1
|
Hu PJ, Tsai PC, Chen CY, Kuo HT, Hung CH, Tseng KC, Lai HC, Peng CY, Wang JH, Chen JJ, Lee PL, Chien RN, Yang CC, Lo GH, Kao JH, Liu CJ, Liu CH, Yan SL, Lin CY, Su WW, Chu CH, Chen CJ, Tung SY, Tai CM, Lin CW, Lo CC, Cheng PN, Chiu YC, Wang CC, Cheng JS, Tsai WL, Lin HC, Huang YH, Yeh ML, Huang CF, Hsieh MH, Huang JF, Dai CY, Chung WL, Wang YH, Yu ML, Bair MJ, T‐COACH Study Group. Impact of Hepatitis C Virus Clearance on Cardiovascular Risk: A Real-World Experience From the Nationwide Taiwan Hepatitis C Virus Registry. Kaohsiung J Med Sci 2025:e70036. [PMID: 40411291 DOI: 10.1002/kjm2.70036] [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: 02/13/2025] [Revised: 04/04/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Hepatitis C virus (HCV) infection is associated with an increased risk of cardiovascular disease (CVD); however, the impact of interferon (IFN)-based therapy on cardiovascular outcomes remains unclear. This nationwide cohort study included 7411 patients with HCV from The Taiwanese Chronic Hepatitis C Cohort registry who received IFN-based therapy between 2003 and 2014. Patients were categorized into sustained virological response (SVR) (n = 5785) and non-SVR (n = 1676) groups. The incidence of new-onset CVD events, including stroke, coronary artery disease, heart failure, and arrhythmia, was assessed using three Cox proportional hazard models adjusted for different sets of confounding factors. The cumulative CVD incidence was comparable in the SVR and non-SVR groups (11.2% vs. 10.2%, p = 0.609). SVR was not significantly associated with a reduced overall CVD risk among the three models [hazards ratio (HR) = 0.88, 95% confidence interval (CI): 0.71-1.05, p = 0.158]. However, a lower risk of stroke was observed in patients who achieved an SVR, although the difference was not significant (HR = 0.84, 95% CI: 0.74-0.94). The results of the sensitivity analyses confirmed these findings. An SVR following IFN-based therapy did not substantially reduce the overall CVD risk; however, a potential reduction in stroke risk was observed. These results emphasize the importance of long-term cardiovascular risk assessments and highlight the need for further research, particularly in the direct-acting antiviral era in which increased cardiovascular benefits may be expected.
Collapse
Affiliation(s)
- Ping-Jen Hu
- Department of Internal Medicine, Division of Gastroenterology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Pei-Chien Tsai
- Hepatobiliary Section, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital; Hepatitis Research Center, School of Medicine and Cohort Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chi-Yi Chen
- Department of Internal Medicine, Chiayi Christian Hospital, Chiayi, Taiwan
| | - Hsing-Tao Kuo
- Division of Hepatogastroenterology, Department of Internal Medicine, Chi-Mei Medical Center, Tainan, Taiwan
| | - Chao-Hung Hung
- Division of Hepatogastroenterology, Department of Internal Medicine, Chiayi Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Kuo-Chih Tseng
- Department of Gastroenterology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Hsueh-Chou Lai
- Division of Hepatogastroenterology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Cheng-Yuan Peng
- Division of Hepatogastroenterology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Houng Wang
- Division of Hepatogastroenterology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jyh-Jou Chen
- Division of Gastroenterology and Hepatology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Pei-Lun Lee
- Division of Gastroenterology and Hepatology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Rong-Nan Chien
- Division of Hepatology, Department of Gastroenterology and Hepatology, Linkou Medical Center, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chi-Chieh Yang
- Division of Gastroenterology, Department of Internal Medicine, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Gin-Ho Lo
- Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Jia-Horng Kao
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Jen Liu
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chen-Hua Liu
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, National Taiwan University Hospital, Taipei, Taiwan
| | - Sheng-Lei Yan
- Division of Gastroenterology, Department of Internal Medicine, Chang Bing Show-Chwan Memorial Hospital, Changhua, Taiwan
| | - Chun-Yen Lin
- Division of Hepatology, Department of Gastroenterology and Hepatology, Linkou Medical Center, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Wei-Wen Su
- Division of Gastroenterology, Department of Internal Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Cheng-Hsin Chu
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chih-Jen Chen
- Division of Gastroenterology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan
| | - Shui-Yi Tung
- Division of Hepatogastroenterology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chi-Ming Tai
- Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chih-Wen Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Ching-Chu Lo
- Department of Internal Medicine, St. Martin De Porres Hospital-Daya, Chiayi, Taiwan
| | - Pin-Nan Cheng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yen-Cheng Chiu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chia-Chi Wang
- Division of Gastroenterology, Department of Internal Medicine, Taipei Tzuchi Hospital, the Buddhist Tzuchi Medical Foundation, New Taipei, Taiwan
| | - Jin-Shiung Cheng
- Division of Gastroenterology and Hepatology, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Lun Tsai
- Division of Gastroenterology and Hepatology, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Han-Chieh Lin
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei, Taiwan
| | - Yi-Hsiang Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Ming-Lun Yeh
- Hepatobiliary Section, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital; Hepatitis Research Center, School of Medicine and Cohort Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Feng Huang
- Hepatobiliary Section, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital; Hepatitis Research Center, School of Medicine and Cohort Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Meng-Hsuan Hsieh
- Hepatobiliary Section, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital; Hepatitis Research Center, School of Medicine and Cohort Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Health Management Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Jee-Fu Huang
- Hepatobiliary Section, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital; Hepatitis Research Center, School of Medicine and Cohort Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chia-Yen Dai
- Hepatobiliary Section, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital; Hepatitis Research Center, School of Medicine and Cohort Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Long Chung
- Hepatobiliary Section, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital; Hepatitis Research Center, School of Medicine and Cohort Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yen-Hsiange Wang
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Lung Yu
- Hepatobiliary Section, Department of Internal Medicine, and Hepatitis Center, Kaohsiung Medical University Hospital; Hepatitis Research Center, School of Medicine and Cohort Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Jong Bair
- Division of Gastroenterology, Department of Internal Medicine, Taitung Mackay Memorial Hospital, Taipei, Taiwan
| | | |
Collapse
|
2
|
Goel RR, Rook AH. Immunobiology and treatment of cutaneous T-cell lymphoma. Expert Rev Clin Immunol 2024; 20:985-996. [PMID: 38450476 DOI: 10.1080/1744666x.2024.2326035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
INTRODUCTION Primary cutaneous T cell lymphomas (CTCL) are a heterogenous group of non-Hodgkin lymphomas derived from skin-homing T cells. These include mycosis fungoides and its leukemic variant Sezary syndrome, as well as the CD30+ lymphoproliferative disorders. AREAS COVERED In this review, we provide a summary of the current literature on CTCL, with a focus on the immunopathogenesis and treatment of mycosis fungoides and Sezary syndrome. EXPERT OPINION Recent advances in immunology have provided new insights into the biology of malignant T cells. This in turn has led to the development of new therapies that modulate the immune system to facilitate tumor clearance or target specific aspects of tumor biology.
Collapse
Affiliation(s)
- Rishi R Goel
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology and Immune Health (I3H), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alain H Rook
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
3
|
Romero E, Tabak E, Fishbein G, Litovsky S, Tallaj J, Liem D, Bakir M, Khachatoorian Y, Piening B, Keating B, Deng M, Cadeiras M. Unsupervised mRNA-seq classification of heart transplant endomyocardial biopsies. Clin Transplant 2023; 37:e15011. [PMID: 37151104 PMCID: PMC10524567 DOI: 10.1111/ctr.15011] [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: 01/21/2023] [Revised: 04/10/2023] [Accepted: 04/28/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND Endomyocardial biopsy (EMB) is currently considered the gold standard for diagnosing cardiac allograft rejection. However, significant limitations related to histological interpretation variability are well-recognized. We sought to develop a methodology to evaluate EMB solely based on gene expression, without relying on histology interpretation. METHODS Sixty-four EMBs were obtained from 47 post-heart transplant recipients, who were evaluated for allograft rejection. EMBs were subjected to mRNA sequencing, in which an unsupervised classification algorithm was used to identify the molecular signatures that best classified the EMBs. Cytokine and natriuretic peptide peripheral blood profiling was also performed. Subsequently, we performed gene network analysis to identify the gene modules and gene ontology to understand their biological relevance. We correlated our findings with the unsupervised and histological classifications. RESULTS Our algorithm classifies EMBs into three categories based solely on clusters of gene expression: unsupervised classes 1, 2, and 3. Unsupervised and histological classifications were closely related, with stronger gene module-phenotype correlations for the unsupervised classes. Gene ontology enrichment analysis revealed processes impacting on the regulation of cardiac and mitochondrial function, immune response, and tissue injury response. Significant levels of cytokines and natriuretic peptides were detected following the unsupervised classification. CONCLUSION We have developed an unsupervised algorithm that classifies EMBs into three distinct categories, without relying on histology interpretation. These categories were highly correlated with mitochondrial, immune, and tissue injury response. Significant cytokine and natriuretic peptide levels were detected within the unsupervised classification. If further validated, the unsupervised classification could offer a more objective EMB evaluation.
