetics, GENCOR, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - Inge Goovaerts
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
- Centrum of Medical Genetics, GENCOR, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - Johan B Saenen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Anne Vorlat
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Tom Vermeulen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Hein Heidbuchel
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Lut Van Laer
- Centrum of Medical Genetics, GENCOR, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - Bart Loeys
- Centrum of Medical Genetics, GENCOR, Antwerp University Hospital and University of Antwerp, Antwerp, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
8
|
Ni MM, Yang JF, Miao J, Xu J. Association between genetic variants of transmembrane transporters and susceptibility to anthracycline-induced cardiotoxicity: Current understanding and existing evidence. Clin Genet 2024; 105:115-129. [PMID: 37961936 DOI: 10.1111/cge.14452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023]
Abstract
Anthracyclines remain the cornerstone of numerous chemotherapeutic protocols, with beneficial effects against haematological malignancies and solid tumours. Unfortunately, the clinical usefulness of anthracyclines is compromised by the development of cardiotoxic side effects, leading to dose limitations or treatment discontinuation. There is no absolute linear correlation between the incidence of cardiotoxicity and the threshold dose, suggesting that genetic factors may modify the association between anthracyclines and cardiotoxicity risk. And the majority of single nucleotide polymorphisms (SNPs) associated with anthracycline pharmacogenomics were identified in the ATP-binding cassette (ABC) and solute carrier (SLC) transporters, generating increasing interest in the pharmacogenetic implications of their genetic variations for anthracycline-induced cardiotoxicity (AIC). This review focuses on the influence of SLC and ABC polymorphisms on AIC and highlights the prospects and clinical significance of pharmacogenetics for individualised preventive approaches.
Collapse
Affiliation(s)
- Ming-Ming Ni
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ju-Fei Yang
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jing Miao
- Department of Pharmacy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
- Research Center for Clinical Pharmacy, Zhejiang University, Hangzhou, China
| | - Jin Xu
- Department of Pharmacy, Children's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Zheng Y, Chen Z, Huang S, Zhang N, Wang Y, Hong S, Chan JSK, Chen KY, Xia Y, Zhang Y, Lip GY, Qin J, Tse G, Liu T. Machine Learning in Cardio-Oncology: New Insights from an Emerging Discipline. Rev Cardiovasc Med 2023; 24:296. [PMID: 39077576 PMCID: PMC11273149 DOI: 10.31083/j.rcm2410296] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 07/31/2024] Open
Abstract
A growing body of evidence on a wide spectrum of adverse cardiac events following oncologic therapies has led to the emergence of cardio-oncology as an increasingly relevant interdisciplinary specialty. This also calls for better risk-stratification for patients undergoing cancer treatment. Machine learning (ML), a popular branch discipline of artificial intelligence that tackles complex big data problems by identifying interaction patterns among variables, has seen increasing usage in cardio-oncology studies for risk stratification. The objective of this comprehensive review is to outline the application of ML approaches in cardio-oncology, including deep learning, artificial neural networks, random forest and summarize the cardiotoxicity identified by ML. The current literature shows that ML has been applied for the prediction, diagnosis and treatment of cardiotoxicity in cancer patients. In addition, role of ML in gender and racial disparities for cardiac outcomes and potential future directions of cardio-oncology are discussed. It is essential to establish dedicated multidisciplinary teams in the hospital and educate medical professionals to become familiar and proficient in ML in the future.
