1
|
Jeon KH, Kang D, Shin DW, Kim H, Choi HL, Yoon SE, Park H, Jeong SM, Cho J. Cardiovascular disease in adolescent and young adult survivors of non-Hodgkin lymphoma in Korea. J Cancer Surviv 2025:10.1007/s11764-025-01816-2. [PMID: 40377886 DOI: 10.1007/s11764-025-01816-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Accepted: 04/25/2025] [Indexed: 05/18/2025]
Abstract
PURPOSE Long-term cardiovascular risk in adolescent and young adult (AYA) survivors of non-Hodgkin lymphoma (NHL) remains insufficiently characterized. This retrospective cohort study investigated the incidence of cardiovascular disease (CVD) among AYA survivors of NHL. METHODS We identified 4553 individuals aged 15-39 years diagnosed with NHL between 2006 and 2019 using the Korean National Health Insurance System database. A control group of 13,659 individuals without a history of cancer or CVD was selected using 1:3 matching based on age, sex, and residential area. The primary outcomes were major adverse cardiovascular events such as myocardial infarction, cardiomyopathy, heart failure, ischemic stroke, and hemorrhagic stroke. Hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using Cox proportional hazard models. RESULTS The mean (SD) age of the participants was 29.5 (6.8) years, and 59% were male. Over a median follow-up of 6.5 years, survivors of NHL had significantly higher risks of cardiomyopathy (HR 6.77; 95% CI 3.44-13.33), heart failure (HR 4.90; 95% CI 3.45-6.97), and hemorrhagic stroke (HR 3.14; 95% CI 1.75-5.65), compared to controls. In the subgroup analyses stratified by treatment modality, the highest risks were observed among patients who underwent hematopoietic stem cell transplantation, which involved high-dose chemotherapy with or without radiotherapy. The risk of myocardial infarction and ischemic stroke did not increase significantly. CONCLUSION AYA survivors of NHL had a significantly higher risk of CVD, including cardiomyopathy, heart failure, and hemorrhagic stroke, than the general population. IMPLICATIONS FOR CANCER SURVIVORS Long-term cardiovascular surveillance is essential for AYA survivors of NHL, particularly those receiving intensive treatment.
Collapse
Affiliation(s)
- Keun Hye Jeon
- Department of Family Medicine, CHA Gumi Medical Center, CHA University School of Medicine, Gumi, Republic of Korea
| | - Danbee Kang
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Dong Wook Shin
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Family Medicine/Supportive Care Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Hyunsoo Kim
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hea Lim Choi
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Family Medicine/Executive Healthcare Clinic, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Eun Yoon
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyunkyung Park
- Department of Hematology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Su-Min Jeong
- Department of Medicine, Seoul National University College of Medicine, 1, Gwanak-Ro, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Department of Family Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Juhee Cho
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Center for Clinical Epidemiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
2
|
Chen JK, Ramesh S, Islam MN, Shibu MA, Kuo CH, Hsieh DJY, Lin SZ, Kuo WW, Huang CY, Ho TJ. Artemisia argyi mitigates doxorubicin-induced cardiotoxicity by inhibiting mitochondrial dysfunction through the IGF-IIR/Drp1/GATA4 signaling pathway. Biotechnol Appl Biochem 2025; 72:388-401. [PMID: 39375847 DOI: 10.1002/bab.2671] [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: 06/04/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/09/2024]
Abstract
Doxorubicin (DOX) is mostly utilized as a wide range of antitumor anthracycline to treat different cancers. The severe antagonistic impacts of DOX on cardiotoxicity constrain its clinical application. Many mechanisms are involved in cardiac toxicity induced by DOX in the human body. Mitochondria is a central part of fatty acid and glucose metabolism. Thus, impaired mitochondrial metabolism can increase heart failure risk, which can play a vital role in cardiomyocyte mitochondrial dysfunction. This study aimed to assess the possible cardioprotective effect of water-extracted Artemisia argyi (AA) against the side effect of DOX in H9c2 cells and whether these protective effects are mediated through IGF-IIR/Drp1/GATA4 signaling pathways. Although several studies proved that AA extract has benefits for various diseases, its cardiac effects have not yet been identified. The H9c2 cells were exposed to 1 μM to establish a model of cardiac toxicity. The results revealed that water-extracted AA could block the expression of IGF-IIR/calcineurin signaling pathways induced by DOX. Notably, our results also showed that AA treatment markedly attenuated Akt phosphorylation and cleaved caspase 3, and the nuclear translocation markers NFATC3 and p-GATA4. Using actin staining for hypertrophy, we determined that AA can reduce the effect of mitochondrial reactive oxygen species and cell size. These findings suggest that water-extracted AA could be a suitable candidate for preventing DOX-induced cardiac damage.
Collapse
Affiliation(s)
- Jhong-Kuei Chen
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Samiraj Ramesh
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Research and Innovation, Institute of Biotechnology, Saveetha School of Engineering (SSE), Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai, India
| | - Md Nazmul Islam
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | | | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, University of Taipei, Taipei, Taiwan
| | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shinn-Zong Lin
- Bioinnovation Center, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Neurosurgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Life Sciences, China Medical University, Taichung, Taiwan
- Ph.D. Program for Biotechnology Industry, China Medical University, Taichung, Taiwan
- School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University, Hualien, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Integration Center of Traditional Chinese and Modern Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
3
|
Makwana B, Malode A, Khadke S, Patel V, Shah R, Patel M, Parikh A, Dani SS, Ganatra S. Cardiac Complications of Immune Checkpoint Inhibitors and Chimeric Antigen Receptor T Cell Therapy. Cardiol Clin 2025; 43:151-167. [PMID: 39551556 DOI: 10.1016/j.ccl.2024.07.001] [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] [Indexed: 11/19/2024]
Abstract
Immune checkpoint inhibitors and chimeric antigen receptor T-cell therapy have revolutionized cancer treatment but can cause life-threatening cardiovascular toxicities through immune-related adverse events. Myocarditis is the most common and potentially fatal toxicity with immune checkpoint inhibitors. T-cell therapies can potentially lead to cytokine release syndrome. Diagnosis of ICI-myocarditis requires a multimodal approach, including biomarkers, imaging, and endomyocardial biopsy, while CRS is characterized by a clinical syndrome resembling distributive shock. Management involves discontinuing the offending therapy, immunosuppression with corticosteroids for ICI-myocarditis, and interleukin-6 antagonists for CRS. Collaboration between oncologists and cardiologists is crucial for early recognition and prompt treatment.
Collapse
Affiliation(s)
- Bhargav Makwana
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Aishwarya Malode
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Sumanth Khadke
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Vahin Patel
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Rushin Shah
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Manav Patel
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Aneri Parikh
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Sourbha S Dani
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Sarju Ganatra
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA; Department of Medicine (Research), Cardio-Oncology Program, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA.
| |
Collapse
|
4
|
Li Z, Xing J, Ma X, Zhang W, Wang C, Wang Y, Qi X, Liu Y, Jian D, Cheng X, Zhu Y, Shi C, Guo Y, Zhao H, Jiang W, Tang H. An orally administered bacterial membrane protein nanodrug ameliorates doxorubicin cardiotoxicity through alleviating impaired intestinal barrier. Bioact Mater 2024; 37:517-532. [PMID: 38698916 PMCID: PMC11063951 DOI: 10.1016/j.bioactmat.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/07/2024] [Accepted: 03/20/2024] [Indexed: 05/05/2024] Open
Abstract
The cardiotoxicity caused by Dox chemotherapy represents a significant limitation to its clinical application and is a major cause of late death in patients undergoing chemotherapy. Currently, there are no effective treatments available. Our analysis of 295 clinical samples from 132 chemotherapy patients and 163 individuals undergoing physical examination revealed a strong positive correlation between intestinal barrier injury and the development of cardiotoxicity in chemotherapy patients. We developed a novel orally available and intestinal targeting protein nanodrug by assembling membrane protein Amuc_1100 (obtained from intestinal bacteria Akkermansia muciniphila), fluorinated polyetherimide, and hyaluronic acid. The protein nanodrug demonstrated favorable stability against hydrolysis compared with free Amuc_1100. The in vivo results demonstrated that the protein nanodrug can alleviate Dox-induced cardiac toxicity by improving gut microbiota, increasing the proportion of short-chain fatty acid-producing bacteria from the Lachnospiraceae family, and further enhancing the levels of butyrate and pentanoic acids, ultimately regulating the homeostasis repair of lymphocytes in the spleen and heart. Therefore, we believe that the integrity of the intestinal barrier plays an important role in the development of chemotherapy-induced cardiotoxicity. Protective interventions targeting the intestinal barrier may hold promise as a general clinical treatment regimen for reducing Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zhen Li
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Junyue Xing
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xiaohan Ma
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Wanjun Zhang
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Chuan Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Yingying Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xinkun Qi
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Yanhui Liu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou, Henan, 450003, China
| | - Dongdong Jian
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Xiaolei Cheng
- Department of Anesthesiology, Affiliated Drum Tower Hospital of Medical School of Nanjing University, Nanjing, Jiangsu, 210008, China
| | - Yanjie Zhu
- Department of Pathology, Central Hospital of Kaifeng City, Kaifeng, Henan, 475000, China
| | - Chao Shi
- Henan Key Laboratory of Molecular Pathology, Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Yongjun Guo
- Henan Key Laboratory of Molecular Pathology, Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan, 450008, China
| | - Huan Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Jiang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative Medicine, Central China Subcenter of National Center for Cardiovascular Diseases, Henan Cardiovascular Disease Center, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
- Henan Key Laboratory of Chronic Disease Management, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
- Zhengzhou Key Laboratory of Cardiovascular Aging, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 451464, China
| |
Collapse
|
5
|
Cannavale KL, Xu L, Nau CL, Armenian SH, Bhatia S, Wong FL, Huang PYS, Cooper R, Chao CR. Neighborhood factors associated with late effects among survivors of adolescent and young adult cancer. J Cancer Surviv 2024:10.1007/s11764-024-01596-1. [PMID: 38839694 DOI: 10.1007/s11764-024-01596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/05/2024] [Indexed: 06/07/2024]
Abstract
PURPOSE The purpose of this study is to evaluate the associations between neighborhood income, education, and neighborhood racial composition (measured as a low percentage of white residents) and risk of developing cardiovascular diseases (CVD), diabetes (DM), and severe depression among survivors of AYA cancer and matched non-cancer peers. METHODS Two-year survivors of AYA cancers diagnosed at age 15-39 yrs at Kaiser Permanente Southern California (diagnosed 2000-2012) and individually matched (1:13) non-cancer subjects were included. The development of CVD, DM, and severe depression was ascertained via electronic health records. Neighborhood characteristics were obtained from census-based geocoded data. Cox regression evaluated associations between neighborhood characteristics and the health outcomes of interest among both the cancer survivors and the non-cancer comparison cohort and effect modification by cancer survivor status on these relationships. RESULTS Among cancer survivors (n = 6774), living in mostly non-white neighborhoods, was associated with risk of CVD (hazard ratio (HR) = 1.54 (95% CI 1.00-2.36)), while lower education level (HR = 1.41 (95% CI 1.02-1.94)) was associated with risk of severe depression. None of the neighborhood characteristics were associated with risk of DM. Effect modification was found for neighborhood education and risk of DM and severe depression. CONCLUSION When jointly considered, cancer survivors who resided in the most disadvantaged neighborhoods were at the highest risk of developing these health outcomes compared to other subgroups. IMPLICATIONS FOR CANCER SURVIVORS Our findings may inform screening strategy and addressing social determinants of health among AYA cancer survivors.
Collapse
Affiliation(s)
- Kimberly L Cannavale
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Lanfang Xu
- MedHealth Analytics Inc., Sugar Land, TX, USA
| | - Claudia L Nau
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA
| | - Saro H Armenian
- Department of Population Sciences, City of Hope, Duarte, CA, USA
- Department of Pediatrics, City of Hope, Duarte, CA, USA
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - F Lennie Wong
- Department of Population Sciences, City of Hope, Duarte, CA, USA
| | - Po-Yin Samuel Huang
- Department of Family Medicine, Los Angeles Medical Center, Kaiser Permanente Southern California, Los Angeles, CA, USA
| | - Robert Cooper
- Department of Pediatric Oncology, Los Angeles Medical Center, Kaiser Permanente Southern California, Los Angeles, CA, USA
| | - Chun R Chao
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA, USA.
- Department of Health Systems Science, Kaiser Permanente Bernard J Tyson School of Medicine, Pasadena, CA, USA.
| |
Collapse
|
6
|
Berkman AM, Andersen CR, Landstrom AP, Hildebrandt MA, Gilchrist SC, Roth ME. Cardiovascular Disease in Childhood, Adolescent, and Young Adult Cancer Survivors: The Impact of Family History of Premature Heart Disease. J Adolesc Young Adult Oncol 2024; 13:548-556. [PMID: 38261412 PMCID: PMC12054696 DOI: 10.1089/jayao.2023.0130] [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] [Indexed: 01/25/2024] Open
Abstract
Purpose: Childhood, adolescent, and young adult (CAYA) cancer survivors (age 0-39 years at diagnosis) are at increased risk of cardiovascular disease (CVD). Family history of early heart disease increases the risk of CVD in the general population; however, it is unknown whether this association is seen in CAYA cancer survivors. Methods: Self-report data from the National Health and Nutrition Examination Survey (2005-2018) were used to identify CAYA survivors (>5 years post-diagnosis). The risk of CVD based on family history status (parent or sibling with a diagnosis of heart attack or angina before age 50 years), personal sociodemographic factors, personal medical history factors, and personal behavioral risk factors was determined using logistic regression models. Results: Included were 95 CAYA survivors with CVD and 491 CAYA survivors without CVD. The odds of CVD were significantly higher in survivors with a first-degree family history of early heart disease (odds ratio [OR]: 2.06, 95% confidence interval [CI]: 1.14-3.74). A history of diabetes (OR: 2.61, 95% CI: 1.41-4.84), hypertension (OR: 1.81, 95% CI: 1.04-3.16), and any smoking (OR: 2.19, 95% CI: 1.19-4.02) was also associated with higher odds of CVD in CAYA survivors. Reporting any physical activity in the past month was associated with lower odds (OR: 0.54, 95% CI: 0.30-0.97) of CVD. Conclusions: Family history of early heart disease was associated with increased odds of CVD in CAYA cancer survivors. Obtaining complete and accurate family history information is important both at time of diagnosis and throughout follow-up.