Collapse
Affiliation(s)
- Erick Romero
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, CA
| | - Esteban Tabak
- Courant Institute of Mathematical Sciences, New York University, New York, NY
| | - Gregory Fishbein
- David Geffen School of Medicine, UCLA Medical Center, Los Angeles, CA
| | - Silvio Litovsky
- UAB School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Jose Tallaj
- UAB School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - David Liem
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, CA
| | - Maral Bakir
- David Geffen School of Medicine, UCLA Medical Center, Los Angeles, CA
| | | | - Brian Piening
- Earle A. Chiles Research Institute, Providence, Portland, Oregon
| | - Brendan Keating
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, PA
| | - Mario Deng
- David Geffen School of Medicine, UCLA Medical Center, Los Angeles, CA
| | - Martin Cadeiras
- Division of Cardiovascular Medicine, UC Davis Medical Center, Sacramento, CA
| |
Collapse
|
4
|
Jayakodi S, Senthilnathan R, Swaminathan A, Shanmugam VK, Shanmugam RK, Krishnan A, Ponnusamy VK, Tsai PC, Lin YC, Chen YH. Bio-inspired nanoparticles mediated from plant extract biomolecules and their therapeutic application in cardiovascular diseases: A review. Int J Biol Macromol 2023:125025. [PMID: 37245774 DOI: 10.1016/j.ijbiomac.2023.125025] [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: 03/06/2023] [Revised: 05/05/2023] [Accepted: 05/20/2023] [Indexed: 05/30/2023]
Abstract
Nanoparticles (NPs) have gained recognition for diagnosis, drug delivery, and therapy in fatal diseases. This review focuses on the benefits of green synthesis of bioinspired NPs using various plant extract (containing various biomolecules such as sugars, proteins, and other phytochemical compounds) and their therapeutic application in cardiovascular diseases (CVDs). Multiple factors including inflammation, mitochondrial and cardiomyocyte mutations, endothelial cell apoptosis, and administration of non-cardiac drugs, can trigger the cause of cardiac disorders. Furthermore, the interruption of reactive oxygen species (ROS) synchronization from mitochondria causes oxidative stress in the cardiac system, leading to chronic diseases such as atherosclerosis and myocardial infarction. NPs can decrease the interaction with biomolecules and prevent the incitement of ROS. Understanding this mechanism can pave the way for using green synthesized elemental NPs to reduce the risk of CVD. This review delivers information on the different methods, classifications, mechanisms and benefits of using NPs, as well as the formation and progression of CVDs and their effects on the body.
Collapse
Affiliation(s)
- Santhoshkumar Jayakodi
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India
| | - Raghul Senthilnathan
- Global Business School for Health, University College London, Gower St, London WC1E 6BT, United Kingdom
| | - Akila Swaminathan
- Clinical Virology, Manipal Institute of Virology, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Venkat Kumar Shanmugam
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Rajesh Kumar Shanmugam
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Chennai, Tamil Nadu 600077, India
| | - Anbarasu Krishnan
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India.
| | - Vinoth Kumar Ponnusamy
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City 807, Taiwan; Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung City 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital (KMUH), Kaohsiung City 807, Taiwan; Center for Emerging Contaminants Research, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| | - Pei-Chien Tsai
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University (KMU), Kaohsiung City 807, Taiwan
| | - Yuan-Chung Lin
- Center for Emerging Contaminants Research, National Sun Yat-Sen University, Kaohsiung 804, Taiwan; Institute of Environmental Engineering, National Sun Yat-Sen University, Kaohsiung 804, Taiwan
| | - Yi-Hsun Chen
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung City, Taiwan.
| |
Collapse
|
5
|
Akkaif MA, Sha’aban A, Cesaro A, Jaber AAS, Vergara A, Yunusa I, Jatau AI, Mohammed M, Govindasamy GS, Al-Mansoub MA, Sheikh Abdul Kader MA, Ibrahim B. The impact of SARS-CoV-2 treatment on the cardiovascular system: an updated review. Inflammopharmacology 2022; 30:1143-1151. [PMID: 35701719 PMCID: PMC9196858 DOI: 10.1007/s10787-022-01009-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/12/2022] [Indexed: 02/08/2023]
Abstract
The coronavirus disease-2019 (COVID-19) pandemic has become a major global health problem. COVID-19 is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and exhibits pulmonary and extrapulmonary effects, including cardiovascular involvement. There are several attempts to identify drugs that could treat COVID-19. Moreover, many patients infected with COVID-19 have underlying diseases, particularly cardiovascular diseases. These patients are more likely to develop severe illnesses and would require optimized treatment strategies. The current study gathered information from various databases, including relevant studies, reviews, trials, or meta-analyses until April 2022 to identify the impact of SARS-CoV-2 treatment on the cardiovascular system. Studies have shown that the prognosis of patients with underlying cardiovascular disease is worsened by COVID-19, with some COVID-19 medications interfering with the cardiovascular system. The COVID-19 treatment strategy should consider many factors and parameters to avoid medication-induced cardiac injury, mainly in elderly patients. Therefore, this article provides a synthesis of evidence on the impact of different COVID-19 medications on the cardiovascular system and related disease conditions.
Collapse
Affiliation(s)
- Mohammed Ahmed Akkaif
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Abubakar Sha’aban
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Arturo Cesaro
- Department of Cardio-Thoracic and Respiratory Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Ammar Ali Saleh Jaber
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, 19099 Dubai, United Arab Emirates
| | - Andrea Vergara
- Department of Cardio-Thoracic and Respiratory Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Ismaeel Yunusa
- College of Pharmacy, University of South Carolina, Columbia, United States
| | - Abubakar Ibrahim Jatau
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Tasmania, Australia
| | - Mustapha Mohammed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 Penang, Malaysia
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmaceutical Sciences, Ahmadu Bello University, Zaria, Kaduna, Nigeria
| | | | | | | | - Baharudin Ibrahim
- Faculty of Pharmacy, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Iwanski J, Kazmouz SG, Li S, Stansfield B, Salem TT, Perez-Miller S, Kazui T, Jena L, Uhrlaub JL, Lick S, Nikolich-Žugich J, Konhilas JP, Gregorio CC, Khanna M, Campos SK, Churko JM. Antihypertensive drug treatment and susceptibility to SARS-CoV-2 infection in human PSC-derived cardiomyocytes and primary endothelial cells. Stem Cell Reports 2021; 16:2459-2472. [PMID: 34525378 PMCID: PMC8407952 DOI: 10.1016/j.stemcr.2021.08.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 01/08/2023] Open
Abstract
The pathogenicity of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been attributed to its ability to enter through the membrane-bound angiotensin-converting enzyme 2 (ACE2) receptor. Therefore, it has been heavily speculated that angiotensin-converting enzyme inhibitor (ACEI) or angiotensin receptor blocker (ARB) therapy may modulate SARS-CoV-2 infection. In this study, exposure of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) and human endothelial cells (hECs) to SARS-CoV-2 identified significant differences in protein coding genes involved in immunity, viral response, and cardiomyocyte/endothelial structure. Specifically, transcriptome changes were identified in the tumor necrosis factor (TNF), interferon α/β, and mitogen-activated protein kinase (MAPK) (hPSC-CMs) as well as nuclear factor kappa-B (NF-κB) (hECs) signaling pathways. However, pre-treatment of hPSC-CMs or hECs with two widely prescribed antihypertensive medications, losartan and lisinopril, did not affect the susceptibility of either cell type to SARS-CoV-2 infection. These findings demonstrate the toxic effects of SARS-CoV-2 in hPSC-CMs/hECs and, taken together with newly emerging multicenter trials, suggest that antihypertensive drug treatment alone does not alter SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jessika Iwanski
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Sobhi G Kazmouz
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Shuaizhi Li
- Department of Immunobiology and the University of Arizona Center on Aging, The University of Arizona, Tucson, AZ, USA
| | - Ben Stansfield
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA; Department of Physiology, The University of Arizona, Tucson, AZ, USA
| | - Tori T Salem
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA; Department of Physiology, The University of Arizona, Tucson, AZ, USA
| | - Samantha Perez-Miller
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Toshinobu Kazui
- Division of Cardiothoracic Surgery, University of Arizona, Tucson, AZ, USA
| | - Lipsa Jena
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology and the University of Arizona Center on Aging, The University of Arizona, Tucson, AZ, USA
| | - Scott Lick
- Division of Cardiothoracic Surgery, University of Arizona, Tucson, AZ, USA
| | - Janko Nikolich-Žugich
- Department of Immunobiology and the University of Arizona Center on Aging, The University of Arizona, Tucson, AZ, USA; BIO5 Institute, The University of Arizona, Tucson, AZ, USA
| | - John P Konhilas
- Department of Physiology, The University of Arizona, Tucson, AZ, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - May Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Samuel K Campos
- Department of Immunobiology and the University of Arizona Center on Aging, The University of Arizona, Tucson, AZ, USA; Department of Molecular & Cellular Biology, The University of Arizona, Tucson, AZ, USA; Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ, USA; BIO5 Institute, The University of Arizona, Tucson, AZ, USA
| | - Jared M Churko
- Department of Cellular and Molecular Medicine and Sarver Heart Center Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA; Department of Physiology, The University of Arizona, Tucson, AZ, USA; BIO5 Institute, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
7
|
Costa IBSDS, Andrade FTDA, Carter D, Seleme VB, Costa MS, Campos CM, Hajjar LA. Challenges and Management of Acute Coronary Syndrome in Cancer Patients. Front Cardiovasc Med 2021; 8:590016. [PMID: 34179121 PMCID: PMC8219848 DOI: 10.3389/fcvm.2021.590016] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 04/20/2021] [Indexed: 01/12/2023] Open
Abstract
Cancer and cardiovascular disease are the leading causes of mortality in the world. The prevalence of cardiovascular risk factors and coronary artery disease in cancer patients is elevated, and it is associated with high mortality. Several mechanisms, such as the proinflammatory and procoagulant states present in cancer patients, may contribute to these scenarios. Oncological therapy can predispose patients to acute thrombosis, accelerated atherosclerosis and coronary spasm. Treatment decisions must be individualized and based on the cancer history and balancing bleeding and thrombosis risks.