Collapse
Affiliation(s)
- Yi Zheng
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular
Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second
Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Ziliang Chen
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular
Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second
Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Shan Huang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular
Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second
Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Nan Zhang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular
Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second
Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Yueying Wang
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular
Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second
Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Shenda Hong
- National Institute of Health Data Science at Peking University, Peking
University, 100871 Beijing, China
- Institute of Medical Technology, Peking University Health Science Center,
100871 Beijing, China
| | - Jeffrey Shi Kai Chan
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Limited, 999077 Hong
Kong, China
| | - Kang-Yin Chen
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular
Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second
Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Yunlong Xia
- Department of Cardiology, First Affiliated Hospital of Dalian Medical
University, 116011 Dalian, Liaoning, China
| | - Yuhui Zhang
- Heart Failure Center, State Key Laboratory of Cardiovascular Disease,
Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of
Medical Sciences and Peking Union Medical College, 100037 Beijing, China
| | - Gregory Y.H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool,
Liverpool John Moores University and Liverpool Heart & Chest Hospital, L69 3BX
Liverpool, UK
- Danish Center for Health Services Research, Department of Clinical Medicine,
Aalborg University, 999017 Aalborg, Denmark
| | - Juan Qin
- Section of Cardio-Oncology & Immunology, Division of Cardiology and the
Cardiovascular Research Institute, University of California San Francisco, San
Francisco, CA 94143, USA
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular
Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second
Hospital of Tianjin Medical University, 300211 Tianjin, China
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Limited, 999077 Hong
Kong, China
- School of Nursing and Health Studies, Hong Kong Metropolitan University,
999077 Hong Kong, China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular
Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second
Hospital of Tianjin Medical University, 300211 Tianjin, China
| |
Collapse
|
10
|
Kistamás K, Müller A, Muenthaisong S, Lamberto F, Zana M, Dulac M, Leal F, Maziz A, Costa P, Bernotiene E, Bergaud C, Dinnyés A. Multifactorial approaches to enhance maturation of human iPSC-derived cardiomyocytes. J Mol Liq 2023; 387:122668. [DOI: 10.1016/j.molliq.2023.122668] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
11
|
Roman-Naranjo P, Parra-Perez AM, Lopez-Escamez JA. A systematic review on machine learning approaches in the diagnosis and prognosis of rare genetic diseases. J Biomed Inform 2023:104429. [PMID: 37352901 DOI: 10.1016/j.jbi.2023.104429] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 06/05/2023] [Accepted: 06/17/2023] [Indexed: 06/25/2023]
Abstract
BACKGROUND The diagnosis of rare genetic diseases is often challenging due to the complexity of the genetic underpinnings of these conditions and the limited availability of diagnostic tools. Machine learning (ML) algorithms have the potential to improve the accuracy and speed of diagnosis by analyzing large amounts of genomic data and identifying complex multiallelic patterns that may be associated with specific diseases. In this systematic review, we aimed to identify the methodological trends and the ML application areas in rare genetic diseases. METHODS We performed a systematic review of the literature following the PRISMA guidelines to search studies that used ML approaches to enhance the diagnosis of rare genetic diseases. Studies that used DNA-based sequencing data and a variety of ML algorithms were included, summarized, and analyzed using bibliometric methods, visualization tools, and a feature co-occurrence analysis. FINDINGS Our search identified 22 studies that met the inclusion criteria. We found that exome sequencing was the most frequently used sequencing technology (59%), and rare neoplastic diseases were the most prevalent disease scenario (59%). In rare neoplasms, the most frequent applications of ML models were the differential diagnosis or stratification of patients (38.5%) and the identification of somatic mutations (30.8%). In other rare diseases, the most frequent goals were the prioritization of rare variants or genes (55.5%) and the identification of biallelic or digenic inheritance (33.3%). The most employed method was the random forest algorithm (54.5%). In addition, the features of the datasets needed for training these algorithms were distinctive depending on the goal pursued, including the mutational load in each gene for the differential diagnosis of patients, or the combination of genotype features and sequence-derived features (such as GC-content) for the identification of somatic mutations. CONCLUSIONS ML algorithms based on sequencing data are mainly used for the diagnosis of rare neoplastic diseases, with random forest being the most common approach. We identified key features in the datasets used for training these ML models according to the objective pursued. These features can support the development of future ML models in the diagnosis of rare genetic diseases.