Collapse
Affiliation(s)
- Amy M. Berkman
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Clark R. Andersen
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew P. Landstrom
- Division of Pediatric Cardiology, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Michelle A.T. Hildebrandt
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Susan C. Gilchrist
- Department of Clinical Cancer Prevention and Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael E. Roth
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
7
|
Caudal A, Snyder MP, Wu JC. Harnessing human genetics and stem cells for precision cardiovascular medicine. CELL GENOMICS 2024; 4:100445. [PMID: 38359791 PMCID: PMC10879032 DOI: 10.1016/j.xgen.2023.100445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/22/2023] [Accepted: 10/25/2023] [Indexed: 02/17/2024]
Abstract
Human induced pluripotent stem cell (iPSC) platforms are valuable for biomedical and pharmaceutical research by providing tissue-specific human cells that retain patients' genetic integrity and display disease phenotypes in a dish. Looking forward, combining iPSC phenotyping platforms with genomic and screening technologies will continue to pave new directions for precision medicine, including genetic prediction, visualization, and treatment of heart disease. This review summarizes the recent use of iPSC technology to unpack the influence of genetic variants in cardiovascular pathology. We focus on various state-of-the-art genomic tools for cardiovascular therapies-including the expansion of genetic toolkits for molecular interrogation, in vitro population studies, and function-based drug screening-and their current applications in patient- and genome-edited iPSC platforms that are heralding new avenues for cardiovascular research.
Collapse
Affiliation(s)
- Arianne Caudal
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Greenstone Biosciences, Palo Alto, CA 94304, USA.
| |
Collapse
|
8
|
Fonoudi H, Jouni M, Cejas RB, Magdy T, Blancard M, Ge N, Shah DA, Lyra-Leite DM, Neupane A, Gharib M, Jiang Z, Sapkota Y, Burridge PW. Functional Validation of Doxorubicin-Induced Cardiotoxicity-Related Genes. JACC CardioOncol 2024; 6:38-50. [PMID: 38510289 PMCID: PMC10950437 DOI: 10.1016/j.jaccao.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 03/22/2024] Open
Abstract
Background Genome-wide association studies and candidate gene association studies have identified more than 180 genetic variants statistically associated with anthracycline-induced cardiotoxicity (AIC). However, the lack of functional validation has hindered the clinical translation of these findings. Objectives The aim of this study was to functionally validate all genes associated with AIC using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Methods Through a systemic literature search, 80 genes containing variants significantly associated with AIC were identified. Additionally, 3 more genes with potential roles in AIC (GSTM1, CBR1, and ERBB2) were included. Of these, 38 genes exhibited expression in human fetal heart, adult heart, and hiPSC-CMs. Using clustered regularly interspaced short palindromic repeats/Cas9-based genome editing, each of these 38 genes was systematically knocked out in control hiPSC-CMs, and the resulting doxorubicin-induced cardiotoxicity (DIC) phenotype was assessed using hiPSC-CMs. Subsequently, functional assays were conducted for each gene knockout on the basis of hypothesized mechanistic implications in DIC. Results Knockout of 26 genes increased the susceptibility of hiPSC-CMs to DIC. Notable genes included efflux transporters (ABCC10, ABCC2, ABCB4, ABCC5, and ABCC9), well-established DIC-associated genes (CBR1, CBR3, and RAC2), and genome-wide association study-discovered genes (RARG and CELF4). Conversely, knockout of ATP2B1, HNMT, POR, CYBA, WDR4, and COL1A2 had no significant effect on the in vitro DIC phenotype of hiPSC-CMs. Furthermore, knockout of the uptake transporters (SLC28A3, SLC22A17, and SLC28A1) demonstrated a protective effect against DIC. Conclusions The present findings establish a comprehensive platform for the functional validation of DIC-associated genes, providing insights for future studies in DIC variant associations and potential mechanistic targets for the development of cardioprotective drugs.
Collapse
Affiliation(s)
- Hananeh Fonoudi
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mariam Jouni
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Romina B. Cejas
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Malorie Blancard
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ning Ge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Disheet A. Shah
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Davi M. Lyra-Leite
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Achal Neupane
- Department of Epidemiology and Cancer Control, St. Jude Children’s Hospital, Memphis, Tennessee, USA
| | - Mennat Gharib
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zhengxin Jiang
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children’s Hospital, Memphis, Tennessee, USA
| | - Paul W. Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
9
|
Abstract
Anthracycline-induced cardiotoxicity (AIC) is a serious and common side effect of anthracycline therapy. Identification of genes and genetic variants associated with AIC risk has clinical potential as a cardiotoxicity predictive tool and to allow the development of personalized therapies. In this review, we provide an overview of the function of known AIC genes identified by association studies and categorize them based on their mechanistic implication in AIC. We also discuss the importance of functional validation of AIC-associated variants in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) to advance the implementation of genetic predictive biomarkers. Finally, we review how patient-specific hiPSC-CMs can be used to identify novel patient-relevant functional targets and for the discovery of cardioprotectant drugs to prevent AIC. Implementation of functional validation and use of hiPSC-CMs for drug discovery will identify the next generation of highly effective and personalized cardioprotectants and accelerate the inclusion of approved AIC biomarkers into clinical practice.
Collapse
Affiliation(s)
- Romina B Cejas
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA;
| | - Kateryna Petrykey
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Paul W Burridge
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA;
| |
Collapse
|
10
|
Al-Ansari S, Stolze J, Bresters D, Brook AH, Laheij AMGA, Brand HS, Dahllöf G, Rozema FR, Raber-Durlacher JE. Late Complications in Long-Term Childhood Cancer Survivors: What the Oral Health Professional Needs to Know. Dent J (Basel) 2024; 12:17. [PMID: 38275678 PMCID: PMC10813876 DOI: 10.3390/dj12010017] [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: 11/29/2023] [Revised: 01/06/2024] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
With diagnostic and therapeutic advances, over 80% of children diagnosed with cancer become long-term survivors. As the number of childhood cancer survivors (CCS) continues to increase, dental practitioners become more likely to have CCS among their patients. CCS may develop late complications from damage caused by their cancer treatment to endocrine, cardiovascular, musculoskeletal, and other organ systems. These complications may surface decades after the completion of treatment. Adverse outcomes of childhood cancer treatment frequently involve oral and craniofacial structures including the dentition. Tooth development, salivary gland function, craniofacial growth, and temporomandibular joint function may be disturbed, increasing oral health risks in these individuals. Moreover, CCS are at risk of developing subsequent malignancies, which may manifest in or near the oral cavity. It is important that dental practitioners are aware of the childhood cancer history of their patients and have knowledge of potential late complications. Therefore, this narrative review aims to inform dental practitioners of late oral complications of cancer treatment modalities commonly used in pediatric oncology. Furthermore, selected common non-oral late sequelae of cancer therapy that could have an impact on oral health and on delivering dental care will be discussed.
Collapse
Affiliation(s)
- Sali Al-Ansari
- Department of Oral Medicine, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands; (S.A.-A.); (A.M.G.A.L.)
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC, University of Amsterdam, 1081 HZ Amsterdam, The Netherlands
- Department Maxillofacial Surgery, Fachklinik Horneide, 48157 Münster, Germany
| | - Juliette Stolze
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands; (J.S.); (H.S.B.)
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
- Department of Oral and Maxillofacial Surgery, Prosthodontics and Special Dental Care, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| | - Dorine Bresters
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands;
| | - Alan Henry Brook
- Adelaide Dental School, University of Adelaide, Adelaide 5005, Australia;
- Institute of Dentistry, Queen Mary University of London, London E12AD, UK
| | - Alexa M. G. A. Laheij
- Department of Oral Medicine, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands; (S.A.-A.); (A.M.G.A.L.)
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC, University of Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Henk S. Brand
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands; (J.S.); (H.S.B.)
| | - Göran Dahllöf
- Division of Orthodontics and Pediatric Dentistry, Karolinska Institutet, 14152 Huddinge, Sweden;
- Center for Oral Health Services and Research, Mid-Norway (TkMidt), 100098 Trondheim, Norway
| | - Frederik R. Rozema
- Department of Oral Medicine, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands; (S.A.-A.); (A.M.G.A.L.)
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC, University of Amsterdam, 1081 HZ Amsterdam, The Netherlands
| | - Judith E. Raber-Durlacher
- Department of Oral Medicine, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands; (S.A.-A.); (A.M.G.A.L.)
- Department of Oral and Maxillofacial Surgery, Amsterdam UMC, University of Amsterdam, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
11
|
Caru M, Dubois P, Curnier D, Andelfinger G, Krajinovic M, Laverdière C, Sinnett D, Périé D. Echocardiographic Parameters Associated With Cardiorespiratory Fitness and Physical Activity in Childhood Acute Lymphoblastic Leukemia Survivors. J Phys Act Health 2023; 20:1152-1161. [PMID: 37793652 DOI: 10.1123/jpah.2023-0100] [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: 03/09/2023] [Revised: 06/30/2023] [Accepted: 08/15/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND Children's exposure to chemotherapeutic agents causes several long-term adverse effects but physical activity has been evidenced to be an effective strategy to improve cardiac function. This cross-sectional study aimed to explore the association between physical activity levels, cardiorespiratory fitness, and cardiac parameters measured by echocardiography. METHODS Participants were 216n childhood acute lymphoblastic leukemia survivors who underwent a maximal cardiopulmonary exercise test and self-reported their daily minutes of moderate to vigorous physical activity. They underwent a complete transthoracic echocardiographic assessment. Systolic and diastolic function analysis and strain images analysis were performed. The associations were studied through the preventive fraction (examined with univariate crude and adjusted logistic regression models) of regular physical activity (≥150 min·wk-1) and adequate cardiorespiratory fitness levels (above the median ≥ 32.0 mL·kg-1·min-1) on cardiac parameters. RESULTS Crude analysis shows that regular physical activity was associated with a significant preventive fraction in mitral E/A ratio (56%; P = .013), while adjusted analyses highlighted a nonsignificant reduction of 74% to 37% in the prevalence of cardiac parameters associated with physical activity. Similar associations of adequate cardiorespiratory fitness on cardiac parameters were observed. Adjusted analyses revealed a nonsignificant reduction of 7% to 86% in the prevalence of cardiac parameters associated with cardiorespiratory fitness. CONCLUSION This study reports that regular physical activity and adequate cardiorespiratory fitness were associated with a higher preventive fraction. Thus, engaging in physical activity prevents childhood acute lymphoblastic leukemia survivors' cardiac dysfunctions. These findings are novel and clinically relevant in pediatric cardiooncology and provide additional evidence to strengthen the benefits of exercise as long-term care in childhood cancer survivors.
Collapse
Affiliation(s)
- Maxime Caru
- Laboratory of Pathophysiology of EXercise (LPEX), School of Kinesiology and Physical Activity Sciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
- Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
- Department of Mechanical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Pierre Dubois
- Department of Mechanical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| | - Daniel Curnier
- Laboratory of Pathophysiology of EXercise (LPEX), School of Kinesiology and Physical Activity Sciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada
- Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Gregor Andelfinger
- Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Maja Krajinovic
- Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Caroline Laverdière
- Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Daniel Sinnett
- Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
- Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| | - Delphine Périé
- Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
- Department of Mechanical Engineering, Polytechnique Montreal, Montreal, QC, Canada
| |
Collapse
|
12
|
Lazar DR, Cainap S, Maniu D, Blag C, Bota M, Lazar FL, Achim A, Colceriu MC, Zdrenghea M. Anthracycline's Effects on Heart Rate Variability in Children with Acute Lymphoblastic Leukemia: Early Toxicity Signs-Pilot Study. J Clin Med 2023; 12:7052. [PMID: 38002666 PMCID: PMC10672281 DOI: 10.3390/jcm12227052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Anthracycline treatments are known to cause cardiotoxic long-term side effects in cancer survivors. Recently, a decrease in heart rate variability (HRV) has been identified in these patients, signaling autonomic dysfunction and altered cardiac fitness. This study aimed at evaluating changes in HRV in children treated with anthracyclines. A total of 35 pediatric patients with acute lymphoblastic leukemia were evaluated by means of a 24 h Holter ECG, at baseline and after reaching half the total cumulative dose of doxorubicin equivalent (120 mg/m2). Parameters of HRV were assessed, as well as any arrhythmic episodes, bradycardia and tachycardia percentages. The results showed a significant decrease in both time-domain and frequency-domain HRV parameters, following anthracycline treatment. The low-frequency (LF) to high-frequency (HF) parameters' ratio also displayed a significant difference (p = 0.035), suggestive of early cardiac autonomic dysfunction. Of note, none of the patients presented symptoms of heart disease or elevated troponins, and only two patients presented echocardiographic signs of diastolic dysfunction. The present study showed that cardiac autonomic nervous system regulation is compromised in children treated with anthracyclines even before reaching the total cumulative dose. Therefore, HRV parameters could be the first indicators of subclinical cardiac toxicity, making Holter ECG monitoring of the oncological patient a necessity.
Collapse
Affiliation(s)
- Diana R. Lazar
- Department No. 11, Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Pediatric Cardiology, Emergency Hospital for Children, 400394 Cluj-Napoca, Romania
| | - Simona Cainap
- Department of Pediatric Cardiology, Emergency Hospital for Children, 400394 Cluj-Napoca, Romania
- Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Dana Maniu
- Biomolecular Physics Department, Faculty of Physics, “Babes-Bolyai” University, 400347 Cluj-Napoca, Romania
| | - Cristina Blag
- Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Pediatric Oncology and Hematology, Emergency Hospital for Children, 400394 Cluj-Napoca, Romania
| | - Madalina Bota
- Department of Mother and Child, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Pediatric Oncology and Hematology, Emergency Hospital for Children, 400394 Cluj-Napoca, Romania
| | - Florin-Leontin Lazar
- Department No. 5, Internal Medicine, Medical Clinic Number 1, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Alexandru Achim
- Department of Cardiology, “Niculae Stancioiu” Heart Institute, Motilor 19-21, 400001 Cluj-Napoca, Romania
| | - Marius C. Colceriu
- Department of Functional Biosciences, Discipline of Physiology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Mihnea Zdrenghea
- Department No. 11, Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
- Department of Hematology, “Ion Chiricuta” Oncology Institute, 400015 Cluj-Napoca, Romania
| |
Collapse
|
13
|
Cohen-Cutler S, Kaplan C, Olch A, Wong K, Malvar J, Constine LS, Freyer DR. Impact of Volumetric Dosimetry on the Projected Cost of Radiation-Related Late Effects Screening After Childhood Cancer: A Real-World Cohort Analysis. Oncologist 2023; 28:e784-e792. [PMID: 37284853 PMCID: PMC10485276 DOI: 10.1093/oncolo/oyad136] [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: 11/14/2022] [Accepted: 04/13/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND Screening guidelines for childhood cancer survivors treated with radiation currently rely on broad anatomic irradiated regions (IR) to determine risk for late effects. However, contemporary radiotherapy techniques use volumetric dosimetry (VD) to define organ-specific exposure, which supports more specific screening recommendations that could be less costly. PATIENTS AND METHODS This was a cross-sectional study of 132 patients treated with irradiation at Children's Hospital Los Angeles from 2000 to 2016. For 5 key organs (cochlea, breast, heart, lung, and colon), radiation exposure was determined retrospectively using both IR and VD methods. Under each method, Children's Oncology Group Long-Term Follow-Up Guidelines were used to identify organs flagged for screening and recommended screening tests. Projected screening costs incurred under each method were computed through age 65 using insurance claims data. RESULTS Median age at the end of treatment was 10.6 years (range, 1.4-20.4). Brain tumor was the most common diagnosis (45%) and head/brain the most common irradiated region (61%). For all 5 organs, use of VD rather than IR resulted in fewer recommended screening tests. This led to average cumulative estimated savings of $3769 (P = .099), with significant savings in patients with CNS tumors (P = .012). Among patients with savings, average savings were $9620 per patient (P = .016) and significantly more likely for females than males (P = .027). CONCLUSION Use of VD to enhance precision of guideline-based screening for radiation-related late effects permits fewer recommended screening tests and generates cost-savings.