Collapse
Affiliation(s)
| | | | - Diego Carter
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Vinicius B. Seleme
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | | | - Carlos M. Campos
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Ludhmila Abrahão Hajjar
- Cancer Institute University of São Paulo, São Paulo, Brazil
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
8
|
Gordon HP, Katz MG, Fazal S, Gillespie VL, Fargnoli AS, Gubara SM, Madjarova SJ, Cohen JA. Inflammatory Responses with Left Ventricular Compromise after Induction of Myocardial Infarcts in Sheep (Ovis aries). Comp Med 2021; 71:240-246. [PMID: 34082856 DOI: 10.30802/aalas-cm-21-000005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Ischemic myocardial disease is a major cause of death among humans worldwide; it results in scarring and pallor of the myocardium and triggers an inflammatory response that contributes to impaired left ventricular function. This response includes and is evidenced by the production of several inflammatory cytokines including TNFα, IL1β, IL4, IFNγ, IL10 and IL6. In the current study, myocardial infarcts were induced in 6 mo old male castrated sheep by ligation of the left circumflex obtuse marginal arteries (OM 1 and 2). MRI was used to measure parameters of left ventricular function that include EDV, ESV, EF, SVI, dp/dt max and dp/dt min at baseline and at 4 wk and 3 mo after infarct induction. We also measured serum concentrations of an array of cytokines. Postmortem histologic findings corroborate the existence of left ventricular myocardial injury and deterioration. Our data show a correlation between serum cytokine concentrations and the development of myocardial damage and left ventricular functional compromise.
Collapse
Affiliation(s)
- Hylton P Gordon
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mt. Sinai, New York, New York;,
| | - Michael G Katz
- Department of Cardiology, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Shahood Fazal
- Department of Cardiology, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Virginia L Gillespie
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Anthony S Fargnoli
- Department of Cardiology, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Sarah M Gubara
- Department of Cardiology, Icahn School of Medicine at Mt. Sinai, New York, New York
| | - Sophia J Madjarova
- Department of Biology, Columbia University School of Medicine, New York, New York
| | - Jonathan A Cohen
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mt. Sinai, New York, New York
| |
Collapse
|
9
|
Baik AH, Oluwole OO, Johnson DB, Shah N, Salem JE, Tsai KK, Moslehi JJ. Mechanisms of Cardiovascular Toxicities Associated With Immunotherapies. Circ Res 2021; 128:1780-1801. [PMID: 33934609 PMCID: PMC8159878 DOI: 10.1161/circresaha.120.315894] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immune-based therapies have revolutionized cancer treatments. Cardiovascular sequelae from these treatments, however, have emerged as critical complications, representing new challenges in cardio-oncology. Immune therapies include a broad range of novel drugs, from antibodies and other biologics, including immune checkpoint inhibitors and bispecific T-cell engagers, to cell-based therapies, such as chimeric-antigen receptor T-cell therapies. The recognition of immunotherapy-associated cardiovascular side effects has also catapulted new research questions revolving around the interactions between the immune and cardiovascular systems, and the signaling cascades affected by T cell activation, cytokine release, and immune system dysregulation. Here, we review the specific mechanisms of immune activation from immunotherapies and the resulting cardiovascular toxicities associated with immune activation and excess cytokine production.
Collapse
Affiliation(s)
- Alan H Baik
- Division of Cardiovascular Medicine, Department of Medicine, UCSF, San Francisco, CA (A.H.B.)
| | - Olalekan O Oluwole
- Division of Oncology (D.B.J., J.J.M., O.O.O.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Douglas B Johnson
- Division of Oncology (D.B.J., J.J.M., O.O.O.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Nina Shah
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA (N.S., K.K.T.)
| | - Joe-Elie Salem
- Department of Pharmacology, Cardio-oncology Program, CIC-1901, APHP.Sorbonne Université, Paris, France (J.-E.S.)
- Cardio-Oncology Program, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.-E.S., J.J.M.)
| | - Katy K Tsai
- Division of Hematology and Oncology, Department of Medicine, UCSF, San Francisco, CA (N.S., K.K.T.)
| | - Javid J Moslehi
- Division of Cardiovascular Medicine (J.J.M.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Division of Oncology (D.B.J., J.J.M., O.O.O.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Cardio-Oncology Program, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (J.-E.S., J.J.M.)
| |
Collapse
|
10
|
Distinct cytokine profiles associated with COVID-19 severity and mortality. J Allergy Clin Immunol 2021; 147:2098-2107. [PMID: 33894209 PMCID: PMC8061091 DOI: 10.1016/j.jaci.2021.03.047] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/20/2021] [Accepted: 03/25/2021] [Indexed: 12/21/2022]
Abstract
Background Markedly elevated levels of proinflammatory cytokines and defective type-I interferon responses were reported in patients with coronavirus disease 2019 (COVID-19). Objective We sought to determine whether particular cytokine profiles are associated with COVID-19 severity and mortality. Methods Cytokine concentrations and severe acute respiratory syndrome coronavirus 2 antigen were measured at hospital admission in serum of symptomatic patients with COVID-19 (N = 115), classified at hospitalization into 3 respiratory severity groups: no need for mechanical ventilatory support (No-MVS), intermediate severity requiring mechanical ventilatory support (MVS), and critical severity requiring extracorporeal membrane oxygenation (ECMO). Principal-component analysis was used to characterize cytokine profiles associated with severity and mortality. The results were thereafter confirmed in an independent validation cohort (N = 86). Results At time of hospitalization, ECMO patients presented a dominant proinflammatory response with elevated levels of TNF-α, IL-6, IL-8, and IL-10. In contrast, an elevated type-I interferon response involving IFN-α and IFN-β was characteristic of No-MVS patients, whereas MVS patients exhibited both profiles. Mortality at 1 month was associated with higher levels of proinflammatory cytokines in ECMO patients, higher levels of type-I interferons in No-MVS patients, and their combination in MVS patients, resulting in a combined mortality prediction accuracy of 88.5% (risk ratio, 24.3; P < .0001). Severe acute respiratory syndrome coronavirus 2 antigen levels correlated with type-I interferon levels and were associated with mortality, but not with proinflammatory response or severity. Conclusions Distinct cytokine profiles are observed in association with COVID-19 severity and are differentially predictive of mortality according to oxygen support modalities. These results warrant personalized treatment of COVID-19 patients based on cytokine profiling.
Collapse
|
11
|
Cao Q, Lei H, Yang M, Wei L, Dong Y, Xu J, Nasser M, Liu M, Zhu P, Xu L, Zhao M. Impact of Cardiovascular Diseases on COVID-19: A Systematic Review. Med Sci Monit 2021; 27:e930032. [PMID: 33820904 PMCID: PMC8035813 DOI: 10.12659/msm.930032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
In December 2019, pneumonia of unknown cause broke out, and currently more than 150 countries around the world have been affected. Globally, as of 5: 46 pm CET, 6 November 2020, the World Health Organization (WHO) had reported 48 534 508 confirmed cases of COVID-19, including 1 231 017 deaths. The novel coronavirus disease (COVID-19) outbreak, caused by the SARS-CoV-2 virus, is the most important medical challenge in decades. Previous research mainly focused on the exploration of lung changes. However, with development of the disease and deepening research, more and more patients showed cardiovascular diseases, even in those without respiratory symptoms, and some researchers have found that underlying cardiovascular diseases increase the risk of infection. Although the related mechanism is not thoroughly studied, based on existing research, we speculate that the interaction between the virus and its receptor, inflammatory factors, various forms of the stress response, hypoxic environment, and drug administration could all induce the development of cardiac adverse events. Interventions to control these pathogenic factors may effectively reduce the occurrence of cardiovascular complications. This review summarizes the latest research on the relationship between COVID-19 and its associated cardiovascular complications, and we also explore possible mechanisms and treatments.
