1
|
Qu HQ, Wang JF, Rosa-Campos A, Hakonarson H, Feldman AM. The Role of BAG3 Protein Interactions in Cardiomyopathies. Int J Mol Sci 2024; 25:11308. [PMID: 39457090 DOI: 10.3390/ijms252011308] [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: 09/20/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Bcl-2-associated athanogene 3 (BAG3) plays an important function in cellular protein quality control (PQC) maintaining proteome stability. Mutations in the BAG3 gene result in cardiomyopathies. Due to its roles in cardiomyopathies and the complexity of BAG3-protein interactions, it is important to understand these protein interactions given the importance of the multifunctional cochaperone BAG3 in cardiomyocytes, using an in vitro cardiomyocyte model. The experimental assay was conducted using high pressure liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) in the human AC16 cardiomyocyte cell line with BioID technology. Proteins with BAG3-interaction were identified in all the 28 hallmark gene sets enriched in idiopathic cardiomyopathies and/or ischemic disease. Among the 24 hallmark gene sets enriched in both idiopathic cardiomyopathies and ischemic disease, 15 gene sets had at least 3 proteins with BAG3-interaction. This study highlights BAG3 protein interactions, unveiling the key gene sets affected in cardiomyopathies, which help to explain the molecular mechanisms of the cardioprotective effects of BAG3. In addition, this study also highlighted the complexity of proteins with BAG3 interactions, implying unwanted effects of BAG3.
Collapse
Affiliation(s)
- Hui-Qi Qu
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ju-Fang Wang
- Department of Medicine, Division of Cardiology, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Alexandre Rosa-Campos
- Proteomics Facility, Sanford-Burnham-Presby Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Hakon Hakonarson
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Human Genetics, Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Faculty of Medicine, University of Iceland, 102 Reykjavík, Iceland
| | - Arthur M Feldman
- Department of Medicine, Division of Cardiology, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
2
|
Gurevic I, Meudec L, Mariette X, Nocturne G, McCoy SS. 0JAK Inhibitor Withdrawal Causes a Transient Proinflammatory Cascade: A Potential Mechanism for Major Adverse Cardiac Events. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.615051. [PMID: 39386576 PMCID: PMC11463359 DOI: 10.1101/2024.09.25.615051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Objective Our objective was to define the effect of JAK1/2 inhibitor (JAKinib) withdrawal on JAK/STAT biochemical response in the context of systemic rheumatic diseases. Methods We tested Type I (bind kinase active conformation) and Type II (bind kinase inactive conformation) JAKinibs in vitro using mesenchymal stromal cells and endothelial cells. We translated our findings in vivo studying NK cells from rheumatoid arthritis (RA) patients treated with Type I JAKinibs or methotrexate. Results Type I JAKinibs (ruxolitinib and baricitinib) increased phosphoJAK1 (pJAK1) and pJAK2 of IFNγ-stimulated MSCs and HUVECs in a time- and dose- dependent manner, with effect peaking after 24 hours. As expected, pSTAT1 was completely suppressed by JAKinibs. We found a marked and rapid increase of pSTATs upon discontinuation of Type I JAKinibs, that occurred to a lesser extent after Type II JAKinib withdrawal. Type I JAKinib withdrawal increased interferon and urokinase expression when compared to Type II JAKinib withdrawal. We found NK cells from RA patients taking Type I JAKinibs had a pro-inflammatory profile after JAKinib withdrawal compared to patients on methotrexate. Conclusions Type I JAKinibs paradoxically accumulate functionally defective pJAK. Upon withdrawal, the primed pJAKs are de-repressed and initiate a pSTAT signaling cascade, resulting in high interferon and urokinase. Type II JAKinibs do not cause pJAK accumulation, pSTAT cascade, and subsequent pro-inflammatory transcripts. The resultant cytokines and proteins produced from this cascade might be associated with adverse cardiac outcomes. Thus, JAKinib withdrawal is a possible mechanism contributing to the major adverse cardiac events described with JAKinib therapy.