Collapse
Affiliation(s)
- P Roman-Naranjo
- Division of Otolaryngology, Department of Surgery, Instituto de Investigación Biosanitaria, ibs.GRANADA, Universidad de Granada, Granada, Spain; Otology and Neurotology Group CTS495, Department of Genomic Medicine, GENYO - Centre for Genomics and Oncological Research - Pfizer, University of Granada, Junta de Andalucía, PTS, Granada, Spain; Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain.
| | - A M Parra-Perez
- Division of Otolaryngology, Department of Surgery, Instituto de Investigación Biosanitaria, ibs.GRANADA, Universidad de Granada, Granada, Spain; Otology and Neurotology Group CTS495, Department of Genomic Medicine, GENYO - Centre for Genomics and Oncological Research - Pfizer, University of Granada, Junta de Andalucía, PTS, Granada, Spain; Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
| | - J A Lopez-Escamez
- Division of Otolaryngology, Department of Surgery, Instituto de Investigación Biosanitaria, ibs.GRANADA, Universidad de Granada, Granada, Spain; Otology and Neurotology Group CTS495, Department of Genomic Medicine, GENYO - Centre for Genomics and Oncological Research - Pfizer, University of Granada, Junta de Andalucía, PTS, Granada, Spain; Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain; Meniere's Disease Neuroscience Research Program, Faculty of Medicine & Health, School of Medical Sciences, The Kolling Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
12
|
Ng CT, Tan LL, Sohn IS, Gonzalez Bonilla H, Oka T, Yinchoncharoen T, Chang WT, Chong JH, Cruz Tan MK, Cruz RR, Astuti A, Agarwala V, Chien V, Youn JC, Tong J, Herrmann J. Advancing Cardio-Oncology in Asia. Korean Circ J 2023; 53:69-91. [PMID: 36792558 PMCID: PMC9932224 DOI: 10.4070/kcj.2022.0255] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/25/2022] [Indexed: 11/25/2022] Open
Abstract
Cardio-oncology is an emerging multi-disciplinary field, which aims to reduce morbidity and mortality of cancer patients by preventing and managing cancer treatment-related cardiovascular toxicities. With the exponential growth in cancer and cardiovascular diseases in Asia, there is an emerging need for cardio-oncology awareness among physicians and country-specific cardio-oncology initiatives. In this state-of-the-art review, we sought to describe the burden of cancer and cardiovascular disease in Asia, a region with rich cultural and socio-economic diversity. From describing the uniqueness and challenges (such as socio-economic disparity, ethnical and racial diversity, and limited training opportunities) in establishing cardio-oncology in Asia, and outlining ways to overcome any barriers, this article aims to help advance the field of cardio-oncology in Asia.
Collapse
Affiliation(s)
- Choon Ta Ng
- Department of Cardiovascular Medicine, Mayo Clinic Rochester, Rochester, MN, USA
- Department of Cardiology, National Heart Centre Singapore, Singapore.
| | - Li Ling Tan
- Department of Cardiology, National University Heart Centre Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Il Suk Sohn
- Department of Cardiology, Kyung Hee University Hospital at Gangdong, Seoul, Korea
| | | | - Toru Oka
- Onco-Cardiology Unit, Department of Internal Medicine, Saitama Cancer Center, Saitama, Japan
| | | | - Wei-Ting Chang
- Division of Cardiology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Jun Hua Chong
- Department of Cardiology, National Heart Centre Singapore, Singapore
| | | | - Rochelle Regina Cruz
- Department of Cardiology, Cardinal Santos Medical Center, Metro Manila, The Philippines
| | - Astri Astuti
- Department of Cardiology and Vascular Medicine, Hasan Sadikin General Hospital, Bandung, Indonesia
| | - Vivek Agarwala
- Department of Medical Oncology and Haemato-Oncology, Narayana Superspeciality Hospital and Cancer Institute, Howrah, India
| | - Van Chien
- Department of Cardiology, National Heart Institute, Hanoi, Vietnam
| | - Jong-Chan Youn
- Seoul St. Mary's Hospital, Catholic Research Institute for Intractable Cardiovascular Disease, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieli Tong
- Department of Cardiology, Tan Tock Seng Hospital, Singapore
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic Rochester, Rochester, MN, USA.