Collapse
Affiliation(s)
- Qingtai Cao
- Hunan Normal University School of Medicine, Changsha, Hunan, China (mainland)
| | - HanYu Lei
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - MengLing Yang
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Le Wei
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - YinMiao Dong
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| | - JiaHao Xu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Mi Nasser
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China (mainland)
| | - MengQi Liu
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland).,Xiangya School of Medicine, Central South University, Changsha, Hunan, China (mainland)
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China (mainland)
| | - LinYong Xu
- School of Life Science, Central South University, Changsha, Hunan, China (mainland)
| | - MingYi Zhao
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China (mainland)
| |
Collapse
|
12
|
Weiner DM, Durgin JS, Wysocka M, Rook AH. The immunopathogenesis and immunotherapy of cutaneous T cell lymphoma: Current and future approaches. J Am Acad Dermatol 2020; 84:597-604. [PMID: 33352268 DOI: 10.1016/j.jaad.2020.12.026] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 01/31/2023]
Abstract
In the past few decades, immunotherapy has emerged as an effective therapeutic option for patients with cutaneous T cell lymphoma (CTCL). CTCL is characterized by progressive impairment of multiple arms of the immune system. Immunotherapy targets these deficits to stimulate a more robust antitumor response, thereby both clearing the malignant T cells and repairing the immune dysfunction. By potentiating rather than suppressing the immune system, immunotherapy can result in longer treatment responses than alternatives such as chemotherapy. In recent years, advances in our understanding of the pathogenesis of CTCL have led to the development of several new agents with promising efficacy profiles. The second article in this continuing medical education series describes the current immunotherapeutic options for treatment of CTCL, with a focus on how they interact with the immune system and their treatment outcomes in case studies and clinical trials. We will discuss established CTCL immunotherapies, such as interferons, photopheresis, and retinoids; emerging therapies, such as interleukin-12 and Toll-like receptor agonists; and new approaches to targeting tumor antigens and checkpoint molecules, such as mogamulizumab, anti-programmed cell death protein 1, anti-CD47, and chimeric antigen receptor T cell therapy. We also describe the principles of multimodality immunotherapy and the use of total skin electron beam therapy in such regimens.
Collapse
Affiliation(s)
- David M Weiner
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Joseph S Durgin
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maria Wysocka
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alain H Rook
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
13
|
Peng W, Wu H, Tan Y, Li M, Yang D, Li S. Mechanisms and treatments of myocardial injury in patients with corona virus disease 2019. Life Sci 2020; 262:118496. [PMID: 32987060 PMCID: PMC7518803 DOI: 10.1016/j.lfs.2020.118496] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/06/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022]
Abstract
The infection epidemic event of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was formally declared a pandemic by World Health Organization on March 11th, 2020. Corona Virus Disease 2019 (COVID-19) is caused by SARS-CoV-2, a new type of coronavirus, which has high contagion and mainly causes respiratory symptoms. With the increase in confirmed cases, however, the infection symptoms turn to be diverse with secondary or first clinical symptoms relating to damage of the cardiovascular system and changes of myocardial enzyme spectrum, cardiac troponin I, electrocardiogram, cardiac function. The occurrence of extra-pulmonary manifestations, including immediately and long-term damage, means that the overall health burden caused by SARS-CoV-2 infection may be under-estimated because COVID-19 patients developed cardiovascular system injury are more likely to become serious. The factors such as directly pathogen-mediated damage to cardiomyocytes, down-regulated angiotensin-converting enzyme 2 (ACE2) expression, excessive inflammatory response, hypoxia and adverse drug reaction, are closely related to the occurrence and development of the course of COVID-19. In combination with recently published medical data of patients having SARS-CoV-2 infection and the latest studies, the manifestations of damage to cardiovascular system by COVID-19, possible pathogenic mechanisms and advances of the treatment are proposed in this article.
Collapse
Affiliation(s)
- Wenyi Peng
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu Military General Hospital, China; School of Medicine, Southwest Jiaotong University, Chengdu 610083, China
| | - Hao Wu
- Department of Toxicology, The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Shannxi Key Laboratory of Free Radical Biology and Medicine, School of Public Health, Fourth Military Medical University, China
| | - Yan Tan
- Medical Management Department, The General Hospital of Western Theater Command, Chengdu Military General Hospital, China
| | - Mei Li
- School of Medicine, Southwest Jiaotong University, Chengdu 610083, China
| | - Dachun Yang
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu Military General Hospital, China.
| | - Shuang Li
- Department of Cardiology, The General Hospital of Western Theater Command, Chengdu Military General Hospital, China.
| |
Collapse
|
14
|
Cheraghmakani H, Samaee H, Ghazaeian M. Interferon Beta-1a Cardiomyopathy in a Patient with Multiple Sclerosis: Case Report. Mult Scler Relat Disord 2020; 44:102219. [DOI: 10.1016/j.msard.2020.102219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/11/2020] [Accepted: 05/18/2020] [Indexed: 10/24/2022]
|
15
|
Aggarwal G, Henry BM, Aggarwal S, Bangalore S. Cardiovascular Safety of Potential Drugs for the Treatment of Coronavirus Disease 2019. Am J Cardiol 2020; 128:147-150. [PMID: 32425199 PMCID: PMC7228893 DOI: 10.1016/j.amjcard.2020.04.054] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/13/2020] [Accepted: 04/20/2020] [Indexed: 12/23/2022]
Abstract
Coronavirus disease 2019 (COVID-19) has become a global pandemic. It is still uncontrolled in most countries and no therapies are currently available. Various drugs are under investigation for its treatment. The disease is known to have worse outcomes in patients who have underlying cardiovascular disease. Chloroquine/hydroxychloroquine, azithromycin, remdesivir and lopinavir/ritonavir are currently being studied in trials and show some promise. Conduction disorders, heart failure, and mortality have been reported with the use of these drugs. It is important to have knowledge of potential cardiotoxic effects of these drugs before using them for COVID-19 patients for better allocation of healthcare resources and improvement in clinical outcomes.
Collapse
|
16
|
Abstract
Coronavirus disease 2019 (COVID-19) is a pandemic infection caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). COVID-19 significantly affects multiple systems including the cardiovascular system. Most importantly, in addition to the direct injury from the virus per se, the subsequent cytokine storm, an overproduction of immune cells and their activating compounds, causes devastating damage. To date, emerging anti-SARS-CoV-2 treatments are warranted to control epidemics. Several candidate drugs have been screened and are currently under investigation. These primarily include antiviral regimens and immunomodulatory regimens. However, beyond the anti-SARS-CoV-2 effects, these drugs may also have risks to the cardiovascular system, especially altering cardiac conduction. Herein, we review the cardiovascular risks of potential anti-COVID-19 drugs.
Collapse
|
17
|
Lapumnuaypol K, Thongprayoon C, Wijarnpreecha K, Cheungpasitporn W. Impact of hepatitis C sustained viral response on cardiovascular diseases: a meta-analysis. Hosp Pract (1995) 2019; 47:105-110. [PMID: 31018721 DOI: 10.1080/21548331.2019.1612066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background: Hepatitis C virus-infected patients are found to have increased risks of cardiovascular disease (CVD)-related morbidity and mortality. However, the effect of treatment on cardiovascular risk remains unknown. We performed a systematic review and meta-analysis to assess the effect of Sustained Virologic Response (SVR) on cardiovascular outcome in chronic HCV-infected patients. Methods: A systematic review was conducted in MEDLINE, EMBASE, Cochrane databases from inception through November 2018 to identify studies that assessed the effect of SVR on CVDs. Effect estimates from the individual study were extracted and combined using random-effect, generic inverse variance method of DerSimonian and Laird. Results: Seven cohort studies with a total of 53,841 HCV-infected patients with average follow-up time of 5 years were enrolled. When compared with HCV-infected patients who do not achieve SVR, patients with SVR have a reduced risk of overall CVDs with the pooled hazard ratio of 0.76 (95% confidence interval 0.61-0.94). Egger's regression asymmetry test was performed and showed no publication bias. Conclusions: Our study demonstrates a significant association between SVR after HCV treatment and reduced risk of overall CVDs.
Collapse
Affiliation(s)
- Kamolyut Lapumnuaypol
- Department of Internal Medicine, Albert Einstein Medical Center , Philadelphia , PA , USA
| | - Charat Thongprayoon
- Department of Nephrology and Hypertension, Mayo Clinic , Rochester , MN , USA
| | - Karn Wijarnpreecha
- Department of Gastroenterology, Mayo Clinic Hospital , Jacksonville , FL , USA
| | - Wisit Cheungpasitporn
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center , Jackson , MS , USA
| |
Collapse
|
18
|
Affiliation(s)
- Kenji Onoue
- Department of Cardiovascular Medicine, Nara Medical University
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University
| |
Collapse
|
19
|
Nishio K, Arase T, Tada H, Tachibana H. Interferon related pericarditis: Review. World J Cardiol 2017; 9:553-557. [PMID: 28706591 PMCID: PMC5491473 DOI: 10.4330/wjc.v9.i6.553] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/13/2017] [Accepted: 05/05/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To conduct a review of “interferon related pericarditis”.
METHODS We searched MEDLINE, EMBASE, Cinahl, and the Cochrane Database from the earliest available date through September 2016. A search strategy using the Medical Subject Headings and text keywords “interferon” and ”pericarditis” were used.
RESULTS Nine case reports were eligible for the present study. Six of 8 cases were women and the mean age was 43.8 ± 13.8 years with chronic hepatitis C in 6 cases, malignant melanoma in 2 cases and chronic myelogenous leukemia in 1 case. The patients complained of chest pain in 6 cases, dyspnea in 5 cases and edema in 2 cases. Pericardial friction rub was heard in 3 of 9 cases. Congestive heart failure occurred in 3 of 9 cases. Two mechanisms for pericarditis were demonstrated, one is autoimmune included lupus like syndrome in 2 cases and the other is cardio toxicity in 4 cases. Treatment of interferon related pericarditis is discontinuation of Interferon treatment. Four of 9 cases were treated with prednisone and 4 with nonsteroidal anti-inflammatory drugs.