Collapse
Affiliation(s)
- Ilya Gurevic
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Loic Meudec
- Center for Immunology of Viral Infections and Autoimmune Diseases, INSERM UMR 1184, Université Paris-Saclay, Le Kremlin-Bicêtre, Paris, France
| | - Xavier Mariette
- Université Paris-Saclay, Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Le Kremlin-Bicêtre, Paris, France
- Assistance Publique – Hôpitaux de Paris, Hôpital Bicêtre, Department of Rheumatology, Le Kremlin Bicêtre, France
| | - Gaetane Nocturne
- Université Paris-Saclay, Center for Immunology of Viral Infections and Auto-immune Diseases (IMVA), Institut pour la Santé et la Recherche Médicale (INSERM) UMR 1184, Le Kremlin-Bicêtre, Paris, France
- Assistance Publique – Hôpitaux de Paris, Hôpital Bicêtre, Department of Rheumatology, Le Kremlin Bicêtre, France
| | - Sara S. McCoy
- Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
3
|
Yan H, Wang P, Yang F, Cheng W, Chen C, Zhai B, Zhou Y. Anticancer therapy-induced adverse drug reactions in children and preventive and control measures. Front Pharmacol 2024; 15:1329220. [PMID: 38425652 PMCID: PMC10902428 DOI: 10.3389/fphar.2024.1329220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
In recent years, considerable achievements have been made in pediatric oncology with the innovation and development of antitumor drugs. However, compared to adults, children as a special group have not yet matured fully in terms of liver and kidney function. Moreover, pediatric patients are prone to more adverse drug reactions (ADRs) from the accumulation of antineoplastic drugs due to their smaller body size and larger body surface area. Chemotherapy-related ADRs have become a non-negligible factor that affects cancer remission. To date, studies on ADRs in pediatric cancer patients have emerged internationally, but few systematic summaries are available. Here, we reviewed the various systemic ADRs associated with antitumor drugs in children and adolescent patients, as well as the advances in strategies to cope with ADRs, which consisted of neurotoxicity, hematological toxicity, cardiotoxicity, ADRs of the respiratory system and gastrointestinal system and urinary system, ADRs of the skin and its adnexa, allergic reactions, and other ADRs. For clinicians and researchers, understanding the causes, symptoms, and coping strategies for ADRs caused by anticancer treatments will undoubtedly benefit more children.
Collapse
Affiliation(s)
- Hui Yan
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Penggao Wang
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Fang Yang
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Weyland Cheng
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Congcong Chen
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Bo Zhai
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Yang Zhou
- Henan Provincial Clinical Research Center for Pediatric Diseases, Henan Key Laboratory of Pediatric Genetics and Metabolic Diseases, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
- Department of Cardiothoracic Surgery, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| |
Collapse
|
4
|
Baldassarre LA, Ganatra S, Lopez-Mattei J, Yang EH, Zaha VG, Wong TC, Ayoub C, DeCara JM, Dent S, Deswal A, Ghosh AK, Henry M, Khemka A, Leja M, Rudski L, Villarraga HR, Liu JE, Barac A, Scherrer-Crosbie M. Advances in Multimodality Imaging in Cardio-Oncology: JACC State-of-the-Art Review. J Am Coll Cardiol 2022; 80:1560-1578. [PMID: 36229093 DOI: 10.1016/j.jacc.2022.08.743] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 11/07/2022]
Abstract
The population of patients with cancer is rapidly expanding, and the diagnosis and monitoring of cardiovascular complications greatly rely on imaging. Numerous advances in the field of cardio-oncology and imaging have occurred in recent years. This review presents updated and practical approaches for multimodality cardiovascular imaging in the cardio-oncology patient and provides recommendations for imaging to detect the myriad of adverse cardiovascular effects associated with antineoplastic therapy, such as cardiomyopathy, atherosclerosis, vascular toxicity, myocarditis, valve disease, and cardiac masses. Uniquely, we address the role of cardiovascular imaging in patients with pre-existing cardiomyopathy, pregnant patients, long-term survivors, and populations with limited resources. We also address future avenues of investigation and opportunities for artificial intelligence applications in cardio-oncology imaging. This review provides a uniform practical approach to cardiovascular imaging for patients with cancer.