| |
Collapse
|
13
|
Prediction of chemotherapy-related complications in pediatric oncology patients: artificial intelligence and machine learning implementations. Pediatr Res 2023; 93:390-395. [PMID: 36302858 DOI: 10.1038/s41390-022-02356-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 10/08/2022] [Accepted: 10/12/2022] [Indexed: 11/08/2022]
Abstract
Although the overall incidence of pediatric oncological diseases tends to increase over the years, it is among the rare diseases of the pediatric population. The diagnosis, treatment, and healthcare management of this group of diseases are important. Prevention of treatment-related complications is vital for patients, particularly in the pediatric population. Nowadays, the use of artificial intelligence and machine learning technologies in the management of oncological diseases is becoming increasingly important. With the advancement of software technologies, improvements have been made in the early diagnosis of risk groups in oncological diseases, in radiology, pathology, and imaging technologies, in cancer staging and management. In addition, these technologies can be used to predict the outcome in chemotherapy treatment of oncological diseases. In this context, this study identifies artificial intelligence and machine learning methods used in the prediction of complications due to chemotherapeutic agents used in childhood cancer treatment. For this purpose, the concepts of artificial intelligence and machine learning are explained in this review. A general framework for the use of machine learning in healthcare and pediatric oncology has been drawn and examples of studies conducted on this topic in pediatric oncology have been given. IMPACT: Artificial intelligence and machine learning are advanced tools that can be used to predict chemotherapy-related complications. Algorithms can assist clinicians' decision-making processes in the management of complications. Although studies are using these methods, there is a need to increase the number of studies on artificial intelligence applications in pediatric clinics.
Collapse
|
14
|
Hurkmans EGE, Brand ACAM, Verdonschot JAJ, te Loo DMWM, Coenen MJH. Pharmacogenetics of chemotherapy treatment response and -toxicities in patients with osteosarcoma: a systematic review. BMC Cancer 2022; 22:1326. [PMID: 36536332 PMCID: PMC9761983 DOI: 10.1186/s12885-022-10434-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Osteosarcoma is the most common bone tumor in children and adolescents. Despite multiagent chemotherapy, only 71% of patients survives and these survivors often experience long-term toxicities. The main objective of this systematic review is to provide an overview of the discovery of novel associations of germline polymorphisms with treatment response and/or chemotherapy-induced toxicities in osteosarcoma. METHODS: MEDLINE and Embase were systematically searched (2010-July 2022). Genetic association studies were included if they assessed > 10 germline genetic variants in > 5 genes in relevant drug pathways or if they used a genotyping array or other large-scale genetic analysis. Quality was assessed using adjusted STrengthening the REporting of Genetic Association studies (STREGA)-guidelines. To find additional evidence for the identified associations, literature was searched to identify replication studies. RESULTS After screening 1999 articles, twenty articles met our inclusion criteria. These range from studies focusing on genes in relevant pharmacokinetic pathways to whole genome sequencing. Eleven articles reported on doxorubicin-induced cardiomyopathy. For seven genetic variants in CELF4, GPR35, HAS3, RARG, SLC22A17, SLC22A7 and SLC28A3, replication studies were performed, however without consistent results. Ototoxicity was investigated in one study. Five small studies reported on mucosistis or bone marrow, nephro- and/or hepatotoxicity. Six studies included analysis for treatment efficacy. Genetic variants in ABCC3, ABCC5, FasL, GLDC, GSTP1 were replicated in studies using heterogeneous efficacy outcomes. CONCLUSIONS Despite that results are promising, the majority of associations were poorly reproducible due to small patient cohorts. For the future, hypothesis-generating studies in large patient cohorts will be necessary, especially for cisplatin-induced ototoxicity as these are largely lacking. In order to form large patient cohorts, national and international collaboration will be essential.