Collapse
Affiliation(s)
- Sally Cohen-Cutler
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Cameron Kaplan
- Department of Medicine, Gehr Family Center for Health Systems Science and Innovation, University of Southern California, Los Angeles, CA, USA
| | - Arthur Olch
- Radiation Oncology Program, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kenneth Wong
- Radiation Oncology Program, Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Radiation Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jemily Malvar
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Louis S Constine
- Departments of Radiation Oncology and Pediatrics, James P Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - David R Freyer
- Cancer and Blood Disease Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Pediatrics and Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
14
|
Qian C, Srivastava DK, Pan J, Hudson MM, Rai SN. Estimating Transition Intensity Rate on Interval-censored Data Using Semi-parametric with EM Algorithm Approach. COMMUN STAT-THEOR M 2023; 53:6038-6054. [PMID: 39100716 PMCID: PMC11293461 DOI: 10.1080/03610926.2023.2239397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/17/2023] [Indexed: 08/06/2024]
Abstract
Phase IV clinical trials are designed to monitor long-term side effects of medical treatment. For instance, childhood cancer survivors treated with chest radiation and/or anthracycline are often at risk of developing cardiotoxicity during their adulthood. Often the primary focus of a study could be on estimating the cumulative incidence of a particular outcome of interest such as cardiotoxicity. However, it is challenging to evaluate patients continuously and usually, this information is collected through cross-sectional surveys by following patients longitudinally. This leads to interval-censored data since the exact time of the onset of the toxicity is unknown. Rai et al. computed the transition intensity rate using a parametric model and estimated parameters using maximum likelihood approach in an illness-death model. However, such approach may not be suitable if the underlying parametric assumptions do not hold. This manuscript proposes a semi-parametric model, with a logit relationship for the treatment intensities in two groups, to estimate the transition intensity rates within the context of an illness-death model. The estimation of the parameters is done using an EM algorithm with profile likelihood. Results from the simulation studies suggest that the proposed approach is easy to implement and yields comparable results to the parametric model.
Collapse
Affiliation(s)
- Chen Qian
- Biostatistics and Bioinformatics Facility, James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, 40202, USA
- Department of Biostatistics and Bioinformatics, University of Louisville, Louisville, Kentucky, 40202, USA
- AbbVie Inc, North Chicago, Illinois, 60064, USA
| | - Deo Kumar Srivastava
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, 38105, USA
| | - Jianmin Pan
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, Ohio, 45267, USA
- Cancer Data Science Center, Biostatistics and Informatics Shared Resources, College of Medicine, University of Cincinnati, Cincinnati, Ohio, 45267, USA
| | - Melissa M Hudson
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, Tennessee, 38105, USA
| | - Shesh N. Rai
- Division of Biostatistics and Bioinformatics, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, Ohio, 45267, USA
- Cancer Data Science Center, Biostatistics and Informatics Shared Resources, College of Medicine, University of Cincinnati, Cincinnati, Ohio, 45267, USA
| |
Collapse
|
15
|
Lei Y, VanPortfliet JJ, Chen YF, Bryant JD, Li Y, Fails D, Torres-Odio S, Ragan KB, Deng J, Mohan A, Wang B, Brahms ON, Yates SD, Spencer M, Tong CW, Bosenberg MW, West LC, Shadel GS, Shutt TE, Upton JW, Li P, West AP. Cooperative sensing of mitochondrial DNA by ZBP1 and cGAS promotes cardiotoxicity. Cell 2023; 186:3013-3032.e22. [PMID: 37352855 PMCID: PMC10330843 DOI: 10.1016/j.cell.2023.05.039] [Citation(s) in RCA: 114] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 02/03/2023] [Accepted: 05/26/2023] [Indexed: 06/25/2023]
Abstract
Mitochondrial DNA (mtDNA) is a potent agonist of the innate immune system; however, the exact immunostimulatory features of mtDNA and the kinetics of detection by cytosolic nucleic acid sensors remain poorly defined. Here, we show that mitochondrial genome instability promotes Z-form DNA accumulation. Z-DNA binding protein 1 (ZBP1) stabilizes Z-form mtDNA and nucleates a cytosolic complex containing cGAS, RIPK1, and RIPK3 to sustain STAT1 phosphorylation and type I interferon (IFN-I) signaling. Elevated Z-form mtDNA, ZBP1 expression, and IFN-I signaling are observed in cardiomyocytes after exposure to Doxorubicin, a first-line chemotherapeutic agent that induces frequent cardiotoxicity in cancer patients. Strikingly, mice lacking ZBP1 or IFN-I signaling are protected from Doxorubicin-induced cardiotoxicity. Our findings reveal ZBP1 as a cooperative partner for cGAS that sustains IFN-I responses to mitochondrial genome instability and highlight ZBP1 as a potential target in heart failure and other disorders where mtDNA stress contributes to interferon-related pathology.
Collapse
Affiliation(s)
- Yuanjiu Lei
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Jordyn J VanPortfliet
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Yi-Fan Chen
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Joshua D Bryant
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Ying Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | | | - Sylvia Torres-Odio
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Katherine B Ragan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Jingti Deng
- Departments of Medical Genetics and Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Armaan Mohan
- Departments of Medical Genetics and Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Bing Wang
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Olivia N Brahms
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Shawn D Yates
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | | | - Carl W Tong
- Department of Medical Physiology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Marcus W Bosenberg
- Departments of Pathology, Dermatology, and Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Laura Ciaccia West
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Gerald S Shadel
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Timothy E Shutt
- Departments of Medical Genetics and Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Jason W Upton
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX 77807, USA.
| |
Collapse
|
16
|
Corvest V, Marec-Bérard P, Lervat C, Pacquement H, Toulmonde M, Gentet JC, Laurence V, Cleirec M, Mansuy L, Bompas E, Castex MP, Taque S, Filhon B, Tabone MD, Verité C, Entz-Werle N, Saumet L, Guimard G, Pondrom M, Chevreau C, Flandrin J, Duranteau L, Rousset-Jablonski C, Brugières L, Jimenez M, Le Deley MC, Gaspar N, Fresneau B. Late toxicity comparison of alkylating-based maintenance regimen with cyclophosphamide (VAC) vs ifosfamide (VAI) in Ewing sarcoma survivors treated in the randomized clinical trial Euro-EWING99-R1 in France. Int J Cancer 2023; 152:1659-1667. [PMID: 36250317 PMCID: PMC10092329 DOI: 10.1002/ijc.34326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/12/2022] [Accepted: 09/07/2022] [Indexed: 11/06/2022]
Abstract
In Euro-EWING99-R1 randomized trial, cyclophosphamide was shown to be noninferior to ifosfamide in the consolidation of standard-risk Ewing sarcoma (SR-EWS) after a common induction with VIDE (vincristine-ifosfamide-doxorubicin-etoposide). We present the results of the late effects analysis of VAC (vincristine-dactinomycin-cyclophoshamide) vs VAI (vincristine-dactinomycin-ifosfamide) conducted in Euro-EWING99-R1 French cohort. Of 267 French randomized patients, 204 were alive and free-of-relapse at 5-years including 172 with available long-term follow-up data concerning cardiac, renal and/or gonadal functions (sex-ratio M/F = 1.3, median age at diagnosis = 14 years): 84 randomized in VAC (median cumulative doses: cyclophosphamide = 9.7 g/m2 , ifosfamide = 59.4 g/m2 ) and 88 in VAI (ifosfamide = 97.1 g/m2 ). With a median follow-up of 10 years (range = 5-17), five late relapses and five second malignancies were recorded. The 10-year event-free survival among 5-year free-of-relapse survivors was similar between VAC and VAI (93% vs 95%, P = .63). We estimated the 10-year cumulative probabilities of cardiac and kidney toxicities at 4.4% (95% confidence interval [95% CI] = 1.1%-7.6%) and 34.8% (95% CI = 26.8%-42.0%), respectively. Cardiac toxicity cumulative probability was similar in both arms, whereas kidney toxicity was higher in VAI (at 10 years, 43.0% vs 25.7%, P = .02), resulting from significant difference in glomerular toxicity (31.1% vs 13.1%, P < .01). At 10 years, gonadal toxicity was observed in 27% and 28% of pubertal men and women, respectively, without significant difference between VAC and VAI. Kidney and gonadal toxicities represent major issues in Euro-EWING99-R1, with significantly higher risk of kidney toxicities with VAI, without significant gonadal toxicity reduction. These results support the need to limit cumulative doses of both alkylating agents and to use mixed regimen as in VIDE-VAC or VDC/IE (vincristine-doxorubicin-cyclophoshamide/ifosfamide-etoposide).
Collapse
Affiliation(s)
- Victoria Corvest
- Department of Children and Adolescents Oncology, Gustave Roussy, Villejuif, France
| | - Perrine Marec-Bérard
- Department of Oncology for Child and Adolescent, Centre Léon Bérard, Pediatric Oncology and Hematology Institute (IHOPe), Lyon, France
| | - Cyril Lervat
- Centre Oscar Lambret, Unité d'Oncologie Pédiatrique, Lille, France
| | | | - Maud Toulmonde
- Département d'Oncologie Médicale, Institut Bergonié, Unités Sarcomes et Phases Précoces, Bordeaux, France
| | - Jean-Claude Gentet
- Department of Pediatric Hematology, Immunology and Oncology, APHM - La Timone Children's Hospital, Marseille, France
| | - Valérie Laurence
- Medical Oncology, Adolescents and Young Adults Unit, Institut Curie, Paris, France
| | - Morgane Cleirec
- Service d'Oncologie Pédiatrique, Centre Hospitalier Universitaire de Nantes, Nantes, France
| | - Ludovic Mansuy
- Department of Pediatric Hematology and Oncology, Nancy University Hospital, Vandœuvre-lès-Nancy, France
| | - Emmanuelle Bompas
- Department of Medical Oncology, Centre René Gauducheau, Nantes, France
| | | | - Sophie Taque
- Department of Pediatric Onco-Hematology, Rennes University Hospital, Rennes, France
| | - Bruno Filhon
- Department of Pediatric Hematology and Oncology, Rouen University Hospital, Rouen, France
| | - Marie-Dominique Tabone
- Service d'Hémato-Oncologie Pédiatrique, Hôpital Armand Trousseau - AP-HP, Sorbonne Université, Paris, France
| | - Cécile Verité
- Department of Pediatric and Adolescent Hematology and Oncology, Pellegrin Hospital, Bordeaux, France
| | - Natacha Entz-Werle
- Pédiatrie Onco-Hématologie, Hôpital Universitaire de Strasbourg, Strasbourg, France
| | - Laure Saumet
- Service d'Onco-Hématologie Pédiatrique, Hôpital Arnaud de Villeneuve, Montpellier, France
| | - Gregory Guimard
- Department of Paediatric Oncology/Hematology, CHU de Reims, American Hospital, Reims, France
| | - Morgane Pondrom
- Department of Pediatric Hemato-Oncology, Nice University Hospital, Nice, France
| | - Christine Chevreau
- Department of Medical Oncology, Institut Claudius Regaud IUCT-O, Toulouse, France
| | - Jennifer Flandrin
- Service de Gynécologie Adolescente et Jeune Adulte (GYNADO), Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Lise Duranteau
- Service de Gynécologie Adolescente et Jeune Adulte (GYNADO), Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - Christine Rousset-Jablonski
- Département de Chirurgie, Centre Léon Bérard, INSERM U1290, RESearch in HealthcAre PErformance (RESHAPE), Lyon, France
| | - Laurence Brugières
- Department of Children and Adolescents Oncology, Gustave Roussy, Villejuif, France
| | - Marta Jimenez
- Research and Development Department, Unicancer, Paris, France
| | - Marie-Cécile Le Deley
- Unité de Méthodologie et Biostatistiques, Centre Oscar Lambret, Lille, France.,Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| | - Nathalie Gaspar
- Department of Children and Adolescents Oncology, Gustave Roussy, Villejuif, France
| | - Brice Fresneau
- Department of Children and Adolescents Oncology, Gustave Roussy, Villejuif, France.,Université Paris-Saclay, UVSQ, Inserm, CESP, Villejuif, France
| |
Collapse
|
17
|
Caru M, Curnier D, Dubois P, Friedrich MG, Andelfinger G, Krajinovic M, Laverdière C, Sinnett D, Périé D. Cardiorespiratory Fitness and Cardiac Magnetic Resonance Imaging in Childhood Acute Lymphoblastic Leukemia Survivors. J Phys Act Health 2023; 20:522-530. [PMID: 36972702 DOI: 10.1123/jpah.2022-0179] [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: 04/04/2022] [Revised: 11/29/2022] [Accepted: 02/09/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND Childhood acute lymphoblastic leukemia survivors' anthracycline-induced cardiotoxicity could be prevented with good cardiorespiratory fitness levels and regular physical activity. This cross-sectional study aimed to assess the association between cardiorespiratory fitness and physical activity with cardiac magnetic resonance parameters. METHODS A total of 96 childhood acute lymphoblastic leukemia survivors underwent a maximal cardiopulmonary exercise test and answered physical activity questionnaires. We calculated the odds ratio of the preventive fraction of regular physical activity (≥150 min/wk) and adequate cardiorespiratory fitness levels (above the median ≥31.4 mL·kg-1·min-1) on cardiac magnetic resonance parameters (left ventricular [LV] and right ventricular [RV] morphological and functional parameters). RESULTS An adequate cardiorespiratory fitness was associated with a significant preventive fraction for LV (up to 84% for LV end-diastolic volume) and RV volumes (up to 88% for RV end-systolic volume). The adjusted analyses highlighted a preventive fraction of 36% to 91% between an adequate cardiorespiratory fitness and LV and RV parameters, late gadolinium enhancement fibrosis, and cardiac magnetic resonance relaxation times. No associations were reported with regular physical activity. CONCLUSIONS This study provides additional evidence regarding the benefits of an adequate cardiorespiratory fitness level for childhood cancer survivors' cardiac health.