Collapse
Affiliation(s)
- Lauren A Baldassarre
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Sarju Ganatra
- Cardio-Oncology and Cardiac MRI Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Beth Israel Lahey Health, Burlington, Massachusetts, USA
| | - Juan Lopez-Mattei
- Cardiovascular Imaging Program, Department of Cardiovascular Medicine, Lee Health, Fort Myers, Florida, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, California, USA
| | - Vlad G Zaha
- Cardio-Oncology Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Timothy C Wong
- UPMC Cardiovascular Magnetic Resonance Center, Division of Cardiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Chadi Ayoub
- Division of Cardiovascular Medicine, Mayo Clinic, Scottsdale, Arizona, USA
| | - Jeanne M DeCara
- Cardio-Oncology Program, Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Arjun K Ghosh
- Cardio-Oncology Service, Barts Heart Centre, St Bartholomew's Hospital, London, United Kingdom; Cardio-Oncology Service, University College London Hospital and Hatter Cardiovascular Institute, London, United Kingdom
| | - Mariana Henry
- Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Abhishek Khemka
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Monika Leja
- Cardio-Oncology Program, Department of Cardiovascular Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Lawrence Rudski
- Azrieli Heart Center, Department of Medicine, Jewish General Hospital, McGill University, Montréal, Québec, Canada
| | - Hector R Villarraga
- Department of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jennifer E Liu
- Cardiology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ana Barac
- Medstar Heart and Vascular Institute, Georgetown University, Washington, DC, USA; Cardiovascular Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Marielle Scherrer-Crosbie
- Division of Cardiology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
5
|
Bishai N, Nabawy WE, Fiki ME, Ibrahim M, Garem NE. Dual versus triple therapy in treatment of hepatitis C virus (HCV). Ir J Med Sci 2022:10.1007/s11845-022-03120-9. [PMID: 36040651 DOI: 10.1007/s11845-022-03120-9] [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: 03/25/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The goal of HCV treatment is eradication of the virus to prevent complications associated with the disease and decrease all-cause mortality. This work compared sustained viral response (SVR) 12 weeks after end of treatment of chronic HCV patients with different treatment regimens, namely 4 regimens. Two hundred treatment naive chronic HCV patients were selected and divided into 4 equal groups as follows: group A received pegylated interferon (peg IFN) and ribavirin (RBV); group B received peg IFN, RBV, and sofosbuvir (SOF); group C received RBV and SOF; group D received SOF, daclatasvir (DCV), and RBV. RESULTS The sustained viral response after 12 months of treatment is 57.23%, 72.09%, 64.40%, and 96.42% of patients in groups A, B, C, and D, respectively. Hence, group D regimen showed the best results. CONCLUSION SOF and DCV and RBV have the highest SVR12 and least side effects compared to other treatment regimens. Although group D patients initially had poor pretreatment investigations relative to other groups, they proved to have the highest tolerability to this regimen. Such findings hold promising line of treatment and better prognosis even for chronic HCV patients with poor liver condition.
Collapse
Affiliation(s)
- Nevine Bishai
- Department of Internal Medicine, Faculty of Medicine, Cairo University Hospitals, Kasr El Aini, Cairo, Egypt.
| | - Walid El Nabawy
- Faculty of Medicine, Beni-Sueif University Hospitals, Beni Sueif, Egypt
| | - Mohamed El Fiki
- Faculty of Medicine, Beni-Sueif University Hospitals, Beni Sueif, Egypt
| | - Mohamed Ibrahim
- Faculty of Medicine, Beni-Sueif University Hospitals, Beni Sueif, Egypt
| | - Nouman El Garem
- Faculty of Medicine, Cairo University Hospitals, Kasr El Aini, Cairo, Egypt
| |
Collapse
|
6
|
Li C, Chen Y, Zhao Y, Lung DC, Ye Z, Song W, Liu FF, Cai JP, Wong WM, Yip CCY, Chan JFW, To KKW, Sridhar S, Hung IFN, Chu H, Kok KH, Jin DY, Zhang AJ, Yuen KY. Intravenous Injection of Coronavirus Disease 2019 (COVID-19) mRNA Vaccine Can Induce Acute Myopericarditis in Mouse Model. Clin Infect Dis 2022; 74:1933-1950. [PMID: 34406358 PMCID: PMC8436386 DOI: 10.1093/cid/ciab707] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Post-vaccination myopericarditis is reported after immunization with coronavirus disease 2019 (COVID-19) messenger RNA (mRNA) vaccines. The effect of inadvertent intravenous injection of this vaccine on the heart is unknown. METHODS We compared the clinical manifestations, histopathological changes, tissue mRNA expression, and serum levels of cytokine/chemokine and troponin in Balb/c mice at different time points after intravenous (IV) or intramuscular (IM) vaccine injection with normal saline (NS) control. RESULTS Although significant weight loss and higher serum cytokine/chemokine levels were found in IM group at 1-2 days post-injection (dpi), only IV group developed histopathological changes of myopericarditis as evidenced by cardiomyocyte degeneration, apoptosis, and necrosis with adjacent inflammatory cell infiltration and calcific deposits on visceral pericardium, although evidence of coronary artery or other cardiac pathologies was absent. Serum troponin level was significantly higher in IV group. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike antigen expression by immunostaining was occasionally found in infiltrating immune cells of the heart or injection site, in cardiomyocytes and intracardiac vascular endothelial cells, but not skeletal myocytes. The histological changes of myopericarditis after the first IV-priming dose persisted for 2 weeks and were markedly aggravated by a second IM- or IV-booster dose. Cardiac tissue mRNA expression of interleukin (IL)-1β, interferon (IFN)-β, IL-6, and tumor necrosis factor (TNF)-α increased significantly from 1 dpi to 2 dpi in the IV group but not the IM group, compatible with presence of myopericarditis in the IV group. Ballooning degeneration of hepatocytes was consistently found in the IV group. All other organs appeared normal. CONCLUSIONS This study provided in vivo evidence that inadvertent intravenous injection of COVID-19 mRNA vaccines may induce myopericarditis. Brief withdrawal of syringe plunger to exclude blood aspiration may be one possible way to reduce such risk.
Collapse
Affiliation(s)
- Can Li
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yanxia Chen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Yan Zhao
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - David Christopher Lung
- Department of Pathology, Queen Elizabeth Hospital and Hong Kong Children’s Hospital, Hong Kong, Hong Kong Special Administrative Region, China
| | - Zhanhong Ye
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Wenchen Song
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Fei-Fei Liu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jian-Piao Cai
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Wan-Man Wong
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Cyril Chik-Yan Yip
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jasper Fuk-Woo Chan
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kelvin Kai-Wang To
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Siddharth Sridhar
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Ivan Fan-Ngai Hung
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Hin Chu
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kin-Hang Kok
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Dong-Yan Jin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Anna Jinxia Zhang
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- State Key Laboratory of Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
- Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, China
- Hainan Medical University-The University of Hong Kong Joint Laboratory of Tropical Infectious Diseases, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| |
Collapse
|
7
|
Makunda N, Vallabhaneni S, Lefebvre B, Fradley MG. Cardiotoxicity of Systemic Melanoma Treatments. Curr Treat Options Oncol 2022; 23:240-253. [PMID: 35192138 DOI: 10.1007/s11864-021-00924-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT Melanoma is the least common but most dangerous skin cancer, accounting for 75% of all deaths from a primary cutaneous malignancy, with incidence rates rising significantly over the last decade. Traditional treatments for melanoma including interferon and cytotoxic chemotherapy had marginal efficacy. With the advent of targeted and immunotherapies, the prognosis for patients with advanced melanoma has significantly improved including those with metastatic disease to the heart. BRAF and MEK inhibitors as well as immune checkpoint inhibitors have become front line therapy for eligible patients with metastatic melanoma and have led to long-term durable response and in some cases can be curative. Despite these oncologic advances, various treatment-limiting side effects can occur. In particular, cardiovascular toxicities can contribute to overall morbidity and mortality in these patients. Toxicities range from asymptomatic QT prolongation and mild LV dysfunction to fulminant myocarditis and potentially life-threatening arrhythmias. A multidisciplinary approach to the care of these patients which includes cardio-oncology evaluation is necessary to develop both risk mitigation and treatment strategies to ensure patients continue receiving necessary and effective melanoma treatments while minimizing long-term adverse cardiovascular effects.