Collapse
Affiliation(s)
- Evelien G. E. Hurkmans
- grid.10417.330000 0004 0444 9382Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Annouk C. A. M. Brand
- grid.10417.330000 0004 0444 9382Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Job A. J. Verdonschot
- grid.412966.e0000 0004 0480 1382Department of Clinical Genetics and Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - D. Maroeska W. M. te Loo
- grid.10417.330000 0004 0444 9382Department of Pediatrics, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Marieke J. H. Coenen
- grid.10417.330000 0004 0444 9382Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands ,grid.5645.2000000040459992XDepartment of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
15
|
Wang DD, Li YF, Zhang C, He SM, Chen X. Predicting the effect of sirolimus on disease activity in patients with systemic lupus erythematosus using machine learning. J Clin Pharm Ther 2022; 47:1845-1850. [PMID: 36131617 DOI: 10.1111/jcpt.13778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/03/2022] [Accepted: 09/04/2022] [Indexed: 11/30/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVES The present study aimed to predict the effect of sirolimus on disease activity in patients with systemic lupus erythematosus (SLE) using machine learning and to recommend appropriate sirolimus dosage regimen for patients with SLE. METHODS The Emax model was selected for machine learning, where the evaluation indicator was the change rate of systemic lupus erythematosus disease activity index from baseline value. RESULTS A total 103 patients with SLE were included for modelling, where the Emax , ET50 were -53.9%, 1.53 months in the final model respectively, and the evaluation of the final model was good. Further simulation found that the follow-up time to achieve 25%, 50%, 75% and 80% (plateau) Emax of sirolimus effecting on disease activity in patients with SLE were 0.51, 1.53, 4.59 and 6.12 months, respectively. In addition, the sirolimus dosage was flexible and adjusted according to drug concentration, where the intersection of sirolimus concentration range included in this study was about 8-10 ng/ml. WHAT IS NEW AND CONCLUSIONS This study was the first time to predict the effect of sirolimus on disease activity in patients with SLE and in order to achieve better therapeutic effect maintaining a concentration of 8-10 ng/ml sirolimus for at least 6.12 months was necessary.
Collapse
Affiliation(s)
- Dong-Dong Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ya-Feng Li
- Department of Pharmacy, Feng Xian People's Hospital, Xuzhou, Jiangsu, China
| | - Cun Zhang
- Department of Pharmacy, Xuzhou Oriental Hospital Affiliated to Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Su-Mei He
- Department of Pharmacy, Suzhou Science & Technology Town Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Xiao Chen
- School of Nursing, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
16
|
Siemens A, Anderson SJ, Rassekh SR, Ross CJD, Carleton BC. A Systematic Review of Polygenic Models for Predicting Drug Outcomes. J Pers Med 2022; 12:jpm12091394. [PMID: 36143179 PMCID: PMC9505711 DOI: 10.3390/jpm12091394] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Polygenic models have emerged as promising prediction tools for the prediction of complex traits. Currently, the majority of polygenic models are developed in the context of predicting disease risk, but polygenic models may also prove useful in predicting drug outcomes. This study sought to understand how polygenic models incorporating pharmacogenetic variants are being used in the prediction of drug outcomes. A systematic review was conducted with the aim of gaining insights into the methods used to construct polygenic models, as well as their performance in drug outcome prediction. The search uncovered 89 papers that incorporated pharmacogenetic variants in the development of polygenic models. It was found that the most common polygenic models were constructed for drug dosing predictions in anticoagulant therapies (n = 27). While nearly all studies found a significant association with their polygenic model and the investigated drug outcome (93.3%), less than half (47.2%) compared the performance of the polygenic model against clinical predictors, and even fewer (40.4%) sought to validate model predictions in an independent cohort. Additionally, the heterogeneity of reported performance measures makes the comparison of models across studies challenging. These findings highlight key considerations for future work in developing polygenic models in pharmacogenomic research.