Collapse
Affiliation(s)
- Maxime Caru
- Faculty of Medicine, Laboratory of Pathophysiology of EXercise (LPEX), School of Kinesiology and Physical Activity Sciences, University of Montreal, Montreal, QC,Canada
- Sainte-Justine University Health Center, Research Center, Montreal, QC,Canada
- Department of Mechanical Engineering, Polytechnique Montreal, Montreal, QC,Canada
| | - Daniel Curnier
- Faculty of Medicine, Laboratory of Pathophysiology of EXercise (LPEX), School of Kinesiology and Physical Activity Sciences, University of Montreal, Montreal, QC,Canada
- Sainte-Justine University Health Center, Research Center, Montreal, QC,Canada
| | - Pierre Dubois
- Department of Mechanical Engineering, Polytechnique Montreal, Montreal, QC,Canada
| | - Matthias G Friedrich
- Departments of Medicine and Diagnostic Radiology, Research Institute of the McGill University Health Centre, Montreal, QC,Canada
| | - Gregor Andelfinger
- Sainte-Justine University Health Center, Research Center, Montreal, QC,Canada
- Department of Pediatrics, University of Montreal, Montreal, QC,Canada
| | - Maja Krajinovic
- Sainte-Justine University Health Center, Research Center, Montreal, QC,Canada
- Department of Pediatrics, University of Montreal, Montreal, QC,Canada
| | - Caroline Laverdière
- Sainte-Justine University Health Center, Research Center, Montreal, QC,Canada
- Department of Pediatrics, University of Montreal, Montreal, QC,Canada
| | - Daniel Sinnett
- Sainte-Justine University Health Center, Research Center, Montreal, QC,Canada
- Department of Pediatrics, University of Montreal, Montreal, QC,Canada
| | - Delphine Périé
- Sainte-Justine University Health Center, Research Center, Montreal, QC,Canada
- Department of Mechanical Engineering, Polytechnique Montreal, Montreal, QC,Canada
| |
Collapse
|
18
|
Agostinucci K, Grant MKO, Melaku W, Nair C, Zordoky BN. Exposure to Doxorubicin Modulates the Cardiac Response to Isoproterenol in Male and Female Mice. Pharmaceuticals (Basel) 2023; 16:391. [PMID: 36986490 PMCID: PMC10058259 DOI: 10.3390/ph16030391] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/24/2023] [Accepted: 02/28/2023] [Indexed: 03/08/2023] Open
Abstract
Sex is a salient risk factor in the development of doxorubicin-induced cardiotoxicity. Sex differences in the heart's ability to respond to hypertrophic stimuli in doxorubicin-exposed animals have not been reported. We identified the sexual dimorphic effects of isoproterenol in mice pre-exposed to doxorubicin. Male and female intact or gonadectomized C57BL/6N mice underwent five weekly intraperitoneal injections of 4 mg/kg doxorubicin followed by a five-week recovery period. Fourteen days of subcutaneous isoproterenol injections (10 mg/kg/day) were administered after the recovery period. Echocardiography was used to assess heart function one and five weeks after the last doxorubicin injection and on the fourteenth day of isoproterenol treatment. Thereafter, mice were euthanized, and the hearts were weighed and processed for histopathology and gene expression analysis. Doxorubicin did not produce overt cardiac dysfunction in male or female mice before starting isoproterenol treatment. The chronotropic response to a single isoproterenol injection was blunted by doxorubicin, but the inotropic response was maintained in both males and females. Pre-exposure to doxorubicin caused cardiac atrophy in both control and isoproterenol-treated male mice but not in female mice. Counterintuitively, pre-exposure to doxorubicin abrogated isoproterenol-induced cardiac fibrosis. However, there were no sex differences in the expression of markers of pathological hypertrophy, fibrosis, or inflammation. Gonadectomy did not reverse the sexually dimorphic effects of doxorubicin. Additionally, pre-exposure to doxorubicin abrogated the hypertrophic response to isoproterenol in castrated male mice but not in ovariectomized female mice. Therefore, pre-exposure to doxorubicin caused male-specific cardiac atrophy that persisted after isoproterenol treatment, which could not be prevented by gonadectomy.
Collapse
Affiliation(s)
| | | | | | | | - Beshay N. Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
19
|
Bennati E, Girolami F, Spaziani G, Calabri GB, Favre C, Parrini I, Lucà F, Tamburini A, Favilli S. Cardio-Oncology in Childhood: State of the Art. Curr Oncol Rep 2022; 24:1765-1777. [PMID: 36181610 DOI: 10.1007/s11912-022-01329-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Cardio-oncology is an increasingly important field of cardiology that focuses on the detection, monitoring, and treatment of cardiovascular disease (CVD) occurring during and after oncological treatments. The survival rate for childhood cancer patients has dramatically increased thanks to new treatment protocols and cardiovascular (CV) sequelae represent the third most frequent cause of mortality in surviving patients. This study aims to provide a complete and updated review of all the main aspects of cardio-oncology in childhood and to highlight the critical issues. RECENT FINDINGS The problem of CV complications in childhood cancer survivors raises the need to make an early diagnosis of cardiotoxicity by the new imaging and laboratory techniques in order to intervene promptly and to implement pharmacological strategies and lifestyle changes to reduce or even to prevent cardiac injury. Furthermore, a stratification of CV risk, also including new predisposing factors such as the presence of some genetic mutations, is of paramount importance before undertaking oncological treatments. Besides, a systematic and personalized planning of long-term follow-up is fundamental to ensure a transition from pediatric to adult hospital and to avoid missed or late diagnosis of cardiomyopathy. We reviewed the main risk factors for cardiotoxicity in children, both traditional and emerging ones: the mechanisms of toxicity of both old and new antineoplastic therapies, the techniques for detecting cardiac damage, and the current evidence regarding pharmacological cardioprotection. At the end, we focused our attention on the existing guidelines and strategies about the long-term follow-up of childhood cancer survivors.
Collapse
Affiliation(s)
- Elena Bennati
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy.
| | - Francesca Girolami
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | - Gaia Spaziani
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | | | - Claudio Favre
- Department of Pediatric Hematology-Oncology, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | - Iris Parrini
- Cardiology Unit, Mauriziano Umberto I Hospital, Corso Turati 62, Turin, Italy
| | - Fabiana Lucà
- Department of Cardiology, Grande Ospedale Metropolitano, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Angela Tamburini
- Department of Pediatric Hematology-Oncology, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| | - Silvia Favilli
- Pediatric Cardiology Unit, Meyer Children's Hospital, Viale G. Pieraccini 24, Florence, Italy
| |
Collapse
|
20
|
Dhingra R, Rabinovich-Nikitin I, Rothman S, Guberman M, Gang H, Margulets V, Jassal DS, Alagarsamy KN, Dhingra S, Ripoll CV, Billia F, Diwan A, Javaheri A, Kirshenbaum LA. Proteasomal Degradation of TRAF2 Mediates Mitochondrial Dysfunction in Doxorubicin-Cardiomyopathy. Circulation 2022; 146:934-954. [PMID: 35983756 PMCID: PMC10043946 DOI: 10.1161/circulationaha.121.058411] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Cytokines such as tumor necrosis factor-α (TNFα) have been implicated in cardiac dysfunction and toxicity associated with doxorubicin (DOX). Although TNFα can elicit different cellular responses, including survival or death, the mechanisms underlying these divergent outcomes in the heart remain cryptic. The E3 ubiquitin ligase TRAF2 (TNF receptor associated factor 2) provides a critical signaling platform for K63-linked polyubiquitination of RIPK1 (receptor interacting protein 1), crucial for nuclear factor-κB (NF-κB) activation by TNFα and survival. Here, we investigate alterations in TNFα-TRAF2-NF-κB signaling in the pathogenesis of DOX cardiotoxicity. METHODS Using a combination of in vivo (4 weekly injections of DOX 5 mg·kg-1·wk-1) in C57/BL6J mice and in vitro approaches (rat, mouse, and human inducible pluripotent stem cell-derived cardiac myocytes), we monitored TNFα levels, lactate dehydrogenase, cardiac ultrastructure and function, mitochondrial bioenergetics, and cardiac cell viability. RESULTS In contrast to vehicle-treated mice, ultrastructural defects, including cytoplasmic swelling, mitochondrial perturbations, and elevated TNFα levels, were observed in the hearts of mice treated with DOX. While investigating the involvement of TNFα in DOX cardiotoxicity, we discovered that NF-κB was readily activated by TNFα. However, TNFα-mediated NF-κB activation was impaired in cardiac myocytes treated with DOX. This coincided with loss of K63- linked polyubiquitination of RIPK1 from the proteasomal degradation of TRAF2. Furthermore, TRAF2 protein abundance was markedly reduced in hearts of patients with cancer treated with DOX. We further established that the reciprocal actions of the ubiquitinating and deubiquitinating enzymes cellular inhibitors of apoptosis 1 and USP19 (ubiquitin-specific peptidase 19), respectively, regulated the proteasomal degradation of TRAF2 in DOX-treated cardiac myocytes. An E3-ligase mutant of cellular inhibitors of apoptosis 1 (H588A) or gain of function of USP19 prevented proteasomal degradation of TRAF2 and DOX-induced cell death. Furthermore, wild-type TRAF2, but not a RING finger mutant defective for K63-linked polyubiquitination of RIPK1, restored NF-κB signaling and suppressed DOX-induced cardiac cell death. Last, cardiomyocyte-restricted expression of TRAF2 (cardiac troponin T-adeno-associated virus 9-TRAF2) in vivo protected against mitochondrial defects and cardiac dysfunction induced by DOX. CONCLUSIONS Our findings reveal a novel signaling axis that functionally connects the cardiotoxic effects of DOX to proteasomal degradation of TRAF2. Disruption of the critical TRAF2 survival pathway by DOX sensitizes cardiac myocytes to TNFα-mediated necrotic cell death and DOX cardiotoxicity.
Collapse
Affiliation(s)
- Rimpy Dhingra
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Sonny Rothman
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Matthew Guberman
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Hongying Gang
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Victoria Margulets
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Davinder S. Jassal
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Keshav N. Alagarsamy
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
- Regenerative Medicine Program, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Sanjiv Dhingra
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
- Regenerative Medicine Program, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| | - Carla Valenzuela Ripoll
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Filio Billia
- Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada, Peter Munk Cardiac Center, University Health Network, Toronto, Ontario, Canada
| | - Abhinav Diwan
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Ali Javaheri
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO
| | - Lorrie A. Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
- Department of Pharmacology and Therapeutics, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre
| |
Collapse
|
21
|
Wang C, Shen Y, Ni J, Hu W, Yang Y. Effect of chronic stress on tumorigenesis and development. Cell Mol Life Sci 2022; 79:485. [PMID: 35974132 PMCID: PMC11071880 DOI: 10.1007/s00018-022-04455-3] [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: 04/08/2022] [Revised: 06/22/2022] [Accepted: 06/27/2022] [Indexed: 11/03/2022]
Abstract
Chronic stress activates the sympathetic nervous system (SNS) and hypothalamic-pituitary-adrenal (HPA) axis to aggravates tumorigenesis and development. Although the importance of SNS and HPA in maintaining homeostasis has already attracted much attention, there is still a lot remained unknown about the molecular mechanisms by which chronic stress influence the occurrence and development of tumor. While some researches have already concluded the mechanisms underlying the effect of chronic stress on tumor, complicated processes of tumor progression resulted in effects of chronic stress on various stages of tumor remains elusive. In this reviews we concluded recent research progresses of chronic stress and its effects on premalignancy, tumorigenesis and tumor development, we comprehensively summarized the molecular mechanisms in between. And we highlight the available treatments and potential therapies for stressed patients with tumor.
Collapse
Affiliation(s)
- Chen Wang
- State Key Laboratory of Natural Medicines, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, No. 639 Long Mian Avenue, Jiangning District, Nanjing, 211198, Jiangsu, People's Republic of China
| | - Yumeng Shen
- State Key Laboratory of Natural Medicines, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, No. 639 Long Mian Avenue, Jiangning District, Nanjing, 211198, Jiangsu, People's Republic of China
| | - Jiaping Ni
- State Key Laboratory of Natural Medicines, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, No. 639 Long Mian Avenue, Jiangning District, Nanjing, 211198, Jiangsu, People's Republic of China
| | - Weiwei Hu
- State Key Laboratory of Natural Medicines, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, No. 639 Long Mian Avenue, Jiangning District, Nanjing, 211198, Jiangsu, People's Republic of China.
- Lingang Laboratory, Shanghai, 200032, People's Republic of China.
| | - Yong Yang
- State Key Laboratory of Natural Medicines, Center for New Drug Safety Evaluation and Research, China Pharmaceutical University, No. 639 Long Mian Avenue, Jiangning District, Nanjing, 211198, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Bhagat A, Shrestha P, Jeyabal P, Peng Z, Watowich SS, Kleinerman ES. Doxorubicin-induced cardiotoxicity is mediated by neutrophils through release of neutrophil elastase. Front Oncol 2022; 12:947604. [PMID: 36033503 PMCID: PMC9400062 DOI: 10.3389/fonc.2022.947604] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
The mechanisms by which Doxorubicin (Dox) causes acute and late cardiotoxicity are not completely understood. One understudied area is the innate immune response, and in particular the role of neutrophils in Dox-induced cardiotoxicity. Here, using echocardiography, flow cytometry and immunofluorescence staining, we demonstrated increased infiltration of neutrophils that correlated with decreased heart function, disruption of vascular structures and increased collagen deposition in the heart after Dox treatment. Depleting neutrophils protected the heart from Dox-induced cardiotoxicity and changes in vascular structure. Furthermore, our data using neutrophil elastase (NE) knock-out mice and the NE inhibitor AZD9668 suggest that neutrophils cause this damage by releasing NE and that inhibiting NE can prevent Dox-induced cardiotoxicity. This work shows the role of neutrophils and NE in Doxorubicin-induced cardiotoxicity for the first time and suggests a new possible therapeutic intervention.
Collapse
Affiliation(s)
- Anchit Bhagat
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Pradeep Shrestha
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Prince Jeyabal
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Zhanglong Peng
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephanie S. Watowich
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eugenie S. Kleinerman
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Eugenie S. Kleinerman,
| |
Collapse
|
23
|
Cejas RB, Tamaño-Blanco M, Fontecha JE, Blanco JG. Impact of DYRK1A Expression on TNNT2 Splicing and Daunorubicin Toxicity in Human iPSC-Derived Cardiomyocytes. Cardiovasc Toxicol 2022; 22:701-712. [PMID: 35596909 PMCID: PMC9236996 DOI: 10.1007/s12012-022-09746-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/21/2022] [Indexed: 11/26/2022]
Abstract
Cardiac troponin T (encoded by TNNT2) is involved in the contraction of cardiomyocytes during beating. The alternative splicing of TNNT2 results in four transcript variants with differential Ca2+ sensitivity. The splicing of TNNT2 involves phosphorylation of the splicing factor SRSF6 by DYRK1A. Altered TNNT2 splicing patterns have been identified in failing human hearts. There is a paucity of studies describing DYRK1A-SRSF6-TNNT2 interplays in human cardiomyocytes. Also, it is not known whether the sensitivity of cardiomyocytes to cardiotoxic anthracyclines is modified in the context of variable DYRK1A-TNNT2 expression. In this study, we investigated the impact of DYRK1A on the endogenous expression of TNNT2 splicing variants in iPSC-derived cardiomyocytes. We also examined whether DYRK1A expression modifies the sensitivity of cardiomyocytes to the cardiotoxic drug daunorubicin (DAU). DYRK1A over-expression increased the abundance of TNNT2 fetal variants by ~ 58% whereas the abundance of the adult cTnT3 variant decreased by ~ 27%. High DYRK1A expression increased the phosphorylation of SRSF6 by ~ 25-65%. DAU cytotoxicity was similar between cardiomyocytes with variable levels of DYRK1A expression. DYRK1A over-expression ameliorated the impact of DAU on beating frequency. This study lays the foundation to further investigate the contribution of variable DYRK1A-TNNT2 expression to Ca2+ handling and beating in human cardiomyocytes.