CONCLUSION Interferon related pericarditis still remains uncertain. Treatment of interferon related pericarditis rests mainly on early recognition and drug discontinuation. Interferon related pericarditis was treated with steroid and/or nonsteroidal anti-inflammatory drugs.
Collapse
|
20
|
Recurrent Pericarditis, an Unexpected Effect of Adjuvant Interferon Chemotherapy for Malignant Melanoma. Case Rep Cardiol 2016; 2016:1342028. [PMID: 27418981 PMCID: PMC4933855 DOI: 10.1155/2016/1342028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/27/2022] Open
Abstract
Drug-induced pericarditis is a well-described cardiac pathology that can result from a variety of medications; however, interferon-mediated pericarditis is extremely rare. We present a case of a young female with recurrent pericarditis due to interferon therapy. The role of interferon in adjuvant chemotherapy is well known and yields good effect, but this case highlights the very uncommon phenomena of interferon induced pericarditis and the significant distress it can cause.
Collapse
|
21
|
Massilamany C, Koenig A, Reddy J, Huber S, Buskiewicz I. Autoimmunity in picornavirus infections. Curr Opin Virol 2015; 16:8-14. [PMID: 26554915 DOI: 10.1016/j.coviro.2015.10.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/14/2015] [Accepted: 10/18/2015] [Indexed: 12/16/2022]
Abstract
Enteroviruses are small, non-enveloped, positive-sense single-strand RNA viruses, and are ubiquitously found throughout the world. These viruses usually cause asymptomatic or mild febrile illnesses, but have a propensity to induce severe diseases including type 1 diabetes and pancreatitis, paralysis and neuroinflammatory disease, myocarditis, or hepatitis. This pathogenicity may result from induction of autoimmunity to organ-specific antigens. While enterovirus-triggered autoimmunity can arise from multiple mechanisms including antigenic mimicry and release of sequestered antigens, the recent demonstration of T cells expressing dual T cell receptors arising as a natural consequence of Theiler's virus infection is the first demonstration of this autoimmune mechanism.
Collapse
Affiliation(s)
- Chandirasegaran Massilamany
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Andreas Koenig
- Department of Medicine and University of Vermont, Colchester, VT 05446, USA
| | - Jay Reddy
- School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
| | - Sally Huber
- Department of Pathology and Vermont Center for Immunobiology and Infectious Diseases, University of Vermont, Colchester, VT 05446, USA
| | - Iwona Buskiewicz
- Department of Pathology and Vermont Center for Immunobiology and Infectious Diseases, University of Vermont, Colchester, VT 05446, USA.
| |
Collapse
|
22
|
Rodríguez-Carrio J, Alperi-López M, López P, Alonso-Castro S, Ballina-García FJ, Suárez A. Angiogenic T cells are decreased in rheumatoid arthritis patients. Ann Rheum Dis 2015; 74:921-7. [PMID: 24399233 DOI: 10.1136/annrheumdis-2013-204250] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 12/15/2013] [Indexed: 01/15/2023]
Abstract
OBJECTIVE The mechanisms underlying the increased cardiovascular risk (CVR) of rheumatoid arthritis (RA) patients remain unclear. Since the recently discovered angiogenic T cells (Tang) could have a role in endothelial repair through cooperating with endothelial progenitor cells (EPC), the main aim of this study was to analyse the Tang and EPC populations in relation to disease-specific features and traditional CVR factors. METHODS Tang (CD3(+)CD31(+)CXCR4(+)) and EPC (CD34(+)VEGFR2(+)CD133(+)) populations were quantified by flow cytometry in peripheral blood samples from 103 RA patients and 18 matched healthy controls (HC). Clinical features and traditional CVR factors were obtained from clinical records, and 28-joint Disease Activity Score was used for measuring disease activity. Interferon (IFN) α serum levels were measured by immunoassays. RESULTS Tang and EPC were strongly decreased in RA patients. In HC, but not in patients, both populations were positively correlated and inversely related to low density lipoprotein- and total-cholesterol levels. Sex, diabetes, dyslipidaemia, hypertension or obesity did not significantly influence Tang in patients, although detected in smokers. However, Tang were closely related to disease activity, autoantibody positivity and IFNα levels. Multiple regression analysis adjusted for traditional CVR factors confirmed that only disease activity, age at diagnosis, antinuclear antibody positivity and smoking habit could predict Tang frequency. Finally, patients who had suffered a CV event since their RA diagnosis presented higher Tang decrease and IFNα levels than those who were CV event-free. CONCLUSIONS Disease-specific parameters, including disease activity, autoantibody profiles and IFNα levels, are associated with Tang decrease in RA, thus probably accounting for CVR.
Collapse
Affiliation(s)
- Javier Rodríguez-Carrio
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Asturias, Spain
| | - Mercedes Alperi-López
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | - Patricia López
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Asturias, Spain
| | - Sara Alonso-Castro
- Department of Rheumatology, Hospital Universitario Central de Asturias, Oviedo, Asturias, Spain
| | | | - Ana Suárez
- Area of Immunology, Department of Functional Biology, University of Oviedo, Oviedo, Asturias, Spain
| |
Collapse
|
23
|
Type I IFNs as biomarkers in rheumatoid arthritis: towards disease profiling and personalized medicine. Clin Sci (Lond) 2014; 128:449-64. [DOI: 10.1042/cs20140554] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RA (rheumatoid arthritis) is a chronic rheumatic condition hallmarked by joint inflammation and destruction by self-reactive immune responses. Clinical management of RA patients is often hampered by its heterogeneous nature in both clinical presentation and outcome, thereby highlighting the need for new predictive biomarkers. In this sense, several studies have recently revealed a role for type I IFNs (interferons), mainly IFNα, in the pathogenesis of a subset of RA patients. Genetic variants associated with the type I IFN pathway have been linked with RA development, as well as with clinical features. Moreover, a role for IFNα as a trigger for RA development has also been described. Additionally, a type I IFN signature has been associated with the early diagnosis of RA and clinical outcome prediction in patients undergoing biological drug treatment, two challenging issues for decision-making in the clinical setting. Moreover, these cytokines have been related to endothelial damage and vascular repair failure in different autoimmune disorders. Therefore, together with chronic inflammation and disease features, they could probably account for the increased cardiovascular disease morbidity and mortality of these patients. The main aim of the present review is to provide recent evidence supporting a role for type I IFNs in the immunopathology of RA, as well as to analyse their possible role as biomarkers for disease management.
Collapse
|
24
|
Sato A, Yamauchi M, Yamada T, Kumano R, Adachi K, Ishii T, Hayashi M, Kumon D. Successful natural interferon-β plus ribavirin therapy in a chronic hepatitis C patient after discontinuation of interferon-α treatment due to arrhythmia and interstitial pneumonia. Clin J Gastroenterol 2014; 7:355-60. [PMID: 25132867 PMCID: PMC4124242 DOI: 10.1007/s12328-014-0500-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/20/2014] [Indexed: 11/28/2022]
Abstract
A 71-year-old female patient with hepatitis C virus genotype 1 had previously discontinued interferon (IFN)-α plus ribavirin therapy, pegylated IFN-α (pegIFN-α) monotherapy, and natural IFN-α monotherapy because of arrhythmia, interstitial pneumonia, and severe neurovegetative symptoms. She subsequently completed 72 weeks of natural IFN-β plus ribavirin therapy without remarkable adverse effects and achieved a sustained viral response, suggesting differences in the pharmacological properties and biological effects of IFN-α and IFN-β. Thus, natural IFN-β plus ribavirin therapy may be a treatment option for patients with poor tolerance to IFN-α or pegIFN-α treatments.