Collapse
Affiliation(s)
- Angela Siemens
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6H 3N1, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Spencer J. Anderson
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6H 3N1, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - S. Rod Rassekh
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6H 3V4, Canada
- Division of Oncology, Hematology and Bone Marrow Transplant, University of British Columbia, Vancouver, BC V6H 3V4, Canada
| | - Colin J. D. Ross
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6H 3N1, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Bruce C. Carleton
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6H 3N1, Canada
- Division of Translational Therapeutics, Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver, BC V6H 3V4, Canada
- Pharmaceutical Outcomes Programme, British Columbia Children’s Hospital, Vancouver, BC V5Z 4H4, Canada
- Correspondence:
| |
Collapse
|
17
|
Gambril JA, Chum A, Goyal A, Ruz P, Mikrut K, Simonetti O, Dholiya H, Patel B, Addison D. Cardiovascular Imaging in Cardio-Oncology: The Role of Echocardiography and Cardiac MRI in Modern Cardio-Oncology. Heart Fail Clin 2022; 18:455-478. [PMID: 35718419 PMCID: PMC9280694 DOI: 10.1016/j.hfc.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cardiovascular (CV) events are an increasingly common limitation of effective anticancer therapy. Over the last decade imaging has become essential to patients receiving contemporary cancer therapy. Herein we discuss the current state of CV imaging in cardio-oncology. We also provide a practical apparatus for the use of imaging in everyday cardiovascular care of oncology patients to improve outcomes for those at risk for cardiotoxicity, or with established cardiovascular disease. Finally, we consider future directions in the field given the wave of new anticancer therapies.
Collapse
Affiliation(s)
- John Alan Gambril
- Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH, USA; Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA. https://twitter.com/GambrilAlan
| | - Aaron Chum
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA; Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, 473 West 12th Avenue, Suite 200, Columbus, OH 43210, USA
| | - Akash Goyal
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA; Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, 473 West 12th Avenue, Suite 200, Columbus, OH 43210, USA. https://twitter.com/agoyalMD
| | - Patrick Ruz
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA; Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, 473 West 12th Avenue, Suite 200, Columbus, OH 43210, USA
| | - Katarzyna Mikrut
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA. https://twitter.com/KatieMikrut
| | - Orlando Simonetti
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA; Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, 473 West 12th Avenue, Suite 200, Columbus, OH 43210, USA; Department of Internal Medicine, The Ohio State University Medical Center, Columbus, OH, USA; Department of Radiology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Hardeep Dholiya
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA; Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, 473 West 12th Avenue, Suite 200, Columbus, OH 43210, USA. https://twitter.com/Hardeep_10
| | - Brijesh Patel
- Division of Cardiovascular Medicine, Davis Heart & Lung Research Institute, 473 West 12th Avenue, Suite 200, Columbus, OH 43210, USA; Cardio-Oncology Program, Heart and Vascular Institute, West Virginia University, Morgantown, WV, USA
| | - Daniel Addison
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA; Division of Cancer Prevention and Control, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
18
|