Collapse
Affiliation(s)
- Romina Beatriz Cejas
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Miriam Tamaño-Blanco
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - John Edgar Fontecha
- Group for Applied Mathematical Modeling and Analytics (GAMMA), Industrial and Systems Engineering, The State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Javier Guillermo Blanco
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
24
|
El Moukhtari SH, Garbayo E, Fernández-Teijeiro A, Rodríguez-Nogales C, Couvreur P, Blanco-Prieto MJ. Nanomedicines and cell-based therapies for embryonal tumors of the nervous system. J Control Release 2022; 348:553-571. [PMID: 35705114 DOI: 10.1016/j.jconrel.2022.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 11/26/2022]
Abstract
Embryonal tumors of the nervous system are neoplasms predominantly affecting the pediatric population. Among the most common and aggressive ones are neuroblastoma (NB) and medulloblastoma (MB). NB is a sympathetic nervous system tumor, which is the most frequent extracranial solid pediatric cancer, usually detected in children under two. MB originates in the cerebellum and is one of the most lethal brain tumors in early childhood. Their tumorigenesis presents some similarities and both tumors often have treatment resistances and poor prognosis. High-risk (HR) patients require high dose chemotherapy cocktails associated with acute and long-term toxicities. Nanomedicine and cell therapy arise as potential solutions to improve the prognosis and quality of life of children suffering from these tumors. Indeed, nanomedicines have been demonstrated to efficiently reduce drug toxicity and improve drug efficacy. Moreover, these systems have been extensively studied in cancer research over the last few decades and an increasing number of anticancer nanocarriers for adult cancer treatment has reached the clinic. Among cell-based strategies, the clinically most advanced approach is chimeric-antigen receptor (CAR) T therapy for both pathologies, which is currently under investigation in phase I/II clinical trials. However, pediatric drug research is especially hampered due not only to ethical issues but also to the lack of efficient pre-clinical models and the inadequate design of clinical trials. This review provides an update on progress in the treatment of the main embryonal tumors of the nervous system using nanotechnology and cell-based therapies and discusses key issues behind the gap between preclinical studies and clinical trials in this specific area. Some directions to improve their translation into clinical practice and foster their development are also provided.
Collapse
Affiliation(s)
- Souhaila H El Moukhtari
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Elisa Garbayo
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Ana Fernández-Teijeiro
- Pediatric Onco-Hematology Unit, Hospital Universitario Virgen Macarena, School of Medicine, Universidad de Sevilla, Avenida Dr, Fedriani 3, 41009 Sevilla, Spain; Sociedad Española de Hematología y Oncología Pediátricas (SEHOP), Spain
| | - Carlos Rodríguez-Nogales
- School of Pharmaceutical Sciences, University of Geneva, Rue Michel-Servet 1, 1206 Geneva, Switzerland
| | - Patrick Couvreur
- Institut Galien Paris-Sud, UMRCNRS8612,Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry 92296, France
| | - María J Blanco-Prieto
- Department of Pharmaceutical Technology and Chemistry, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain.
| |
Collapse
|
25
|
Lamberg M, Rossman A, Bennett A, Painter S, Goodman R, MacLeod J, Maddula R, Rayan D, Doshi K, Bick A, Bailey S, Brown SA. Next Generation Risk Markers in Preventive Cardio-oncology. Curr Atheroscler Rep 2022; 24:443-456. [PMID: 35441347 PMCID: PMC10026729 DOI: 10.1007/s11883-022-01021-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/10/2022] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Cardiovascular disease (CVD) and cancer are the first and second most common causes of death within the USA. It is well established that a diagnosis of cancer increases risk and predisposes the patient to CVD, and vice versa. Despite these associations, cancer is not yet incorporated into current CVD risk calculators, necessitating additional CV risk markers for improved stratification in this at-risk population. In this review, we consider the utility of breast arterial calcification (BAC), coronary artery calcification (CAC), clonal hematopoiesis of indeterminate potential (CHIP), and cancer and cancer treatment in CVD risk assessment. RECENT FINDINGS There is evidence supporting the use of BAC, CAC, CHIP, and cancer and cancer treatment for improved CV risk stratification in patients with cancer and those who are being screened for cancer. BAC has been shown to predict CAC, coronary atherosclerotic plaque on coronary CTA, coronary artery stenosis on coronary angiography, and CVD events and accordingly enhances CVD risk stratification beyond the atherosclerotic CVD (ASCVD) risk pooled cohort equation. Additionally, CAC visualized on CT utilized for lung cancer screening, radiation planning, and cancer staging is predictive of coronary artery disease (CAD). Furthermore, CHIP can also be utilized in risk stratification, as the presence of CHIP carries a 40% increase in CV risk independent of traditional CV risk factors. Finally, cancer and many oncologic therapies confer a lifelong increased risk of CVD. We propose an emerging set of tools to be incorporated into the routine continuum of CVD risk assessment in individuals who have been treated for cancer or who are being screened for cancer development. In this review, we discuss BAC, CAC, CHIP, and cancer and cancer treatment as emerging risk markers in cardiovascular health assessment. Their effectiveness in predicting and influencing the burden of CVD will be discussed, along with suggestions on their incorporation into preventive cardio-oncology practice. Future research will focus on short- and long-term CVD outcomes in these populations.
Collapse
Affiliation(s)
- Morgan Lamberg
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA
| | | | | | - Sabrina Painter
- Department of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Rachel Goodman
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA
| | | | | | - David Rayan
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA
| | - Krishna Doshi
- Department of Medicine, Advocate Lutheran General Hospital, Park Ridge, IL, USA
| | - Alexander Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Simone Bailey
- Preventive Cardiology, Rochester Regional Health, Rochester, MN, USA
| | - Sherry-Ann Brown
- Cardio-Oncology & Preventive Cardiology Programs, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
26
|
Sitte V, Burkhardt B, Weber R, Kretschmar O, Hersberger M, Bergsträsser E, Christmann M. Advanced Imaging and New Cardiac Biomarkers in Long-term Follow-up After Childhood Cancer. J Pediatr Hematol Oncol 2022; 44:e374-e380. [PMID: 33828032 DOI: 10.1097/mph.0000000000002156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/04/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Pathologic ejection fraction (EF), shortening fraction (FS), and standard heart failure biomarkers (high sensitive troponin T and N-terminal brain natriuretic peptide) during follow-up after childhood cancer have been associated with irreversible cardiac damage. We aimed to evaluate strain imaging values by echocardiography and new biomarkers for heart failure with preserved ejection fraction (HFpEF) as potential more sensitive parameters for cardiac deterioration in childhood cancer survivors (CCS). MATERIALS AND METHODS Prospective study with 50 CCS (median 16.2 y) at a median follow-up of 13 years. In addition to standard echo and laboratory parameters for heart failure, strain measurements and new biomarkers, including myocardial inflammation (interleukin 6), extracellular matrix (ECM) remodeling (C-telopeptide for type I collagen, intact N-terminal propeptide of type III procollagen), and other heart failure biomarkers (galectin 3, solutable ST2, growth differentiation factor 15), were obtained and compared with 50 healthy controls. RESULTS No significant differences in EF, FS, high sensitive troponin T, N-terminal brain natriuretic peptide, interleukin 6, solutable ST2, and galectin 3 were found between study and control groups. In contrast, strain imaging showed significant differences between both groups (global longitudinal strainGLS -16.1% vs. -20.4%, P<0.0001; global circumferential strain -14.3 vs. -21.4%, P<0.0001), detecting 66% (global longitudinal strain) and 76% (global circumferential strain) of patients with pathologic values in contrast to 6% (EF) and 16% (FS) for standard parameters. Markers for disturbances of ECM remodeling (C-telopeptide for type I collagen, intact N-terminal propeptide of type III procollagen, each P<0.0001) and growth differentiation factor 15 (P<0.0001) were significantly different between the groups. CONCLUSION Strain imaging and new cardiac biomarkers used in HFpEF focusing on ECM remodeling appear to be more sensitive in detecting early remodeling processes in CCS than standard echo and laboratory parameters.
Collapse
Affiliation(s)
| | | | - Roland Weber
- Department of Paediatric Cardiology, Heart Center
| | | | | | - Eva Bergsträsser
- Department of Paediatric Oncology, University Children's Hospital, Zurich, Switzerland
| | | |
Collapse
|
27
|
Katzenstein HM, Malogolowkin MH, Krailo MD, Piao J, Towbin AJ, McCarville MB, Tiao GM, Dunn SP, Langham MR, McGahren ED, Finegold MJ, Ranganathan S, Weldon CB, Thompson PA, Trobaugh-Lotrario AD, O’Neill AF, Furman WL, Chung N, Randazzo J, Rodriguez-Galindo C, Meyers RL. Doxorubicin in combination with cisplatin, 5-flourouracil, and vincristine is feasible and effective in unresectable hepatoblastoma: A Children's Oncology Group study. Cancer 2022; 128:1057-1065. [PMID: 34762296 PMCID: PMC9066555 DOI: 10.1002/cncr.34014] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/01/2021] [Accepted: 10/07/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND The Children's Oncology Group (COG) adopted cisplatin, 5-flourouracil, and vincristine (C5V) as standard therapy after the INT-0098 legacy study showed statistically equivalent survival but less toxicity in comparison with cisplatin and doxorubicin. Subsequent experience demonstrated doxorubicin to be effective in patients with recurrent disease after C5V, and this suggested that it could be incorporated to intensify therapy for patients with advanced disease. METHODS In this nonrandomized, phase 3 COG trial, the primary aim was to explore the feasibility and toxicity of a novel therapeutic cisplatin, 5-flourouracil, vincristine, and doxorubicin (C5VD) regimen with the addition of doxorubicin to C5V for patients considered to be at intermediate risk. Patients were eligible if they had unresectable, nonmetastatic disease. Patients with a complete resection at diagnosis and local pathologic evidence of small cell undifferentiated histology were also eligible for an assessment of feasibility. RESULTS One hundred two evaluable patients enrolled between September 14, 2009, and March 12, 2012. Delivery of C5VD was feasible and tolerable: the mean percentages of the target doses delivered were 96% (95% CI, 94%-97%) for cisplatin, 96% (95% CI, 94%-97%) for 5-fluorouracil, 95% (95% CI, 93%-97%) for doxorubicin, and 90% (95% CI, 87%-93%) for vincristine. Toxicity was within expectations, with death as a first event in 1 patient. The most common adverse events were febrile neutropenia (n = 55 [54%]), infection (n = 48 [47%]), mucositis (n = 31 [30%]), hypokalemia (n = 39 [38%]), and elevated aspartate aminotransferase (n = 28 [27%]). The 5-year event-free and overall survival rates for the 93 patients who did not have complete resection at diagnosis were 88% (95% CI, 79%-93%) and 95% (95% CI, 87%-98%), respectively. CONCLUSIONS The addition of doxorubicin to the previous standard regimen of C5V is feasible, tolerable, and efficacious, and this suggests that C5VD is a good regimen for future clinical trials.
Collapse
Affiliation(s)
- Howard M Katzenstein
- Nemours Children’s Specialty Care and Wolfson Children’s Hospital, Jacksonville, FL
| | | | - Mark D Krailo
- University of Southern California Keck School of Medicine, Los Angeles, CA
| | - Jin Piao
- University of Southern California Keck School of Medicine, Los Angeles, CA
| | | | - M Beth McCarville
- University of Tennessee Health Science Center and St Jude Children’s Research Hospital, Memphis, TN
| | - Gregory M Tiao
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | | | - Max R Langham
- University of Tennessee Health Science Center and St Jude Children’s Research Hospital, Memphis, TN
| | | | | | | | | | | | | | - Allison F O’Neill
- Dana-Farber Cancer Institute, and Boston Children’s Hospital, Boston, MA
| | - Wayne L Furman
- University of Tennessee Health Science Center and St Jude Children’s Research Hospital, Memphis, TN
| | | | | | - Carlos Rodriguez-Galindo
- University of Tennessee Health Science Center and St Jude Children’s Research Hospital, Memphis, TN
| | | |
Collapse
|
28
|
Supplementing Soy-Based Diet with Creatine in Rats: Implications for Cardiac Cell Signaling and Response to Doxorubicin. Nutrients 2022; 14:nu14030583. [PMID: 35276943 PMCID: PMC8840593 DOI: 10.3390/nu14030583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/16/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Nutritional habits can have a significant impact on cardiovascular health and disease. This may also apply to cardiotoxicity caused as a frequent side effect of chemotherapeutic drugs, such as doxorubicin (DXR). The aim of this work was to analyze if diet, in particular creatine (Cr) supplementation, can modulate cardiac biochemical (energy status, oxidative damage and antioxidant capacity, DNA integrity, cell signaling) and functional parameters at baseline and upon DXR treatment. Here, male Wistar rats were fed for 4 weeks with either standard rodent diet (NORMAL), soy-based diet (SOY), or Cr-supplemented soy-based diet (SOY + Cr). Hearts were either freeze-clamped in situ or following ex vivo Langendorff perfusion without or with 25 μM DXR and after recording cardiac function. The diets had distinct cardiac effects. Soy-based diet (SOY vs. NORMAL) did not alter cardiac performance but increased phosphorylation of acetyl-CoA carboxylase (ACC), indicating activation of rather pro-catabolic AMP-activated protein kinase (AMPK) signaling, consistent with increased ADP/ATP ratios and lower lipid peroxidation. Creatine addition to the soy-based diet (SOY + Cr vs. SOY) slightly increased left ventricular developed pressure (LVDP) and contractility dp/dt, as measured at baseline in perfused heart, and resulted in activation of the rather pro-anabolic protein kinases Akt and ERK. Challenging perfused heart with DXR, as analyzed across all nutritional regimens, deteriorated most cardiac functional parameters and also altered activation of the AMPK, ERK, and Akt signaling pathways. Despite partial reprogramming of cell signaling and metabolism in the rat heart, diet did not modify the functional response to supraclinical DXR concentrations in the used acute cardiotoxicity model. However, the long-term effect of these diets on cardiac sensitivity to chronic and clinically relevant DXR doses remains to be established.
Collapse
|
29
|
Brickler M, Raskin A, Ryan TD. Current State of Pediatric Cardio-Oncology: A Review. CHILDREN (BASEL, SWITZERLAND) 2022; 9:127. [PMID: 35204848 PMCID: PMC8870613 DOI: 10.3390/children9020127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 01/03/2023]
Abstract
The landscape of pediatric oncology has dramatically changed over the course of the past several decades with five-year survival rates surpassing 80%. Anthracycline therapy has been the cornerstone of many chemotherapy regimens for pediatric patients since its introduction in the 1960s, and recent improved survival has been in large part due to advancements in chemotherapy, refinement of supportive care treatments, and development of novel therapeutics such as small molecule inhibitors, chimeric antigen receptor T-cell therapy, and immune checkpoint inhibitors. Unfortunately, many cancer-targeted therapies can lead to acute and chronic cardiovascular pathologies. The range of cardiotoxicity can vary but includes symptomatic or asymptotic heart failure, arrhythmias, coronary artery disease, valvar disease, pericardial disease, hypertension, and peripheral vascular disease. There is lack of data guiding primary prevention and treatment strategies in the pediatric population, which leads to substantial practice variability. Several important future research directions have been identified, including as they relate to cardiac disease, prevention strategies, management of cardiovascular risk factors, risk prediction, early detection, and the role of genetic susceptibility in development of cardiotoxicity. Continued collaborative research will be key in advancing the field. The ideal model for pediatric cardio-oncology is a proactive partnership between pediatric cardiologists and oncologists in order to better understand, treat, and ideally prevent cardiac disease in pediatric oncology patients.