Collapse
Affiliation(s)
- Akira Sato
- Division of Gastroenterology, Department of Internal Medicine, St. Marianna University School of Medicine Yokohama City Seibu Hospital, 1197-1 Yasashicho, Asahi-ku, Yokohama, 241-0811 Japan
| | - Masahiro Yamauchi
- Division of Cardiology, Department of Internal Medicine, St. Marianna University School of Medicine Yokohama City Seibu Hospital, 1197-1 Yasashicho, Asahi-ku, Yokohama, 241-0811 Japan
| | - Takayuki Yamada
- Department of Radiology, St. Marianna University School of Medicine Yokohama City Seibu Hospital, 1197-1 Yasashicho, Asahi-ku, Yokohama, 241-0811 Japan
| | - Reiko Kumano
- Department of Radiology, St. Marianna University School of Medicine Yokohama City Seibu Hospital, 1197-1 Yasashicho, Asahi-ku, Yokohama, 241-0811 Japan
| | - Kayo Adachi
- Division of Gastroenterology, Department of Internal Medicine, St. Marianna University School of Medicine Yokohama City Seibu Hospital, 1197-1 Yasashicho, Asahi-ku, Yokohama, 241-0811 Japan
| | - Toshiya Ishii
- Division of Gastroenterology, Department of Internal Medicine, St. Marianna University School of Medicine Yokohama City Seibu Hospital, 1197-1 Yasashicho, Asahi-ku, Yokohama, 241-0811 Japan
| | - Mikihito Hayashi
- Division of Gastroenterology, Department of Internal Medicine, St. Marianna University School of Medicine Yokohama City Seibu Hospital, 1197-1 Yasashicho, Asahi-ku, Yokohama, 241-0811 Japan
| | - Daisuke Kumon
- Division of Gastroenterology, Department of Internal Medicine, St. Marianna University School of Medicine Yokohama City Seibu Hospital, 1197-1 Yasashicho, Asahi-ku, Yokohama, 241-0811 Japan
| |
Collapse
|
25
|
Zhao W, Ji F, Yu S, Li Z, Deng H. Dilated cardiomyopathy and hypothyroidism associated with pegylated interferon and ribavirin treatment for chronic hepatitis C: case report and literature review. Braz J Infect Dis 2014; 18:110-113. [PMID: 24120833 PMCID: PMC9425206 DOI: 10.1016/j.bjid.2013.05.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 05/23/2013] [Indexed: 01/20/2023] Open
Abstract
Pegylated interferon alpha (Peg IFN-α) in combination with ribavirin is the backbone of treatment in chronic hepatitis C (CHC). Cardiotoxicity due to interferon therapy is rare. The most frequent cardiovascular complications are arrhythmias and ischemic manifestations. Cardiomyopathy is extremely rare but can be life threatening. We present the case of a 41-year-old female patient with CHC in whom Peg IFN-α induced dilated cardiomyopathy and hypothyroidism. Chest radiography showed an enlarged and globular cardiac silhouette and pulmonary congestion. Echocardiography showed decreased left ventricular systolic function with an ejection fraction of 32% and fractional shortening of 15%. Cardiomyopathy had a complete remission after cessation of antiviral therapy with short-term heart failure medications and supportive care. Then we review the current literature about interferon induced cardiomyopathy in patients with HCV infection, as well as share our clinical experience in diagnosing and managing this rare complication.
Collapse
Affiliation(s)
- Wenxue Zhao
- Department of Infectious Disease, The Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Fanpu Ji
- Department of Infectious Disease, The Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China.
| | - Shanshan Yu
- Department of Ultrasound Medicine, The Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Zongfang Li
- Department of Hepatology and Surgery, The Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Hong Deng
- Department of Infectious Disease, The Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
26
|
Sakabe M, Yoshioka R, Fujiki A. Sick sinus syndrome induced by interferon and ribavirin therapy in a patient with chronic hepatitis C. J Cardiol Cases 2013; 8:173-175. [PMID: 30534284 DOI: 10.1016/j.jccase.2013.08.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 07/04/2013] [Accepted: 08/01/2013] [Indexed: 11/17/2022] Open
Abstract
The therapeutic effect of interferon (IFN) on chronic hepatitis C and its adverse effects have been well documented. Although the incidence of IFN-related cardiotoxicity is low, careful observation is necessary because of its possible fatal outcome. We describe a 45-year-old woman who suffered from sinus node dysfunction after the combination therapy of pegylated IFN-alpha and ribavirin for chronic hepatitis C. Despite the cessation of IFN therapy, sinus node dysfunction was not reversible, and led her to the implantation of permanent pacemaker. Physicians should therefore be aware of the possibility of sinus node dysfunction in patients receiving IFN therapy. <Learning objective: Pegylated interferon-alpha has been widely used to treat hepatitis C virus infection, which is associated with a wide variety of adverse effects. There is a limited number of case reports regarding suspected interferon-induced cardiotoxicity, especially bradyarrhythmia. We report a case with chronic hepatitis C who developed sick sinus syndrome after interferon-alpha therapy.>.
Collapse
Affiliation(s)
- Masao Sakabe
- Division of Cardiology, Shizuoka Red Cross Hospital, 8-2 Otemachi, Aoi-ku, Shizuoka 420-0853, Japan
| | - Ryozo Yoshioka
- Division of Cardiology, Shizuoka Red Cross Hospital, 8-2 Otemachi, Aoi-ku, Shizuoka 420-0853, Japan
| | - Akira Fujiki
- Division of Cardiology, Shizuoka Red Cross Hospital, 8-2 Otemachi, Aoi-ku, Shizuoka 420-0853, Japan
| |
Collapse
|
27
|
Durante-Mangoni E, Iossa D, Pinto D, De Vincentiis L, Ragone E, Utili R. Safety and efficacy of peginterferon alpha plus ribavirin in patients with chronic hepatitis C and coexisting heart disease. Dig Liver Dis 2011; 43:411-5. [PMID: 21310673 DOI: 10.1016/j.dld.2010.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 12/18/2010] [Accepted: 12/30/2010] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic hepatitis C patients with coexisting heart disease are often denied antiviral treatment due to safety concerns. However, this is not evidence-based. AIMS To evaluate safety and efficacy of pegylated interferon and ribavirin in chronic hepatitis C patients with heart disease. METHODS Patients with overt heart disease (ischaemic heart disease, prior mechanical heart valve replacement, chronic arrhythmias and cardiomyopathy) and chronic hepatitis C were treated with standard pegylated interferon/ribavirin doses for standard duration. Cardiovascular safety was monitored by electrocardiography, echocardiography and measurement of troponin and B-type natriuretic peptide. RESULTS Twenty-three patients (65.2% male, median age 57 years, 47.8% genotype 1) were treated. Three patients (13%) suspended treatment prematurely; 52% obtained sustained virological response, 39% relapsed, 9% were non-responders. No serious adverse event was observed. Post-treatment clinical examination, electrocardiography and echocardiography did not show any sign of progression of the pre-existing heart disease. CONCLUSIONS Treatment with pegylated interferon/ribavirin may be safely offered to carefully selected chronic hepatitis C patients with coexisting, clinically significant heart disease. In these patients, the outcome of antiviral treatment overlaps that observed in other patient subgroups.
Collapse
|
28
|
Kastalli S, El Aïdli S, Mourali S, Zaïem A, Daghfous R, Lakhal M. Cardiac arrhythmia induced by interferon beta-1a. Fundam Clin Pharmacol 2011; 26:207-9. [PMID: 21352349 DOI: 10.1111/j.1472-8206.2010.00914.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cardiac adverse effects have never been reported with interferon (INF) beta. We report a case of left bundle branch block in a 35-year-old woman treated with INF beta-1a for multiple sclerosis. Five years after INF therapy, she presented loss of consciousness, retrosternal pains, short breath and lowered tolerance of effort. ECG and Holter 24-h ECG monitoring revealed permanent complete left bundle branch block. Nine months after stopping INF, no abnormalities were found at ECG and echocardiogram examination.
Collapse
Affiliation(s)
- Sarrah Kastalli
- Service de recueil et analyse des effets indésirables, Centre National de Pharmacovigilance, 9 Av Dr Zouheir Essafi 1006 Tunis, Tunisia
| | | | | | | | | | | |
Collapse
|
29
|
Nishio K, Konndo T, Okada S, Enchi M. Pericarditis and chronic inflammatory demyelinating polyneuropathy during therapy with pegylated interferon alfa-2a for chronic hepatitis C. World J Hepatol 2010; 2:358-61. [PMID: 21161021 PMCID: PMC2999299 DOI: 10.4254/wjh.v2.i9.358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 08/15/2010] [Accepted: 08/22/2010] [Indexed: 02/06/2023] Open
Abstract
We report a case of pericarditis and chronic inflammatory demyelinating polyneuropathy with biological signs of a lupus-like syndrome due to pegylated interferon alfa-2a therapy during treatment for chronic hepatitis C. The patient developed moderate weakness in the lower limbs and dyspnea. He was hospitalized for congestive heart failure. An electrocardiogram showed gradual ST-segment elevation in leads V(1) through V(6) without coronary artery disease. A transthoracic cardiac ultrasonographic study revealed moderate pericardial effusion with normal left ventricular function. Anti-DNA antibody and antids DNA IgM were positive. Neurological examination revealed a symmetrical predominantly sensory polyneuropathy with impairment of light touch and pin prick in globe and stoking-like distribution. Treatment with prednisolone improved the pericarditis and motor nerve disturbance and the treatment with intravenous immunoglobulin improved the sensory nerve disturbance.
Collapse
Affiliation(s)
- Kazuaki Nishio
- Kazuaki Nishio, Takeshi Konndo, The Third Department of Internal Medicine, School of Medicine Showa University, Tokyo 142-8666, Japan
| | | | | | | |
Collapse
|
30
|
Marcolino MS, Ribeiro AL, Clementino NCD, Nunes MDCP, Barbosa MM, Silva MHCR, Bittencourt HNS, Geleijnse ML, Boersma E. The use of imatinib mesylate has no adverse effects on the heart function. Results of a pilot study in patients with chronic myeloid leukemia. Leuk Res 2010; 35:317-22. [PMID: 20692037 DOI: 10.1016/j.leukres.2010.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 07/07/2010] [Accepted: 07/07/2010] [Indexed: 10/19/2022]
Abstract
To investigate cardiac effects of imatinib at an extended follow-up (median 12.4 months), 12 chronic myeloid leukemia patients underwent cardiac screening. No significant changes on the frequency of cardiovascular signs and symptoms, electrocardiographic abnormalities, echocardiographic measurements and BNP levels were observed. Median ejection fraction was 67% at baseline versus 68% at follow-up (median intra-patient change 0.5%). Median BNP levels were 8.3 versus 7.3pg/mL (median intra-patient change 0.2pg/mL). Troponin I measures were below the lower limit of detection, whereas strain measures were similar to healthy control. This pilot study suggests that it is probably safe to perform cardiac monitoring on an annual basis.