Collapse
Affiliation(s)
- Neha Makunda
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Srilakshmi Vallabhaneni
- Cardio-Oncology Center of Excellence, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Benedicte Lefebvre
- Cardio-Oncology Center of Excellence, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
8
|
Rezende AGDS, Lopes EP, Batista AD, Filgueira NA, Costa WER, Felix PMDS, Markman B. Treatment of Hepatitis C with Direct-Acting Antivirals does not Induce Significant Arrhythmias. INTERNATIONAL JOURNAL OF CARDIOVASCULAR SCIENCES 2021. [DOI: 10.36660/ijcs.20200220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
9
|
Dziedzic A, Miller E, Bijak M, Przyslo L, Saluk-Bijak J. Increased Pro-Thrombotic Platelet Activity Associated with Thrombin/PAR1-Dependent Pathway Disorder in Patients with Secondary Progressive Multiple Sclerosis. Int J Mol Sci 2020; 21:ijms21207722. [PMID: 33086557 PMCID: PMC7589910 DOI: 10.3390/ijms21207722] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/20/2022] Open
Abstract
Epidemiological studies confirm the high risk of ischemic events in multiple sclerosis (MS) that are associated with increased pro-thrombotic activity of blood platelets. The most potent physiological platelet agonist is thrombin, which activates platelets via cleavage of specific protease-activated receptors (PARs). Our current study is aimed to determine the potential genetics and proteomic abnormalities of PAR1 in both platelets and megakaryocytes, which may have thromboembolic consequences in the course of MS. The obtained results were correlated with the expression level of platelet and megakaryocyte transcripts for APOA1 and A2M genes encoding atherosclerosis biomarkers: apolipoprotein A1 (ApoA1) and α-2-macroglobulin (α2M), respectively. Moreover, PAR1 functionality in MS platelets was assessed by flow cytometry, determining the level of platelet–platelet and platelet–leukocyte aggregates, platelet microparticles and surface expression of P-selectin. As a PAR1 agonist, the synthetic TRAP-6 peptide was used, which made it possible to achieve platelet activation in whole blood without triggering clotting. Comparative analyses showed an elevated level of platelet activation markers in the blood of MS patients compared to controls. The mRNA expression of gene coding α2M was upregulated, whilst ApoA1 was down-regulated, both in platelets and megakaryocytes from MS patients. Furthermore, we observed an increase in both mRNA expression and surface density of PAR1 in platelets and megakaryocytes in MS compared to controls. Both the level of platelet activation markers and PAR1 expression showed a high correlation with the expression of transcripts for APOA1 and A2M genes.
Collapse
Affiliation(s)
- Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Lukasz Przyslo
- Department of Developmental Neurology and Epileptology, Research Institute of Polish Mother’s Memorial Hospital, Rzgowska 281/289, 93-338 Lodz, Poland;
| | - Joanna Saluk-Bijak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
- Correspondence: ; Tel./Fax: +48-42-635-4336
| |
Collapse
|
10
|
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]
|
11
|
Cardiotoxic mechanisms of cancer immunotherapy - A systematic review. Int J Cardiol 2020; 323:179-187. [PMID: 32800915 DOI: 10.1016/j.ijcard.2020.08.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/26/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is a success story of translational medicine that has led to improved survival in patients with different difficult-to-treat types of cancer, such as metastasized melanoma, non-small cell lung cancer or renal cell carcinoma. These novel therapeutic agents exert their antitumor effects by activating the patients' immune system against cancer cells. Immunotherapy can be divided into active agents, such as anti-tumour vaccines or adoptive T-cell transfer, and passive immunotherapies like monoclonal antibodies, checkpoint inhibitors, cytokine therapy, bispecific T-cell engagers. After initial experimental use, broad clinical application revealed a number of important cardiovascular side effects of immunotherapeutics, which limit treatment options and decrease patients' prognosis and quality of life. With the rising rate of new immunotherapeutics at a hand, the number of patients receiving cancer immunotherapy will constantly increase, resulting in improved long-term survival rates. This review aims to summarize available cancer immunotherapies, their mechanism of action, currently known cardiovascular toxicities and their treatment. Further optimization of patient care will depend on the combined efforts by oncologists, cardiologists and cardiac surgeons to identify patients at risk and the implementation of interdisciplinary screening and treatment strategies. It is therefore crucial to familiarize heart specialists with novel cancer therapeutics and their potential adverse effects.