Collapse
Affiliation(s)
| | | | - Thomas D. Ryan
- Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA;
| |
Collapse
|
30
|
Ogugofor MO, Njoku UO, Njoku OU, Batiha GES. Phytochemical analysis and thrombolytic profiling of Costus afer stem fractions. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-021-00392-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The most commonly occurring mechanism driving ischemic heart disease, ischemic stroke, and myocardial infarction is thrombosis. It is normally characterized by platelet activation and aggregation. Thrombolytics have been used in the treatment of several forms of thrombosis, but their adverse effects have limited their usefulness. Thus, there is a need to develop alternatives from medicinal plants known to possess antithrombotic activity such as Costus afer.
Results
The phytochemical evaluations indicated the presence of flavonoids, alkaloids, cardiac glycosides, tannins, terpenoids, and saponins. The antithrombotic profiling showed that streptokinase had the highest percentage clot lysis, followed by ethylacetate fraction of the extract, which was higher than aspirin and other fractions of the extract.
Conclusion
The present findings show that C. afer stem extract and various fractions possess antithrombotic activities. However, further studies are needed to characterize the antithrombotic bioactive compounds present in the different fractions that are responsible for the activities.
Collapse
|
31
|
Bansal N, Joshi C, Adams MJ, Hutchins K, Ray A, Lipshultz SE. Cardiotoxicity in pediatric lymphoma survivors. Expert Rev Cardiovasc Ther 2021; 19:957-974. [PMID: 34958622 DOI: 10.1080/14779072.2021.2013811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Over the past five decades, the diagnosis and management of children with various malignancies have improved tremendously. As a result, an increasing number of children are long-term cancer survivors. With improved survival, however, has come an increased risk of treatment-related cardiovascular complications that can appear decades later. AREAS COVERED This review discusses the pathophysiology, epidemiology and effects of treatment-related cardiovascular complications from anthracyclines and radiotherapy in pediatric lymphoma survivors. There is a paucity of evidence-based recommendations for screening for and treatment of cancer therapy-induced cardiovascular complications. We discuss current preventive measures and strategies for their treatment. EXPERT OPINION Significant cardiac adverse effects occur due to radiation and chemotherapy received by patients treated for lymphoma. Higher lifetime cumulative doses, female sex, longer follow-up, younger age, and preexisting cardiovascular disease are associated with a higher incidence of cardiotoxicity. With deeper understanding of the mechanisms of these adverse cardiac effects and identification of driver mutations causing these effects, personalized cancer therapy to limit cardiotoxic effects while ensuring an adequate anti-neoplastic effect would be ideal. In the meantime, expanding the use of cardioprotective agents with the best evidence such as dexrazoxane should be encouraged and further studied.
Collapse
Affiliation(s)
- Neha Bansal
- Division of Pediatric Cardiology, Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx NY, USA
| | - Chaitya Joshi
- Department of Pediatrics, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo NY, USA
| | - Michael Jacob Adams
- Department of Public Health Sciences, University of Rochester, Rochester NY, USA
| | - Kelley Hutchins
- John A. Burns School of Medicine, Pediatric Hematology/Oncology, Kapiolani Medical Center for Women and Children, Honolulu HI, USA
| | - Andrew Ray
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo NY, USA
| | - Steven E Lipshultz
- Department of Pediatrics, University at Buffalo Jacobs School of Medicine and Biomedical Sciences, Buffalo NY, USA.,Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo NY, USA.,Pediatrics Department, John R. Oishei Children's Hospital, UBMD Pediatrics Practice Group, Buffalo NY, USA
| |
Collapse
|
32
|
Sawicki KT, Sala V, Prever L, Hirsch E, Ardehali H, Ghigo A. Preventing and Treating Anthracycline Cardiotoxicity: New Insights. Annu Rev Pharmacol Toxicol 2021; 61:309-332. [PMID: 33022184 DOI: 10.1146/annurev-pharmtox-030620-104842] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Anthracyclines are the cornerstone of many chemotherapy regimens for a variety of cancers. Unfortunately, their use is limited by a cumulative dose-dependent cardiotoxicity. Despite more than five decades of research, the biological mechanisms underlying anthracycline cardiotoxicity are not completely understood. In this review, we discuss the incidence, risk factors, types, and pathophysiology of anthracycline cardiotoxicity, as well as methods to prevent and treat this condition. We also summarize and discuss advances made in the last decade in the comprehension of the molecular mechanisms underlying the pathology.
Collapse
Affiliation(s)
- Konrad Teodor Sawicki
- Division of Cardiology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Lorenzo Prever
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Hossein Ardehali
- Division of Cardiology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| |
Collapse
|
33
|
Radulescu LM, Radulescu D, Ciuleanu TE, Crisan D, Buzdugan E, Romitan DM, Buzoianu AD. Cardiotoxicity Associated with Chemotherapy Used in Gastrointestinal Tumours. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57080806. [PMID: 34441012 PMCID: PMC8400748 DOI: 10.3390/medicina57080806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/25/2021] [Accepted: 08/04/2021] [Indexed: 12/12/2022]
Abstract
Cardiotoxicity is a well-recognised side effect of cancer-related therapies with a great impact on outcomes and quality of life in the cancer survivor population. The pathogenesis of chemotherapy-induced cardiotoxicity in patients with gastrointestinal cancers involves various molecular mechanisms, and the combined use of various chemotherapies augments the risk of each drug used alone. In terms of cardiotoxicity diagnosis, novel biomarkers, such as troponins, brain natriuretic peptide (BNP), myeloperoxidases and miRNAs have been recently assessed. Echocardiography is a noninvasive imaging method of choice for the primary assessment of chemotherapy-treated patients to generally evaluate the cardiovascular impact of these drugs. Novel echocardiography techniques, like three-dimensional and stress echocardiography, will improve diagnosis efficacy. Cardiac magnetic resonance (CMR) can evaluate cardiac morphology, function and wall structure. Corroborated data have shown the importance of CMR in the early evaluation of patients with gastrointestinal cancers, treated with anticancer drugs, but further studies are required to improve risk stratification in these patients. In this article, we review some important aspects concerning the cardiotoxicity of antineoplastic drugs used in gastrointestinal cancers. We also discuss the mechanism of cardiotoxicity, the role of biomarkers and the imaging methods used in its detection.
Collapse
Affiliation(s)
- Liliana Maria Radulescu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania; (L.M.R.); (A.D.B.)
- Department of Cardiology, Cluj Municipal Hospital, 400005 Cluj-Napoca, Romania; (E.B.); (D.-M.R.)
| | - Dan Radulescu
- Department of Cardiology, Cluj Municipal Hospital, 400005 Cluj-Napoca, Romania; (E.B.); (D.-M.R.)
- Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania;
- Correspondence: ; Tel.: +40-741041707
| | - Tudor-Eliade Ciuleanu
- Department of Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania;
| | - Dana Crisan
- Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania;
- Department of Internal Medicine, Cluj Municipal Hospital, 400139 Cluj-Napoca, Romania
| | - Elena Buzdugan
- Department of Cardiology, Cluj Municipal Hospital, 400005 Cluj-Napoca, Romania; (E.B.); (D.-M.R.)
- Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania;
| | - Dragos-Mihai Romitan
- Department of Cardiology, Cluj Municipal Hospital, 400005 Cluj-Napoca, Romania; (E.B.); (D.-M.R.)
| | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania; (L.M.R.); (A.D.B.)
| |
Collapse
|
34
|
Pinto CM, Horta LS, Soares AP, Carvalho BA, Ferreira E, Lages EB, Ferreira LAM, Faraco AAG, Santiago HC, Goulart GAC. Nanoencapsulated Doxorubicin Prevents Mucositis Development in Mice. Pharmaceutics 2021; 13:1021. [PMID: 34371713 PMCID: PMC8329927 DOI: 10.3390/pharmaceutics13071021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 12/26/2022] Open
Abstract
Doxorubicin (DOX), a chemotherapy drug successfully used in the therapy of various types of cancer, is currently associated with the mucositis development, an inflammation that can cause ulcerative lesions in the mucosa of the gastrointestinal tract, abdominal pain and secondary infections. To increase the safety of the chemotherapy, we loaded DOX into nanostructured lipid carriers (NLCs). The NLC-DOX was characterized by HPLC, DLS, NTA, Zeta potential, FTIR, DSC, TEM and cryogenic-TEM. The ability of NLC-DOX to control the DOX release was evaluated through in vitro release studies. Moreover, the effect of NLC-DOX on intestinal mucosa was compared to a free DOX solution in C57BL/6 mice. The NLC-DOX showed spherical shape, high drug encapsulation efficiency (84.8 ± 4.6%), high drug loading (55.2 ± 3.4 mg/g) and low average diameter (66.0-78.8 nm). The DSC and FTIR analyses showed high interaction between the NLC components, resulting in controlled drug release. Treatment with NLC-DOX attenuated DOX-induced mucositis in mice, improving shortening on villus height and crypt depth, decreased inflammatory parameters, preserved intestinal permeability and increased expression of tight junctions (ZO-1 and Ocludin). These results indicated that encapsulation of DOX in NLCs is viable and reduces the drug toxicity to mucosal structures.
Collapse
Affiliation(s)
- Cristiane M. Pinto
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| | - Laila S. Horta
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.S.H.); (H.C.S.)
| | - Amanda P. Soares
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.S.H.); (H.C.S.)
| | - Bárbara A. Carvalho
- Department of General Pathology, Biological Science Institute, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (B.A.C.); (E.F.)
| | - Enio Ferreira
- Department of General Pathology, Biological Science Institute, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (B.A.C.); (E.F.)
| | - Eduardo B. Lages
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| | - Lucas A. M. Ferreira
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| | - André A. G. Faraco
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| | - Helton C. Santiago
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.S.H.); (H.C.S.)
| | - Gisele A. C. Goulart
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| |
Collapse
|
35
|
Noel CV, Rainusso N, Robertson M, Romero J, Masand P, Coarfa C, Pautler R. Early detection of myocardial changes with and without dexrazoxane using serial magnetic resonance imaging in a pre-clinical mouse model. CARDIO-ONCOLOGY 2021; 7:23. [PMID: 34134789 PMCID: PMC8207719 DOI: 10.1186/s40959-021-00109-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/28/2021] [Indexed: 12/01/2022]
Abstract
Background Cancer therapy-related cardiac dysfunction may occur in pediatric cancer survivors. Identification of early markers of myocardial damage secondary to anthracycline exposure is crucial to develop strategies that may ameliorate this complication. Objectives The purpose of this study was to identify early myocardial changes induced by doxorubicin with and without cardioprotection using dexrazoxane detected by serial cardiac magnetic resonance imaging (CMR) in a pre-clinical mouse model. Methods Serial CMR examinations were performed in 90 mice distributed in 3 groups: 45 received doxorubicin (DOX group), 30 mice received doxorubicin with dexrazoxane (DOX/DEX group) and 15 mice received saline injections (control group). We obtained the following CMR parameters in all mice: T2, extracellular volume quantification (ECV), myocardial deformation, and functional quantification. Results Myocardial edema assessed by T2 time was the earliest parameter demonstrating evidence of myocardial injury, most notable in the DOX group at week 4 and 8 compared with DOX/DEX group. Similarly, global longitudinal strain was abnormal in both the DOX and DOX/DEX groups. However, this change persisted only in the DOX group. The ECV was significantly elevated in the DOX group at the final CMR, while only minimally elevated in the DOX/DEX group. The right and left ejection fraction was decreased, along with the mass to volume ratio in the DOX group. The T2 time, ECV, and deformation correlated with ejection fraction and left ventricular volume. Conclusions T2 time and deformation by CMR identifies early myocardial injury from anthracyclines. Dexrazoxne did not prevent the initial edema, but the inflammatory changes were not sustained. CMR may be useful for early detection of cardiac dysfunction. Serial CMR demonstrates dexrazoxane minimizes cardiac dysfunction and aids recovery in a mouse model. Supplementary Information The online version contains supplementary material available at 10.1186/s40959-021-00109-8.
Collapse
Affiliation(s)
- Cory V Noel
- Pediatric Cardiology of Alaska, Seattle Children's Hospital - Division of Pediatric Cardiology, Anchorage, AK, USA.
| | - Nino Rainusso
- Division of Pediatric Hematology and Oncology, Baylor College of Medicine, Houston, USA.,Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, USA
| | - Matthew Robertson
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, USA
| | - Jonathan Romero
- Baylor College of Medicine Small Animal Imaging Facility, Houston, USA
| | - Prakash Masand
- Texas Children's Hospital Pediatric Radiology, Houston, USA
| | - Cristian Coarfa
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, USA
| | - Robia Pautler
- Baylor College of Medicine Small Animal Imaging Facility, Houston, USA
| |
Collapse
|
36
|
Tao RH, Kobayashi M, Yang Y, Kleinerman ES. Exercise Inhibits Doxorubicin-Induced Damage to Cardiac Vessels and Activation of Hippo/YAP-Mediated Apoptosis. Cancers (Basel) 2021; 13:cancers13112740. [PMID: 34205942 PMCID: PMC8198139 DOI: 10.3390/cancers13112740] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 01/16/2023] Open
Abstract
Dose-related cardiomyopathy is a major side effect following doxorubicin (Dox). To investigate whether exercise (Ex)-induced vasculogenesis plays a role in reducing Dox-induced cardiotoxicity, GFP+ bone marrow (BM) cells from GFP transgenic mice were transplanted into wild-type mice. Transplanted mice were treated with Dox, Ex, Dox+Ex, or control. We found Dox therapy resulted in decreased systolic and diastolic blood flow, decreased ejection fraction and fractional shortening, and decreased vascular endothelial cells and pericytes. These abnormalities were not seen in Dox+Ex hearts. Heart tissues from control-, Ex-, or Dox-treated mice showed a small number of GFP+ cells. By contrast, the Dox+Ex-treated hearts had a significant increase in GFP+ cells. Further analyses demonstrated these GFP+ BM cells had differentiated into vascular endothelial cells (GFP+CD31+) and pericytes (GFP+NG2+). Decreased cardiomyocytes were also seen in Dox-treated but not Dox+Ex-treated hearts. Ex induced an increase in GFP+c-Kit+ cells. However, these c-Kit+ BM stem cells had not differentiated into cardiomyocytes. Dox therapy induced phosphorylation of MST1/2, LATS1, and YAP; a decrease in total YAP; and cleavage of caspase-3 and PARP in the heart tissues. Dox+Ex prevented these effects. Our data demonstrated Dox-induced cardiotoxicity is mediated by vascular damage resulting in decreased cardiac blood flow and through activation of Hippo-YAP signaling resulting in cardiomyocyte apoptosis. Furthermore, Ex inhibited these effects by promoting migration of BM stem cells into the heart to repair the cardiac vessels damaged by Dox and through inhibiting Dox-induced Hippo-YAP signaling-mediated apoptosis. These data support the concept of using exercise as an intervention to decrease Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Rong-Hua Tao
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence: (R.-H.T.); (E.S.K.); Tel.: +1-(713)-563-7333 (R.-H.T.); +1-(713)-792-8110 (E.S.K.); Fax: +1-(713)-563-5407 (R.-H.T.); +1-(713)-563-5407 (E.S.K.)