Kwan JM, Oikonomou EK, Henry ML, Sinusas AJ. Multimodality Advanced Cardiovascular and Molecular Imaging for Early Detection and Monitoring of Cancer Therapy-Associated Cardiotoxicity and the Role of Artificial Intelligence and Big Data. Front Cardiovasc Med 2022; 9:829553. [PMID: 35369354 PMCID: PMC8964995 DOI: 10.3389/fcvm.2022.829553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer mortality has improved due to earlier detection via screening, as well as due to novel cancer therapies such as tyrosine kinase inhibitors and immune checkpoint inhibitions. However, similarly to older cancer therapies such as anthracyclines, these therapies have also been documented to cause cardiotoxic events including cardiomyopathy, myocardial infarction, myocarditis, arrhythmia, hypertension, and thrombosis. Imaging modalities such as echocardiography and magnetic resonance imaging (MRI) are critical in monitoring and evaluating for cardiotoxicity from these treatments, as well as in providing information for the assessment of function and wall motion abnormalities. MRI also allows for additional tissue characterization using T1, T2, extracellular volume (ECV), and delayed gadolinium enhancement (DGE) assessment. Furthermore, emerging technologies may be able to assist with these efforts. Nuclear imaging using targeted radiotracers, some of which are already clinically used, may have more specificity and help provide information on the mechanisms of cardiotoxicity, including in anthracycline mediated cardiomyopathy and checkpoint inhibitor myocarditis. Hyperpolarized MRI may be used to evaluate the effects of oncologic therapy on cardiac metabolism. Lastly, artificial intelligence and big data of imaging modalities may help predict and detect early signs of cardiotoxicity and response to cardioprotective medications as well as provide insights on the added value of molecular imaging and correlations with cardiovascular outcomes. In this review, the current imaging modalities used to assess for cardiotoxicity from cancer treatments are discussed, in addition to ongoing research on targeted molecular radiotracers, hyperpolarized MRI, as well as the role of artificial intelligence (AI) and big data in imaging that would help improve the detection and prognostication of cancer-treatment cardiotoxicity.
Collapse
Affiliation(s)
- Jennifer M. Kwan
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Evangelos K. Oikonomou
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Mariana L. Henry
- Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Albert J. Sinusas
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| |
Collapse
|
19
|
Martinez DSL, Noseworthy PA, Akbilgic O, Herrmann J, Ruddy KJ, Hamid A, Maddula R, Singh A, Davis R, Gunturkun F, Jefferies JL, Brown SA. Artificial intelligence opportunities in cardio-oncology: Overview with spotlight on electrocardiography. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 15:100129. [PMID: 35721662 PMCID: PMC9202996 DOI: 10.1016/j.ahjo.2022.100129] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 01/21/2023]
Abstract
Cardiovascular disease is a leading cause of death among cancer survivors, second only to cancer recurrence or development of new tumors. Cardio-oncology has therefore emerged as a relatively new specialty focused on prevention and management of cardiovascular consequences of cancer therapies. Yet challenges remain regarding precision and accuracy with predicting individuals at highest risk for cardiotoxicity. Barriers such as access to care also limit screening and early diagnosis to improve prognosis. Thus, developing innovative approaches for prediction and early detection of cardiovascular illness in this population is critical. In this review, we provide an overview of the present state of machine learning applications in cardio-oncology. We begin by outlining some factors that should be considered while utilizing machine learning algorithms. We then examine research in which machine learning has been applied to improve prediction of cardiac dysfunction in cancer survivors. We also highlight the use of artificial intelligence (AI) in conjunction with electrocardiogram (ECG) to predict cardiac malfunction and also atrial fibrillation (AF), and we discuss the potential role of wearables. Additionally, the article summarizes future prospects and critical takeaways for the application of machine learning in cardio-oncology. This study is the first in a series on artificial intelligence in cardio-oncology, and complements our manuscript on echocardiography and other forms of imaging relevant to cancer survivors cared for in cardiology clinical practice.