Collapse
|
12
|
Cirulis MM, Beesley SJ, Wilson EL, Stubben C, Olsen TD, Hirshberg EL, Smith LM, Lanspa MJ, Abraham TP, Grissom CK, Rondina MT, Brown SM. The peripheral blood transcriptome in septic cardiomyopathy: an observational, pilot study. Intensive Care Med Exp 2019; 7:57. [PMID: 31650252 PMCID: PMC6813402 DOI: 10.1186/s40635-019-0271-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/24/2019] [Indexed: 01/25/2023] Open
Abstract
Background Septic cardiomyopathy (SCM) is common in sepsis and associated with increased morbidity and mortality. Left ventricular global longitudinal strain (LV GLS), measured by speckle tracking echocardiography, allows improved identification of impaired cardiac contractility. The peripheral blood transcriptome may be an important window into SCM pathophysiology. We therefore studied the peripheral blood transcriptome and LV GLS in a prospective cohort of patients with sepsis. Results In this single-center observational pilot study, we enrolled adult patients (age > 18) with sepsis within 48 h of admission to the ICU. SCM was defined as LV GLS > − 17% based on echocardiograms performed within 72 h of admission. We enrolled 27 patients, 24 of whom had high-quality RNA results; 18 (75%) of 24 had SCM. The group was 50% female and had a median (IQR) age of 59.5 (48.5–67.0) years and admission APACHE II score of 21.0 (16.0–32.3). Forty-six percent had septic shock. After filtering for low-expression and non-coding genes, 15,418 protein coding genes were expressed and 73 had significantly different expression between patients with vs. without SCM. In patients with SCM, 43 genes were upregulated and 30 were downregulated. Pathway analysis identified enrichment in type 1 interferon signaling (adjusted p < 10−5). Conclusions In this hypothesis-generating study, SCM was associated with upregulation of genes in the type 1 interferon signaling pathway. Interferons are cytokines that stimulate the innate and adaptive immune response and are implicated in the early proinflammatory and delayed immunosuppression phases of sepsis. While type 1 interferons have not been implicated previously in SCM, interferon therapy (for viral hepatitis and Kaposi sarcoma) has been associated with reversible cardiomyopathy, perhaps suggesting a role for interferon signaling in SCM.
Collapse
Affiliation(s)
- Meghan M Cirulis
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Medicine, University of Utah, Salt Lake City, UT, USA. .,Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Shock Trauma Intensive Care Unit, 5121 South Cottonwood Street, Murray, UT, 84107 42, USA.
| | - Sarah J Beesley
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Medicine, University of Utah, Salt Lake City, UT, USA.,Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Shock Trauma Intensive Care Unit, 5121 South Cottonwood Street, Murray, UT, 84107 42, USA.,Critical Care Echocardiography Service, Intermountain Medical Center, Murray, UT, USA
| | - Emily L Wilson
- Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Shock Trauma Intensive Care Unit, 5121 South Cottonwood Street, Murray, UT, 84107 42, USA.,Critical Care Echocardiography Service, Intermountain Medical Center, Murray, UT, USA
| | - Chris Stubben
- Bioinformatics Shared Resource, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Troy D Olsen
- Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Shock Trauma Intensive Care Unit, 5121 South Cottonwood Street, Murray, UT, 84107 42, USA
| | - Eliotte L Hirshberg
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Medicine, University of Utah, Salt Lake City, UT, USA.,Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Shock Trauma Intensive Care Unit, 5121 South Cottonwood Street, Murray, UT, 84107 42, USA.,Critical Care Echocardiography Service, Intermountain Medical Center, Murray, UT, USA
| | - Lane M Smith
- Department of Emergency Medicine, Wake Forest School of Medicine, Winston Salem, NC, USA
| | - Michael J Lanspa
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Medicine, University of Utah, Salt Lake City, UT, USA.,Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Shock Trauma Intensive Care Unit, 5121 South Cottonwood Street, Murray, UT, 84107 42, USA.,Critical Care Echocardiography Service, Intermountain Medical Center, Murray, UT, USA
| | - Theodore P Abraham
- Division of Cardiology, Department of Medicine, UCSF, San Francisco, CA, USA
| | - Colin K Grissom
- Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Shock Trauma Intensive Care Unit, 5121 South Cottonwood Street, Murray, UT, 84107 42, USA.,Critical Care Echocardiography Service, Intermountain Medical Center, Murray, UT, USA
| | - Matthew T Rondina
- Department of Emergency Medicine, Wake Forest School of Medicine, Winston Salem, NC, USA.,Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | - Samuel M Brown
- Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, Department of Medicine, University of Utah, Salt Lake City, UT, USA.,Pulmonary and Critical Care Division, Department of Medicine, Intermountain Medical Center, Shock Trauma Intensive Care Unit, 5121 South Cottonwood Street, Murray, UT, 84107 42, USA.,Critical Care Echocardiography Service, Intermountain Medical Center, Murray, UT, USA
| |
Collapse
|
13
|
Heart rate variability analysis in patients with multiple sclerosis. Mult Scler Relat Disord 2018; 24:64-68. [PMID: 29957350 DOI: 10.1016/j.msard.2018.06.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 01/09/2018] [Accepted: 06/17/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Multiple sclerosis can cause cardiovascular autonomic dysfunction. It is assumed that is caused by multiple demyelinating plaques localized in the brain stem and spinal cord. Previous studies have determined this using tilt table test, heart rate responses to Valsalva maneuver and deep breathing and heart rate variability analysis with 24 h Holter monitoring. However there is not a consensus regarding the presence of the relationship between autonomic dysfunction and severity of multiple sclerosis, type of multiple sclerosis and expanded disability status scale. The aim of the study is comparison of heart rate variability between recently diagnosed patients with relapsing-remitting multiple sclerosis and healthy controls by using 24 h Holter monitoring. Also we intended to investigate relationship between Expanded Disability Status Scale score, Multiple Sclerosis Functional Composite scores and cranial and spinal magnetic resonance imaging findings and hearth rate variability. METHOD Fifty-one patients with newly diagnosed relapsing-remitting multiple sclerosis and 44 age- and sex-matched healthy controls were compared in this study. Patients with multiple sclerosis, who were already under immunomodulatory or immunosuppressive treatment, were excluded from the study. Echocardiography and hearth rate variability analysis using 24 h period Holter monitoring were performed in all of the subjects. Echocardiography was used to detect the presence of cardiac pathology. One multiple sclerosis patient with right ventricular dilatation and mobile intratrial septum was excluded from the study. All the patients underwent cranial and cervical spinal magnetic resonance imaging to determine the relationship between autonomic abnormalities and magnetic resonance imaging. RESULTS Our results showed that hearth rate variability values were significantly lower in patients with multiple sclerosis when compared with healthy controls: SDNN index (the mean of all the 5 min standard deviations of normal RR intervals during the 24 h period) (59.80 ± 17.33 vs. 67.20 ± 21.28, p = 0,044), the root-mean-square successive difference (rMSSD) (34.40 ± 17.50 vs. 38.25 ± 12.95, p = 0,042), spectral hearth rat variability total power (3738.84 ± 2085.51 vs. 4427.44 ± 1965.71, p = 0,037), spectral hearth rate variability low frequency (852.03 ± 450.54 vs. 1011.75 ± 370.06, p = 0,018). Ten patients (20%) had brainstem lesion, 25 patients (50%) had cervical lesions and 10 patients (20%) had thoracic spinal lesions on magnetic resonance imaging. There was no significant relationship between location of the lesions and heart rate variability analyses. Also there was no significant relationship between hearth rate variability values and Expanded Disability Status Scale score, Multiple Sclerosis Functional Composite scores or number of multiple sclerosis attack (p > 0,05). CONCLUSION These findings reveals that our study population with multiple sclerosis had decreased heart rate variability compared to healthy controls. This was reflected by dysfunction of both parasympathetic and sympathetic parameters of hearth rate variability analysis. However, there is no significant relationship between hearth rate variability analysis and the findings on cranial, cervical, thoracic spinal magnetic resonance imaging findings, number of attack, Expanded Disability Status Scale score or Multiple Sclerosis Functional Composite scores in patients with multiple sclerosis.