| | - Masato Kobayashi
- School of Health Sciences, Institutes of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa 920-0942, Japan;
| | - Yuanzheng Yang
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
| | - Eugenie S. Kleinerman
- Department of Pediatrics-Research, Division of Pediatrics, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA;
- Correspondence: (R.-H.T.); (E.S.K.); Tel.: +1-(713)-563-7333 (R.-H.T.); +1-(713)-792-8110 (E.S.K.); Fax: +1-(713)-563-5407 (R.-H.T.); +1-(713)-563-5407 (E.S.K.)
| |
Collapse
|
37
|
Sofia R, Melita V, De Vita A, Ruggiero A, Romano A, Attinà G, Birritella L, Lamendola P, Lombardo A, Lanza GA, Delogu AB. Cardiac Surveillance for Early Detection of Late Subclinical Cardiac Dysfunction in Childhood Cancer Survivors After Anthracycline Therapy. Front Oncol 2021; 11:624057. [PMID: 34055601 PMCID: PMC8162652 DOI: 10.3389/fonc.2021.624057] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/20/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In childhood cancer survivors (CCSs) anthracycline-related cardiotoxicity is an important cause of morbidity and late mortality, but the optimal modality of cardiac surveillance still remains to be defined. The aim of this study was to assess whether non-invasive echocardiography-based functional cardiac measures can detect early subclinical myocardial changes in long-term pediatric cancer survivors who received anthracycline therapy. METHODS Twenty anthracycline-treated long-term CCSs and 20 age, sex, and body surface area matched healthy controls were enrolled in this study. Among cancer survivors, mean age at diagnosis was 6.5 ± 4.4 years, and the mean cumulative anthracycline dose was 234.5 ± 87.4 mg/m2. All subjects underwent a comprehensive functional echocardiographic protocol study including two-dimensional echocardiography (2D Echo), tissue Doppler imaging (TDI), speckle tracking (STE) and three-dimensional echocardiography (3D Echo). Patients were studied at a mean follow-up time of 6.5 ± 2.8 years from the end of therapy. RESULTS No significant differences in two-dimensional left ventricle ejection fraction (LVEF), diastolic parameters and speckle tracking (STE)-derived myocardial strain were observed between patients treated with anthracyclines and controls. Myocardial performance index was significantly prolonged (p = 0.005) and three-dimensional LVEF was significantly reduced (p = 0.002) in CCSs compared to controls, even though most values were within the normal range. There were no significant correlations between 2D, STE, and 3D echocardiographic parameters and age at diagnosis or duration of follow-up. No significant differences in echocardiographic parameters were found when stratifying cancer patients according to established risk factors for anthracycline cardiomyopathy. CONCLUSIONS This study found significantly reduced three-dimensional LVEF in CCSs compared with controls, despite no significant differences in two-dimensional LVEF and longitudinal strain values. These findings suggest that long-term CCSs who had received anthracycline therapy may be found to have subclinical features of myocardial dysfunction. However, further studies are needed to demonstrate the validity of new imaging techniques, including STE and 3D Echo, to identify patients at risk for cardiomyopathy in the long-term follow-up of CCSs.
Collapse
Affiliation(s)
- Rosaria Sofia
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of The Sacred Heart, Rome, Italy
| | - Veronica Melita
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio De Vita
- Catholic University of The Sacred Heart, Rome, Italy
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Ruggiero
- Catholic University of The Sacred Heart, Rome, Italy
- Pediatric Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Alberto Romano
- Catholic University of The Sacred Heart, Rome, Italy
- Pediatric Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giorgio Attinà
- Catholic University of The Sacred Heart, Rome, Italy
- Pediatric Oncology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Lisa Birritella
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of The Sacred Heart, Rome, Italy
| | - Priscilla Lamendola
- Catholic University of The Sacred Heart, Rome, Italy
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonella Lombardo
- Catholic University of The Sacred Heart, Rome, Italy
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gaetano Antonio Lanza
- Catholic University of The Sacred Heart, Rome, Italy
- Department of Cardiovascular and Thoracic Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Angelica Bibiana Delogu
- Unit of Pediatrics, Pediatric Cardiology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Catholic University of The Sacred Heart, Rome, Italy
| |
Collapse
|
38
|
Effect of traditional Chinese medicine on anthracycline-induced cardiotoxicity in animal models: A systematic review and meta-analysis. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2021. [DOI: 10.1016/j.jtcms.2021.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
39
|
Visscher H, Otth M, Feijen EAML, Nathan PC, Kuehni CE. Cardiovascular and Pulmonary Challenges After Treatment of Childhood Cancer. Pediatr Clin North Am 2020; 67:1155-1170. [PMID: 33131539 DOI: 10.1016/j.pcl.2020.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Childhood cancer survivors are at risk for developing cardiovascular disease and pulmonary disease related to cancer treatment. This might not become apparent until many years after treatment and varies from subclinical to life-threatening disease. Important causes are anthracyclines and radiotherapy involving heart, head, or neck for cardiovascular disease, and bleomycin, busulfan, nitrosoureas, radiation to the chest, and lung or chest surgery for pulmonary disease. Most effects are dose dependent, but genetic risk factors have been discovered. Treatment options are limited. Prevention and regular screening are crucial. Survivors should be encouraged to adopt a healthy lifestyle, and modifiable risk factors should be addressed.
Collapse
Affiliation(s)
- Henk Visscher
- Division of Haematology/Oncology, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada; Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, The Netherlands.
| | - Maria Otth
- Childhood Cancer Research Platform, Institute of Social and Preventive Medicine, University of Bern, Mittelstrasse 43, Bern 3012, Switzerland; Division of Hematology-Oncology, Department of Pediatrics, Kantonsspital Aarau, Switzerland
| | - E A M Lieke Feijen
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, The Netherlands
| | - Paul C Nathan
- AfterCare Program, Division of Haematology/Oncology, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada
| | - Claudia E Kuehni
- Childhood Cancer Research Platform, Institute of Social and Preventive Medicine, University of Bern, Mittelstrasse 43, Bern 3012, Switzerland; Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Mittelstrasse 43, Bern 3012, Switzerland
| |
Collapse
|
40
|
Sapkota Y. Harnessing Genomics to Predict and Prevent Anthracycline-Associated Cardiotoxicity. JACC CardioOncol 2020; 2:707-709. [PMID: 34396284 PMCID: PMC8352100 DOI: 10.1016/j.jaccao.2020.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
41
|
Lalmuansangi C, Zosangzuali M, Lalremruati M, Tochhawng L, Siama Z. Evaluation of the protective effects of Ganoderma applanatum against doxorubicin-induced toxicity in Dalton’s Lymphoma Ascites (DLA) bearing mice. Drug Chem Toxicol 2020; 45:1243-1253. [DOI: 10.1080/01480545.2020.1812630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- C. Lalmuansangi
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | - Mary Zosangzuali
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| | | | | | - Zothan Siama
- Department of Zoology, Mizoram University, Aizawl, Mizoram, India
| |
Collapse
|
42
|
Ma P, Qin Y, Cao H, Erben U, Ni C, Qin Z. Temporary blockade of interferon-γ ameliorates doxorubicin-induced cardiotoxicity without influencing the anti-tumor effect. Biomed Pharmacother 2020; 130:110587. [PMID: 32763819 DOI: 10.1016/j.biopha.2020.110587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 11/25/2022] Open
Abstract
Doxorubicin (DOX) is commonly used as an anti-cancer agent. However, its severe cardiotoxicity often makes it life threatening even long after DOX therapy during childhood. We recently reported interferon-γ (IFN-γ) necessary for DOX-induced acute cardiotoxicity in a p38 dependent way and, asked here for the potential of IFN-γ blockade to prevent DOX-induced chronic cardiotoxicity during tumor therapy. In our model system, mice without or with growing tumors repeatedly received DOX treatment. Simultaneous injection of anti-IFN-γ antibody R46-A2 with DOX to block IFN-γ signal efficiently protected the cardiac function of DOX treated recipients. Importantly, a single late injection of R46-A2 after DOX exposure also ameliorated DOX induced cardiac dysfunction in tumor-bearing mice. The anti-IFN-γ treatment did not affect the DOX-mediated tumor suppression effect and it left the main cellular immune response intact. Therefore, temporary blockade of IFN-γ may represent a novel strategy to ameliorate established DOX induced cardiotoxicity (DIC) or prevent its development in tumor therapy.
Collapse
Affiliation(s)
- Pan Ma
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yue Qin
- National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Hong Cao
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ulrike Erben
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Ni
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
43
|
Bellary A, Villarreal A, Eslami R, Undseth QJ, Lec B, Defnet AM, Bagrodia N, Kandel JJ, Borden MA, Shaikh S, Chopra R, Laetsch TW, Delaney LJ, Shaw CM, Eisenbrey JR, Hernandez SL, Sirsi SR. Perfusion-guided sonopermeation of neuroblastoma: a novel strategy for monitoring and predicting liposomal doxorubicin uptake in vivo. Theranostics 2020; 10:8143-8161. [PMID: 32724463 PMCID: PMC7381728 DOI: 10.7150/thno.45903] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/03/2020] [Indexed: 12/31/2022] Open
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in infants and children, and imposes significant morbidity and mortality in this population. The aggressive chemoradiotherapy required to treat high-risk NB results in survival of less than 50%, yet is associated with significant long-term adverse effects in survivors. Boosting efficacy and reducing morbidity are therefore key goals of treatment for affected children. We hypothesize that these may be achieved by developing strategies that both focus and limit toxic therapies to the region of the tumor. One such strategy is the use of targeted image-guided drug delivery (IGDD), which is growing in popularity in personalized therapy to simultaneously improve on-target drug deposition and assess drug pharmacodynamics in individual patients. IGDD strategies can utilize a variety of imaging modalities and methods of actively targeting pharmaceutical drugs, however in vivo imaging in combination with focused ultrasound is one of the most promising approaches already being deployed for clinical applications. Over the last two decades, IGDD using focused ultrasound with "microbubble" ultrasound contrast agents (UCAs) has been increasingly explored as a method of targeting a wide variety of diseases, including cancer. This technique, known as sonopermeation, mechanically augments vascular permeability, enabling increased penetration of drugs into target tissue. However, to date, methods of monitoring the vascular bioeffects of sonopermeation in vivo are lacking. UCAs are excellent vascular probes in contrast-enhanced ultrasound (CEUS) imaging, and are thus uniquely suited for monitoring the effects of sonopermeation in tumors. Methods: To monitor the therapeutic efficacy of sonopermeation in vivo, we developed a novel system using 2D and 3D quantitative contrast-enhanced ultrasound imaging (qCEUS). 3D tumor volume and contrast enhancement was used to evaluate changes in blood volume during sonopermeation. 2D qCEUS-derived time-intensity curves (TICs) were used to assess reperfusion rates following sonopermeation therapy. Intratumoral doxorubicin (and liposome) uptake in NB was evalauted ex vivo along with associated vascular changes. Results: In this study, we demonstrate that combining focused ultrasound therapy with UCAs can significantly enhance chemotherapeutic payload to NB in an orthotopic xenograft model, by improving delivery and tumoral uptake of long-circulating liposomal doxorubicin (L-DOX) nanoparticles. qCEUS imaging suggests that changes in flow rates are highly sensitive to sonopermeation and could be used to monitor the efficacy of treatment in vivo. Additionally, initial tumor perfusion may be a good predictor of drug uptake during sonopermeation. Following sonopermeation treatment, vascular biomarkers show increased permeability due to reduced pericyte coverage and rapid onset of doxorubicin-induced apoptosis of NB cells but without damage to blood vessels. Conclusion: Our results suggest that significant L-DOX uptake can occur by increasing tumor vascular permeability with microbubble sonopermeation without otherwise damaging the vasculature, as confirmed by in vivo qCEUS imaging and ex vivo analysis. The use of qCEUS imaging to monitor sonopermeation efficiency and predict drug uptake could potentially provide real-time feedback to clinicians for determining treatment efficacy in tumors, leading to better and more efficient personalized therapies. Finally, we demonstrate how the IGDD strategy outlined in this study could be implemented in human patients using a single case study.
Collapse
Affiliation(s)
- Aditi Bellary
- Department of Biomedical Engineering, University of Texas at Dallas, Richardson, TX, USA
| | - Arelly Villarreal
- Department of Biomedical Engineering, University of Texas at Dallas, Richardson, TX, USA
| | - Rojin Eslami
- Department of Biomedical Engineering, University of Texas at Dallas, Richardson, TX, USA
| | - Quincy J. Undseth
- Department of Biomedical Engineering, University of Texas at Dallas, Richardson, TX, USA
| | - Bianca Lec
- Department of Surgery, University of Chicago Medical School, Chicago, IL, USA
| | - Ann M. Defnet
- Department of Surgery, University of Chicago Medical School, Chicago, IL, USA
| | - Naina Bagrodia
- Department of Surgery, University of Chicago Medical School, Chicago, IL, USA
| | - Jessica J. Kandel
- Department of Surgery, University of Chicago Medical School, Chicago, IL, USA
| | - Mark A. Borden
- Biomedical Engineering, Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Sumbul Shaikh
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rajiv Chopra
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Theodore W. Laetsch
- Department of Pediatrics and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center and Children's Health, Dallas, TX, USA
| | - Lauren J. Delaney
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Colette M. Shaw
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - John R. Eisenbrey
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sonia L. Hernandez
- Department of Surgery, University of Chicago Medical School, Chicago, IL, USA
| | - Shashank R. Sirsi
- Department of Biomedical Engineering, University of Texas at Dallas, Richardson, TX, USA
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
44
|
Caru M, Curnier D. Sex and Gender Considerations After Surviving Acute Lymphoblastic Leukemia: An Exercise Oncology Context. J Adolesc Young Adult Oncol 2020; 9:441-444. [DOI: 10.1089/jayao.2019.0137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Maxime Caru
- Laboratory of Pathophysiology of Exercise (LPEX), School of Kinesiology and Physical Activity Sciences, Faculty of Medicine, University of Montreal, Montreal, Canada
- Sainte-Justine University Health Center, Research Center, Montreal, Canada
- Laboratoire EA 4430–Clinique Psychanalyse Developpement (CliPsyD), University of Paris Nanterre, Nanterre, France
| | - Daniel Curnier
- Laboratory of Pathophysiology of Exercise (LPEX), School of Kinesiology and Physical Activity Sciences, Faculty of Medicine, University of Montreal, Montreal, Canada
- Sainte-Justine University Health Center, Research Center, Montreal, Canada
| |
Collapse
|
45
|
Pinheiro EA, Magdy T, Burridge PW. Human In Vitro Models for Assessing the Genomic Basis of Chemotherapy-Induced Cardiovascular Toxicity. J Cardiovasc Transl Res 2020; 13:377-389. [PMID: 32078739 PMCID: PMC7365753 DOI: 10.1007/s12265-020-09962-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/22/2020] [Indexed: 12/20/2022]
Abstract
Chemotherapy-induced cardiovascular toxicity (CICT) is a well-established risk for cancer survivors and causes diseases such as heart failure, arrhythmia, vascular dysfunction, and atherosclerosis. As our knowledge of the precise cardiovascular risks of each chemotherapy agent has improved, it has become clear that genomics is one of the most influential predictors of which patients will experience cardiovascular toxicity. Most recently, GWAS-led, top-down approaches have identified novel genetic variants and their related genes that are statistically related to CICT. Importantly, the advent of human-induced pluripotent stem cell (hiPSC) models provides a system to experimentally test the effect of these genomic findings in vitro, query the underlying mechanisms, and develop novel strategies to mitigate the cardiovascular toxicity liabilities due to these mechanisms. Here we review the cardiovascular toxicities of chemotherapy drugs, discuss how these can be modeled in vitro, and suggest how these models can be used to validate genetic variants that predispose patients to these effects.