Collapse
|
31
|
Charniot JC, Vignat N, Monsuez JJ, Kidouche R, Avramova B, Artigou JY, Albertini JP. Cardiogenic shock associated with reversible dilated cardiomyopathy during therapy with regular doses of venlafaxine. Am J Emerg Med 2010; 28:256.e1-5. [DOI: 10.1016/j.ajem.2009.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2009] [Revised: 05/17/2009] [Accepted: 05/17/2009] [Indexed: 10/19/2022] Open
|
32
|
Takase B, Uehata A, Fujioka T, Kondo T, Nishioka T, Isojima K, Satomura K, Ohsuzu F, Kurita A. Endothelial dysfunction and decreased exercise tolerance in interferon-alpha therapy in chronic hepatitis C: relation between exercise hyperemia and endothelial function. Clin Cardiol 2009; 24:286-90. [PMID: 11303695 PMCID: PMC6654793 DOI: 10.1002/clc.4960240406] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND We previously reported that reversible endothelial dysfunction is caused by interferon-alpha therapy (IFN) in patients with chronic hepatitis C. In experimental studies, limb blood flow during exercise is reported to be dependent on endothelium-derived nitric oxide. HYPOTHESIS The purpose of this study was to confirm the effect of IFN on endothelial function and to investigate whether exercise hyperemia is dependent on endothelial function in humans. METHODS We performed symptom-limited exercise treadmill testing and measured flow-mediated vasodilation (FMD, endothelium-dependent vasodilation) and sublingual glyceryl-trinitrate-induced dilation (GTN-D, 0.3 mg, endothelium-independent vasodilation) in the brachial artery by using high-resolution ultrasound in 10 patients with chronic active hepatitis C (age 53 +/- 11 years, 2 men, 8 women) before and immediately after administration of recombinant interferon 2b (10 million U/day) for 4 weeks. RESULTS There were no significant abnormal findings in any patients in routine studies of 24-h ambulatory electrocardiogram monitoring, two-dimensional echocardiography, and exercise treadmill testing both before and after treatment. Leg fatigue and exhaustion were the reasons for termination of exercise treadmill testing in each patient. Pressure rate product was calculated at rest and peak exercise. Interferon-alpha therapy significantly (p<0.05) decreased FMD (6.8 +/- 3.1 vs. 1.9 +/- 2.6%), exercise treadmill testing tolerance time (437 +/- 89 vs. 395 +/- 62 s) and peak pressure rate product (283 +/- 41 vs. 241 +/- 47 mmHg x beats/min x 10(-2)), but not GTN-D (13.4 +/- 5.4 vs. 17.0 +/- 5.5%). The change of FMD due to IFN significantly and highly correlated with exercise treadmill testing tolerance time (r = 0.86, p<0.001), but not with change of peak pressure rate product, suggesting that FMD is more closely related to the condition of the peripheral circulation than is cardiac performance. CONCLUSION These results suggest that IFN in patients with chronic hepatitis C impairs endothelial function and exercise tolerance, and that endothelial function might be at least partly involved in exercise hyperemia in humans.
Collapse
Affiliation(s)
- B Takase
- National Defense Medical College, Internal Medicine-1, Research Center, Tokorozawa, Saitama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hashemi N, Rossi S, Navarro VJ, Herrine SK. Safety of peginterferon in the treatment of chronic hepatitis C. Expert Opin Drug Saf 2009; 7:771-81. [PMID: 18983223 DOI: 10.1517/14740330802423291] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Combination of 'pegylated' interferons (IFNs) plus ribavirin, the standard treatment of chronic hepatitis C (CHC), is frequently associated with side effects. Anticipation, recognition and proper management of these side effects are important to ensure compliance with therapy and achievement of sustained virologic response. OBJECTIVE To illustrate the side effect profile of pegIFN-alpha in the treatment of CHC. METHODS Studies and abstracts were identified through a computerized, English language literature search. Key search terms included peginterferon and CHC. Information available only in abstract form was retrieved from national and international hepatology associations. RESULTS Most adverse events occurring with combination therapy can be anticipated and managed appropriately; therefore, premature discontinuation of therapy owing to side effects is not required in most patients.
Collapse
Affiliation(s)
- Nikroo Hashemi
- Thomas Jefferson University, Division of Gastroenterology and Hepatology, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
34
|
|
35
|
Kartal ED, Alpat SN, Ozgunes I, Usluer G. Adverse effects of high-dose interferon-alpha-2a treatment for chronic hepatitis B. Adv Ther 2007; 24:963-71. [PMID: 18029321 DOI: 10.1007/bf02877700] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of this study was to assess morbidity and the incidence of adverse effects during interferon (IFN)-alpha-2a treatment of patients with chronic hepatitis B. This prospective study included 48 consecutive patients with chronic hepatitis B who underwent IFN-alpha-2a treatment from January 2003 to August 2005. Adverse effects related to IFN treatment were recorded during this period and for 6 mo after treatment. Adverse effects that led to dose reduction or early discontinuation of IFN treatment were examined. Complete response was reported in 25% of patients. At least 1 adverse effect was documented in 88% of patients. Flu-like symptoms were the most frequently observed adverse effects (88%), and thrombocytopenia (63%), leukopenia (54%), and anemia (23%) were also reported. Bleeding occurred in 2 patients. Other adverse effects included neuropsychiatric signs (21%), alopecia (19%), weight loss (17%), thyroid disorders (19%), menstrual cycle irregularities (8%), skin lesions (8%), and dry cough (4%). Adverse effects that led to dose reduction or early discontinuation of IFN treatment occurred in 19% of patients and included impotence, depression, seizure, thyroid disorders, severe thrombocytopenia, and intestinal bleeding. These effects were found to be unrelated to treatment response. No relationship was detected between patient age, duration of treatment, and adverse effects of IFN. Although IFN-alpha-2a treatment induced various adverse effects in patients with chronic hepatitis B, most of these effects were reversible or could be ameliorated. Adverse effects that led to dose reduction or early discontinuation of IFN treatment were found to be unrelated to complete response.
Collapse
Affiliation(s)
- Elif Doyuk Kartal
- Department of Infectious Diseases and Clinical Bacteriology, Eskisehir Osmangazi University Medical Faculty, Eskisehir, Turkey.
| | | | | | | |
Collapse
|
36
|
Condat B, Asselah T, Zanditenas D, Estampes B, Cohen A, O'Toole D, Bonnet J, Ngo Y, Marcellin P, Blazquez M. Fatal cardiomyopathy associated with pegylated interferon/ribavirin in a patient with chronic hepatitis C. Eur J Gastroenterol Hepatol 2006; 18:287-9. [PMID: 16462543 DOI: 10.1097/00042737-200603000-00010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
We report the case of a 45-year-old man with HCV treated with pegylated interferon-alpha/ribavirin, in whom fatal cardiomyopathy occurred. Cardiomyopathy is a rare complication of high dose of standard interferon but has never been reported with pegylated interferon. The relationship between pegylated interferon-alpha/ribavirin and the development of cardiomyopathy is highly probable for the following reasons: (1) a cardiologist consultation with specific investigations had been performed before treatment excluding a pre-existing cardiomyopathy; (2) symptoms of advanced dilated cardiomyopathy appeared immediately after the end of treatment; (3) other causes of cardiomyopathy have been ruled out. In all except one of the 21 reported cases with standard interferon, cardiomyopathy was reversible. In our patient, fatal cardiomyopathy occurred with a usual dose of pegylated interferon-alpha. Clinicians should be aware of this potential complication when evaluating the ratio benefit/risk of treatment in patients with chronic hepatitis C infection.
Collapse
Affiliation(s)
- Bertrand Condat
- Service d'hépato-gastroentérologie, Hôpital Saint-Camille, Bry sur Marne, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Takase B, Hamabe A, Uehata A, Hujioka T, Kondo T, Matsui T, Ohsuzu F, Ishihara M. Recombinant interferon alpha treatment decreases heart rate variability indices and impairs exercise tolerance in patients with chronic hepatitis. Biomed Pharmacother 2005; 59 Suppl 1:S163-8. [PMID: 16275487 DOI: 10.1016/s0753-3322(05)80025-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Complications of interferon (IFN) therapy include cardiac arrhythmias, impaired cardiac function and myocardial ischemia. Decreased heart rate variability (HRV) indices, impaired exercise tolerance and decreased left ventricular (LV) function are related to unfavorable outcome of heart disease. To investigate the effect of IFN therapy on HRV, exercise tolerance and cardiac function, 24-h ambulatory electrocardiographic monitoring (AECG), two-dimensional echocardiography, and exercise treadmill testing (ETT) was performed in 9 patients (age 56 +/- 9 years-old) with chronic hepatitis and without underlying heart disease before and after treatment with IFN (recombinant alpha 2b; 10 x 10(6) U/day for 4 weeks). HRV parameters consisted of standard deviation of RR interval (sdNN, ms), SDANN (ms), S.D. index (ms), rMSSD (ms), pNN50 (%) and frequency analysis of heart rate spectrum resulted in low (ms, 0.04-0.15 Hz), high (ms, 0.15-0.40 Hz) and total (ms, 0.01-1.00 Hz) frequency components. Ischemia was not detected by AECG or ETT, and LV function was normal after INF treatment in all patients. However, INF treatment resulted in a decrease in exercise tolerance time (449 +/- 94 s vs. 329 +/- 67 s, P < 0.05) and a decrease in several HRV parameters (S.D. index, 42 +/- 5 ms vs. 37 +/- 9 ms; rMSSD, 22 +/- 5 ms vs. 19 +/- 4 ms; pNN50, 4 +/- 3% vs. 2 +/- 1%; P < 0.05). Further, patients treated with INF tended to have a lower sdNN and total frequency spectra, although this difference did not reach the level of statistical significance. These data suggest that the arrhythmogenic effect of INF may be mediated by decreases in HRV and impairment of exercise tolerance even in patients without overt heart diseases. Further, INF therapy may be contraindicated in patients with predisposing severe cardiac disorders, including arrhythmias, ischemia and decreased LV function.