Collapse
Affiliation(s)
- Daniel Sierra-Lara Martinez
- Coronary Care Unit, National Institute of Cardiology/Instituto Nacional de Cardiologia, Ciudad de Mexico, Mexico
| | | | - Oguz Akbilgic
- Department of Health Informatics and Data Science, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Wake Forest, NC, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Ashima Singh
- Institute of Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert Davis
- Center for Biomedical Informatics, University of Tennessee Health Sciences Center, USA
| | - Fatma Gunturkun
- Center for Biomedical Informatics, University of Tennessee Health Sciences Center, USA
| | - John L. Jefferies
- Division of Cardiovascular Diseases, University of Tennessee Health Sciences Center, USA
- Department of Epidemiology, St. Jude Children's Research Hospital, USA
| | - Sherry-Ann Brown
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
20
|
Berkman AM, Hildebrandt MA, Landstrom AP. The genetic underpinnings of anthracycline-induced cardiomyopathy predisposition. Clin Genet 2021; 100:132-143. [PMID: 33871046 PMCID: PMC9902211 DOI: 10.1111/cge.13968] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/24/2021] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
Anthracyclines, chemotherapeutic agents that have contributed to significant improvements in cancer survival, also carry risk of both acute and chronic cardiotoxicity. This has led to significantly elevated risks of cardiac morbidity and mortality among cancer survivors treated with these agents. Certain treatment related, demographic, and medical factors increase an individual's risk of anthracycline induced cardiotoxicity; however, significant variability among those affected suggests that there is an underlying genetic predisposition to anthracycline induced cardiotoxicity. The current narrative review seeks to summarize the literature to date that has identified genetic variants associated with anthracycline induced cardiotoxicity. These include variants found in genes that encode proteins associated with anthracycline transportation and metabolism, those that encode proteins associated with the generation of reactive oxygen species, and those known to be associated with cardiac disease. While there is strong evidence that susceptibility to anthracycline induced cardiotoxicity has genetic underpinnings, the majority of work to date has been candidate gene analyses. Future work should focus on genome-wide analyses including genome-wide association and sequencing-based studies to confirm and expand these findings.
Collapse
Affiliation(s)
- Amy M. Berkman
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, 2301 Erwin Drive, Durham, North Carolina, United States
| | - Michelle A.T. Hildebrandt
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas, United States
| | - Andrew P. Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, 2301 Erwin Drive, Durham, North Carolina, United States
- Department of Cell Biology, Duke University School of Medicine, 2301 Erwin Drive, Durham, North Carolina, United States
| |
Collapse
|
21
|
Hahn VS, Zhang KW, Sun L, Narayan V, Lenihan DJ, Ky B. Heart Failure With Targeted Cancer Therapies: Mechanisms and Cardioprotection. Circ Res 2021; 128:1576-1593. [PMID: 33983833 DOI: 10.1161/circresaha.121.318223] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Oncology has seen growing use of newly developed targeted therapies. Although this has resulted in dramatic improvements in progression-free and overall survival, challenges in the management of toxicities related to longer-term treatment of these therapies have also become evident. Although a targeted approach often exploits the differences between cancer cells and noncancer cells, overlap in signaling pathways necessary for the maintenance of function and survival in multiple cell types has resulted in systemic toxicities. In particular, cardiovascular toxicities are of important concern. In this review, we highlight several targeted therapies commonly used across a variety of cancer types, including HER2 (human epidermal growth factor receptor 2)+ targeted therapies, tyrosine kinase inhibitors, immune checkpoint inhibitors, proteasome inhibitors, androgen deprivation therapies, and MEK (mitogen-activated protein kinase kinase)/BRAF (v-raf murine sarcoma viral oncogene homolog B) inhibitors. We present the oncological indications, heart failure incidence, hypothesized mechanisms of cardiotoxicity, and potential mechanistic rationale for specific cardioprotective strategies.
Collapse
Affiliation(s)
- Virginia S Hahn
- Division of Cardiology, Johns Hopkins School of Medicine, Baltimore, MD (V.S.H.)
| | - Kathleen W Zhang
- Cardio-Oncology Center of Excellence, Washington University, St Louis, MO (K.W.Z., D.J.L.)
| | - Lova Sun
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Vivek Narayan
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Daniel J Lenihan
- Cardio-Oncology Center of Excellence, Washington University, St Louis, MO (K.W.Z., D.J.L.)
| | - Bonnie Ky
- Penn Cardio-Oncology Translational Center of Excellence, Abramson Cancer Center (L.S., V.N., B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Division of Cardiovascular Medicine (B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Division of Biostatistics (B.K.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| |
Collapse
|