Collapse
Affiliation(s)
- Emily A Pinheiro
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
46
|
Giaccone L, Felicetti F, Butera S, Faraci D, Cerrano M, Dionisi Vici M, Brunello L, Fortunati N, Brignardello E, Bruno B. Optimal Delivery of Follow-Up Care After Allogeneic Hematopoietic Stem-Cell Transplant: Improving Patient Outcomes with a Multidisciplinary Approach. J Blood Med 2020; 11:141-162. [PMID: 32523389 PMCID: PMC7237112 DOI: 10.2147/jbm.s206027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/02/2020] [Indexed: 01/05/2023] Open
Abstract
The increasing indications for allogeneic stem-cell transplant in patients with hematologic malignancies and non-malignant diseases combined with improved clinical outcomes have contributed to increase the number of long-term survivors. However, survivors are at increased risk of developing a unique set of complications and late effects, besides graft-versus-host disease and disease relapse. In this setting, the management capacity of a single health-care provider can easily be overwhelmed. Thus, to provide appropriate survivorship care, a multidisciplinary approach for the long-term follow-up is essential. This review aims at summarizing the most relevant information that a health-care provider should know to establish a follow-up care plan, in the light of individual exposures and risk factors, that includes all organ systems and considers the psychological burden of these patients.
Collapse
Affiliation(s)
- Luisa Giaccone
- Division of Hematology, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, University of Torino, Torino, Italy
| | - Francesco Felicetti
- Transition Unit for Childhood Cancer Survivors, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Sara Butera
- Division of Hematology, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, University of Torino, Torino, Italy
| | - Danilo Faraci
- Division of Hematology, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, University of Torino, Torino, Italy
| | - Marco Cerrano
- Division of Hematology, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, University of Torino, Torino, Italy
| | - Margherita Dionisi Vici
- Transition Unit for Childhood Cancer Survivors, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Lucia Brunello
- Division of Hematology, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, University of Torino, Torino, Italy
| | - Nicoletta Fortunati
- Transition Unit for Childhood Cancer Survivors, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Enrico Brignardello
- Transition Unit for Childhood Cancer Survivors, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy
| | - Benedetto Bruno
- Division of Hematology, Azienda Ospedaliera Universitaria Città della Salute e della Scienza di Torino, University of Torino, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, University of Torino, Torino, Italy
| |
Collapse
|
47
|
Zhang Y, Ma C, Liu C, Wei F. Luteolin attenuates doxorubicin-induced cardiotoxicity by modulating the PHLPP1/AKT/Bcl-2 signalling pathway. PeerJ 2020; 8:e8845. [PMID: 32435528 PMCID: PMC7224230 DOI: 10.7717/peerj.8845] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Background Luteolin (LUT) is a flavonoid found in vegetables and fruits that has diverse functions. Doxorubicin (DOX) is an anthracycline antibiotic that is frequently used for the treatment of various cancers. Unfortunately, the clinical efficacy of DOX is limited by its dose-related cardiotoxicity. In this study, we aimed to investigate the potential mechanism through which LUT attenuates cardiotoxicity in vivo. Methods We evaluated the body weight, heart weight, electrocardiogram, and pathological changes before and after administration of LUT. Moreover, the effects of LUT (50 mg/kg in the low dose group, 100 mg/kg in the high dose group) on biochemical parameters (brain natriuretic peptide, creatine kinase MB, cardiac troponin T, and dehydrogenation of lactate enzyme) and oxidative stress parameters (malondialdehyde and superoxide dismutase) were studied in the sera of cardiotoxicity model rats. We also identified the apoptotic mediators whose expression was induced by LUT by quantitative real-time reverse transcription-polymerase chain reaction (RT-qPCR) evaluation. In addition, we used network analysis to predict DOX-induced cardiotoxicity and protection afforded by LUT. Western blotting was used to detect the expression of associated proteins. Results LUT significantly improved DOX-induced cardiotoxicity in a dose-dependent fashion. LUT ameliorated DOX-induced weight loss and heart weight changes, as well as changes in biochemical parameters and oxidative stress parameters in heart injury model rats. LUT’s protective effect was observed via regulation of the apoptotic markers Bcl-2, Bax, and caspase-3 mRNA and protein expression levels. Network analysis showed that the AKT/Bcl-2 signalling pathway was activated; specifically, the PH domain leucine-rich repeats protein phosphatase 1 (phlpp1) was involved in the AKT/Bcl-2 signal pathway. LUT inhibited the activity of phlpp1 leading to positive regulation of the AKT/Bcl-2 pathway, which attenuated doxorubicin-induced cardiotoxicity. Conclusions These results demonstrate that LUT exerted protective effects against DOX-induced cardiotoxicity in vivo by alleviating oxidative stress, suppressing phlpp1 activity, and activating the AKT/Bcl-2 signalling pathway.
Collapse
Affiliation(s)
- YanDong Zhang
- Department of Rheumatology, First Hospital, Jilin University, ChangChun, Jilin, China
| | - ChengYuan Ma
- Department of Neurosurgery, First Hospital, Jilin University, ChangChun, Jilin, China
| | - ChunShui Liu
- Department of Hematology , First Hospital, Jilin University, ChangChun, Jilin, China
| | - Feng Wei
- Department of Hepatobiliary & Pancreas Surgery, First Hospital, Jilin University, Changchun, Jilin, China
| |
Collapse
|
48
|
Dhingra R, Guberman M, Rabinovich-Nikitin I, Gerstein J, Margulets V, Gang H, Madden N, Thliveris J, Kirshenbaum LA. Impaired NF-κB signalling underlies cyclophilin D-mediated mitochondrial permeability transition pore opening in doxorubicin cardiomyopathy. Cardiovasc Res 2020; 116:1161-1174. [PMID: 31566215 PMCID: PMC7177490 DOI: 10.1093/cvr/cvz240] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/15/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022] Open
Abstract
AIMS The chemotherapy drug doxorubicin (Dox) is commonly used for treating a variety of human cancers; however, it is highly cardiotoxic and induces heart failure. We previously reported that the Bcl-2 mitochondrial death protein Bcl-2/19kDa interaction protein 3 (Bnip3), is critical for provoking mitochondrial perturbations and necrotic cell death in response to Dox; however, the underlying mechanisms had not been elucidated. Herein, we investigated mechanism that drives Bnip3 gene activation and downstream effectors of Bnip3-mediated mitochondrial perturbations and cell death in cardiac myocytes treated with Dox. METHODS AND RESULTS Nuclear factor-κB (NF-κB) signalling, which transcriptionally silences Bnip3 activation under basal states in cardiac myocytes was dramatically reduced following Dox treatment. This was accompanied by Bnip3 gene activation, mitochondrial injury including calcium influx, permeability transition pore (mPTP) opening, loss of nuclear high mobility group protein 1, reactive oxygen species production, and cell death. Interestingly, impaired NF-κB signalling in cells treated with Dox was accompanied by protein complexes between Bnip3 and cyclophilin D (CypD). Notably, Bnip3-mediated mPTP opening was suppressed by inhibition of CypD-demonstrating that CypD functionally operates downstream of Bnip3. Moreover, restoring IKKβ-NF-κB activity in cardiac myocytes treated with Dox suppressed Bnip3 expression, mitochondrial perturbations, and necrotic cell death. CONCLUSIONS The findings of the present study reveal a novel signalling pathway that functionally couples NF-κB and Dox cardiomyopathy to a mechanism that is mutually dependent upon and obligatorily linked to the transcriptional control of Bnip3. Our findings further demonstrate that mitochondrial injury and necrotic cell death induced by Bnip3 is contingent upon CypD. Hence, maintaining NF-κB signalling may prove beneficial in reducing mitochondrial dysfunction and heart failure in cancer patients undergoing Dox chemotherapy.
Collapse
Affiliation(s)
- Rimpy Dhingra
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Manitoba, Canada
| | - Matthew Guberman
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Manitoba, Canada
| | - Inna Rabinovich-Nikitin
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Manitoba, Canada
| | - Jonathon Gerstein
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Manitoba, Canada
| | - Victoria Margulets
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Manitoba, Canada
| | - Hongying Gang
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Manitoba, Canada
| | - Nicholas Madden
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Manitoba, Canada
| | - James Thliveris
- Department of Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Lorrie A Kirshenbaum
- Department of Physiology and Pathophysiology, The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Manitoba, Canada
| |
Collapse
|
49
|
Getz KD, Sung L, Alonzo TA, Leger KJ, Gerbing RB, Pollard JA, Cooper T, Kolb EA, Gamis AS, Ky B, Aplenc R. Effect of Dexrazoxane on Left Ventricular Systolic Function and Treatment Outcomes in Patients With Acute Myeloid Leukemia: A Report From the Children's Oncology Group. J Clin Oncol 2020; 38:2398-2406. [PMID: 32343641 DOI: 10.1200/jco.19.02856] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To determine whether dexrazoxane provides effective cardioprotection during frontline treatment of pediatric acute myeloid leukemia (AML) without increasing relapse risk or noncardiac toxicities of the chemotherapy regimens. PATIENTS AND METHODS This was a multicenter study of all pediatric patients with AML without high allelic ratio FLT3/ITD treated in the Children's Oncology Group trial AAML1031 between 2011 and 2016. Median follow-up was 3.5 years. Dexrazoxane was administered at the discretion of treating physicians and documented at each course. Ejection fraction (EF) and shortening fraction (SF) were recorded after each course and at regular intervals in follow-up. Per protocol, anthracyclines were to be withheld if there was evidence of left ventricular systolic dysfunction (LVSD) defined as SF < 28% or EF < 55%. Occurrence of LVSD, trends in EF and SF, 5-year event-free survival (EFS) and overall survival (OS), and treatment-related mortality (TRM) were compared by dexrazoxane exposure. RESULTS A total of 1,014 patients were included in the analyses; 96 were exposed to dexrazoxane at every anthracycline course, and 918 were never exposed. Distributions of sex, age, race, presenting WBC count, risk group, treatment arm, and compliance with cardiac monitoring were similar for dexrazoxane-exposed and -unexposed patients. Dexrazoxane-exposed patients had significantly smaller EF and SF declines than unexposed patients across courses and a lower risk for LVSD (26.5% v 42.2%; hazard ratio, 0.55; 95% CI, 0.36 to 0.86; P = .009). Dexrazoxane-exposed patients had similar 5-year EFS (49.0% v 45.1%; P = .534) and OS (65.0% v 61.9%; P = .613) to those unexposed; however, there was a suggestion of lower TRM with dexrazoxane (5.7% v 12.7%; P = .068). CONCLUSION Dexrazoxane preserved cardiac function without compromising EFS and OS or increasing noncardiac toxicities. Dexrazoxane should be considered for cardioprotection during frontline treatment of pediatric AML.
Collapse
Affiliation(s)
- Kelly D Getz
- Children's Hospital of Philadelphia, Philadelphia, PA.,University of Pennsylvania, Philadelphia, PA
| | - Lillian Sung
- The Hospital for Sick Children, Toronto, Ontario, Canada
| | | | | | | | - Jessica A Pollard
- Boston Children's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | | | - E Anders Kolb
- Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
| | - Alan S Gamis
- Children's Mercy Hospital and Clinics, Kansas City, MO
| | - Bonnie Ky
- University of Pennsylvania, Philadelphia, PA
| | - Richard Aplenc
- Children's Hospital of Philadelphia, Philadelphia, PA.,University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
50
|
Zhang D, Lv J, Zhang B, Zhang X, Jiang H, Lin Z. The characteristics and regularities of cardiac adverse drug reactions induced by Chinese materia medica: A bibliometric research and association rules analysis. JOURNAL OF ETHNOPHARMACOLOGY 2020; 252:112582. [PMID: 31972324 DOI: 10.1016/j.jep.2020.112582] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/06/2020] [Accepted: 01/15/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chinese materia medica (CMM) has been widely used as an approach of ethnomedicine worldwide. Recently, there are growing concerns related to the potential cardiotoxicity of herbal medicines but comprehensive studies are limited. METHODS Comprehensive literature retrieval via publicly available electronic databases was performed to identify the case reports that focused on cardiac adverse reactions (ADRs) triggered by oral CMMs. And a bibliometric survey was conducted to analyze the most commonly suspected risk factors in terms of responsible CMMs, susceptible patients and clinical administration of cardiac ADRs. Moreover, the techniques of data mining were utilized to investigate the regularities and association between the ADRs status and major contributory factors. RESULTS The available evidence of current research indicated that many influential factors were strongly associated with cardiac ADRs caused by oral CMMs inevitably, including pediatric patients, poisonous CMMs (especially herbs of Aconitum species), overdose and self-medication. Specifically, the timely and effective resuscitation could attribute their favorable capacity to reduce mortality for cardiac ADRs. Notably, the cardiac ADRs cases had often concomitant the ADRs of the nervous system and digestive system. CONCLUSION The comprehensive features and risk factors of cardiac ADRs induced by oral CMMs can be discovered and elucidated through the approaches of bibliometric research, association rules analysis, and data mining technology, which raise the profile and awareness of the rational applications of CMMs and pharmacovigilance within relevant heart side effects.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing, 100029, China.
| | - Jintao Lv
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing, 100029, China.
| | - Bing Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing, 100029, China; Center for Pharmacovigilance and Rational Use of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Xiaomeng Zhang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing, 100029, China.
| | - Hao Jiang
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing, 100029, China.
| | - Zhijian Lin
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, No. 11 North Three-ring East Road, Chao Yang District, Beijing, 100029, China.
| |
Collapse
|