Collapse
Affiliation(s)
- B Takase
- Division of Biomedical Engineering, National Defense Medical College Research Institute, 3-2 Namiki, Tokorozawa, Saitama 359-8513, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Kawai K, Hata K, Kawai H, Takaoka H, Miyata-Fukuoka Y, Okubo H, Yokoyama M. Pathophysiological Characteristics and Responsiveness to Neurohormonal Antagonism in Idiopathic Dilated Cardiomyopathy Patients With Antihepatitis C Virus Antibody. Int Heart J 2005; 46:407-17. [PMID: 16043937 DOI: 10.1536/ihj.46.407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A high prevalence of hepatitis C virus (HCV) infection has been reported among idiopathic dilated cardiomyopathy (DCM) patients. We examined the prevalence of DCM patients with HCV antibody, and the pathophysiological characteristics and responsiveness to neurohormonal antagonism in DCM with HCV. HCV antibodies were determined in 540 patients with cardiac diseases. In 117 DCM patients, clinicopathologic data were evaluated before and 1 year after angiotensin converting enzyme inhibitor and/or beta-blocker (ACE-inhibitor/BB) administration and their prognosis was followed-up for a mean of 72 +/- 41 months. HCV antibodies were found in 12 of 135 DCM patients (8.9%) and in 37 of 405 patients without DCM (9.1%) (P = NS). At baseline, contrary to DCM without HCV, DCM with HCV was associated (P < 0.05) with greater left ventricular (LV) end-diastolic and end-systolic dimension, LV mass, and myocardial diameter in endomyocardial biopsy, and lower % fractional shortening. By multivariate analysis, HCV infection was independently associated with larger LV end-systolic dimension among DCM patients (P = 0.005). The advanced LV dilatation and hypertrophy in DCM with HCV decreased more in response to the ACE-inhibitor/BB therapy compared to DCM without HCV. There were no differences between DCM patients with and without HCV in survival and cardiac event-free rates. In summary, although HCV infection appears not to be the specific cause of DCM, HCV may enhance ventricular remodeling leading to heart failure among DCM patients. Nevertheless, the advanced ventricular remodeling with HCV was adequately reversed by neurohormonal antagonism, and did not lead to an unfavorable outcome.
Collapse
Affiliation(s)
- Keisuke Kawai
- Division of Cardiovascular and Respiratory Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | | | | | | | | | | |
Collapse
|
39
|
Wisniewski B, Denis J, Fischer D, Labayle D. [Pericarditis secondary to interferon alpha in chronic hepatitis C]. ACTA ACUST UNITED AC 2004; 28:315-6. [PMID: 15094688 DOI: 10.1016/s0399-8320(04)94929-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
40
|
Abstract
Interferon and ribavirin combination therapy for chronic hepatitis C produces a number of well-described side effects that are dominated by fatigue, influenza-like symptoms, hematologic abnormalities, and neuropsychiatric symptoms. Combination therapy with pegylated interferons (peginterferon alfa-2a and alfa-2b) yields an adverse event profile similar to standard interferon, although the frequency of certain adverse events may vary by preparation. Premature withdrawal from therapy due to adverse events was required in 10% to 14% of participants in registration trials of these agents. Most adverse events were safely and effectively managed by dose reduction using predetermined criteria. The most common indications for dose reduction were hematologic abnormalities, such as anemia and neutropenia, with the latter more frequent in peginterferon treatment arms. Recent data suggest that maintaining adherence to a prescribed treatment regimen can enhance antiviral response. Strategies to maximize adherence are being developed and, in the future, may include early identification of and therapy for depression and the selective use of hematopoietic growth factors to ameliorate hematologic abnormalities.
Collapse
Affiliation(s)
- Michael W Fried
- Division of Digestive Diseases, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| |
Collapse
|
41
|
Abstract
Interferon and ribavirin combination therapy for chronic hepatitis C produces a number of well-described side effects that are dominated by fatigue, influenza-like symptoms, hematologic abnormalities, and neuropsychiatric symptoms. Combination therapy with pegylated interferons (peginterferon alfa-2a and alfa-2b) yields an adverse event profile similar to standard interferon, although the frequency of certain adverse events may vary by preparation. Premature withdrawal from therapy due to adverse events was required in 10% to 14% of participants in registration trials of these agents. Most adverse events were safely and effectively managed by dose reduction using predetermined criteria. The most common indications for dose reduction were hematologic abnormalities, such as anemia and neutropenia, with the latter more frequent in peginterferon treatment arms. Recent data suggest that maintaining adherence to a prescribed treatment regimen can enhance antiviral response. Strategies to maximize adherence are being developed and, in the future, may include early identification of and therapy for depression and the selective use of hematopoietic growth factors to ameliorate hematologic abnormalities.
Collapse
Affiliation(s)
- Michael W Fried
- Division of Digestive Diseases, University of North Carolina, Chapel Hill, North Carolina 27599, USA.
| |
Collapse
|
42
|
Uto H, Matsuoka H, Murata M, Okamoto T, Miyata Y, Hori T, Ido A, Hirono S, Hayashi K, Tsubouchi H. A case of chronic hepatitis C developing insulin-dependent diabetes mellitus associated with various autoantibodies during interferon therapy. Diabetes Res Clin Pract 2000; 49:101-6. [PMID: 10963820 DOI: 10.1016/s0168-8227(00)00143-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We report a case of chronic hepatitis C presenting insulin-dependent diabetes mellitus (IDDM) associated with various autoantibodies including possible anti-insulin receptor antibody (AIRA) during interferon (IFN) therapy. A 57-year-old man having chronic hepatitis C virus (HCV) infection with chronic thyroiditis received IFN therapy. The thyroid function was well-controlled by administration of thyroid hormone, although thyroid autoantibodies were positive. At 15 weeks after starting IFN (reaching 530 million units of total dose), marked thirst happened, with increased fasting plasma glucose level (488 mg/dl) and decreased daily urinary C peptide immunoreactivity level (less than 4.2 microg/day). IDDM occurred with anti-nuclear antibody (ANA), anti-DNA antibody and possible AIRA, and thyroid autoantibodies titers increased, but without pancreatic islet cell antibody and anti-glutamic acid decarboxylase antibody. Administration of IFN was stopped and insulin treatment was started, but plasma glucose level was not controlled well. AIRA became negative 2 months later, however, insulin antibody (IA) was positive when tested after 18 months. Serum HCV RNA has been negative, and a normal level of serum transaminase has been observed since IFN therapy. It is likely that IFN therapy induced the immunological disturbance and resulted in occurrence of various autoantibodies and IDDM in the patient.
Collapse
Affiliation(s)
- H Uto
- Department of Internal Medicine II, Miyazaki Medical College, Kiyotake 5200, 889-1601, Miyazaki, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
The preferred treatment for patients with chronic hepatitis C, either treatment-naive, relapsers or nonresponders to IFN monotherapy, is now IFN-ribavirin combination treatment. The adverse effects of IFN are well established and familiar to hepatologists all over the world. More than 25,000 patients worldwide have been treated with combination therapy. Patients re-treated with a combination regimen are more likely to tolerate IFN better than treatment-naive patients, probably due to better case selection. The safety profile of regimens containing IFN-alpha plus ribavirin is generally consistent with the safety profile of each agent when employed in monotherapy; there is little or no synergistic toxicity. Anorexia, dyspnoea, cough, pruritus and rash are the only adverse events reported at a consistently higher frequency with combination treatment, and are usually mild to moderate in severity and rarely result in dose reductions or discontinuation. The primary cause of dose reduction for combination therapy is haemolytic anaemia, which can be managed effectively. The most common reason for discontinuation of therapy for either type of therapy is psychiatric problems, especially depression, which seems to be closely related to the duration of treatment. In patients receiving combination therapy, anaemia and depression need close monitoring, and dose modification in some cases. Strict guidelines for dose reduction and discontinuation are essential to prevent serious adverse events. Because of the teratogenic risk from ribavirin, pregnancy is contraindicated in patients or their partners during and 6 months after treatment.
Collapse
Affiliation(s)
- A Chutaputti
- Pramongkutklao Hospital, Rajathevee, Bangkok, Thailand.
| |
Collapse
|
44
|
Incidence of electrocardiographic abnormalities during treatment with human leukocyte interferon-alfa in patients with chronic hepatitis C but without preexisting cardiovascular disease. Curr Ther Res Clin Exp 1998. [DOI: 10.1016/s0011-393x(98)85027-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|