Collapse
|
14
|
Hamlett ED, Ledreux A, Potter H, Chial HJ, Patterson D, Espinosa JM, Bettcher BM, Granholm AC. Exosomal biomarkers in Down syndrome and Alzheimer's disease. Free Radic Biol Med 2018; 114:110-121. [PMID: 28882786 PMCID: PMC6135098 DOI: 10.1016/j.freeradbiomed.2017.08.028] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/30/2017] [Accepted: 08/31/2017] [Indexed: 02/07/2023]
Abstract
Every person with Down syndrome (DS) has the characteristic features of Alzheimer's disease (AD) neuropathology in their brain by the age of forty, and most go on to develop AD dementia. Since people with DS show highly variable levels of baseline function, it is often difficult to identify early signs of dementia in this population. The discovery of blood biomarkers predictive of dementia onset and/or progression in DS is critical for developing effective clinical diagnostics. Our recent studies show that neuron-derived exosomes, which are small extracellular vesicles secreted by most cells in the body, contain elevated levels of amyloid-beta peptides and phosphorylated-Tau that could indicate a preclinical AD phase in people with DS starting in childhood. We also found that the relative levels of these biomarkers were altered following dementia onset. Exosome release and signaling are dependent on cellular redox homeostasis as well as on inflammatory processes, and exosomes may be involved in the immune response, suggesting a dual role as both triggers of inflammation in the brain and propagators of inflammatory signals between brain regions. Based on recently reported connections between inflammatory processes and exosome release, the elevated neuroinflammatory state observed in people with DS may affect exosomal AD biomarkers. Herein, we discuss findings from studies of people with DS, people with DS and AD (DS-AD), and mouse models of DS showing new connections between neuroinflammatory pathways, oxidative stress, exosomes, and exosome-mediated signaling, which may inform future AD diagnostics, preventions, and treatments in the DS population as well as in the general population.
Collapse
Affiliation(s)
- Eric D Hamlett
- Knoebel Institute for Healthy Aging and the Department of Biological Sciences, University of Denver, Denver, CO, USA; Medical University of South Carolina, Charleston, SC, USA
| | - Aurélie Ledreux
- Knoebel Institute for Healthy Aging and the Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Huntington Potter
- Rocky Mountain Alzheimer's Disease Center, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Neurology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Heidi J Chial
- Rocky Mountain Alzheimer's Disease Center, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Neurology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - David Patterson
- Knoebel Institute for Healthy Aging and the Department of Biological Sciences, University of Denver, Denver, CO, USA
| | - Joaquin M Espinosa
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Pharmacology, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Brianne M Bettcher
- Rocky Mountain Alzheimer's Disease Center, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Neurology, University of Colorado Anschutz Medical Campus, Denver, CO, USA; Department of Neurosurgery, University of Colorado Anschutz Medical Campus, Denver, CO, USA
| | - Ann-Charlotte Granholm
- Knoebel Institute for Healthy Aging and the Department of Biological Sciences, University of Denver, Denver, CO, USA; Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
15
|
Wang DY, Okoye GD, Neilan TG, Johnson DB, Moslehi JJ. Cardiovascular Toxicities Associated with Cancer Immunotherapies. Curr Cardiol Rep 2017; 19:21. [PMID: 28220466 PMCID: PMC10176498 DOI: 10.1007/s11886-017-0835-0] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW We review the cardiovascular toxicities associated with cancer immune therapies and discuss the cardiac manifestations, potential mechanisms, and management strategies. RECENT FINDINGS The recent advances in cancer immune therapy with immune checkpoint inhibitors and adoptive cell transfer have improved clinical outcomes in numerous cancers. The rising use of cancer immune therapy will lead to a higher incidence in immune-related adverse events. Recent studies have highlighted several reports of severe cases of acute cardiotoxic events with immune therapy including fulminant myocarditis. We believe that immune-mediated myocarditis is a driving mechanism behind these cardiovascular toxicities and requires vigilant screening and prompt management with corticosteroids and immune-modulating drugs, especially with combination immune therapies. While the incidence of serious cardiovascular toxicities with immune therapy appears low, these can be life-threatening especially when manifesting as acute immune-mediated myocarditis. Further collaborative studies are needed to effectively identify, characterize, and manage these events.
Collapse
Affiliation(s)
- Daniel Y Wang
- Divisions of Oncology, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, 777 Preston Research Bldg., Nashville, TN, 37232, USA
| | - Gosife Donald Okoye
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Medicine, Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas G Neilan
- Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Medicine, Cardio-Oncology Program, Massachusetts General Hospital, Boston, MA, USA
| | - Douglas B Johnson
- Divisions of Oncology, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, 777 Preston Research Bldg., Nashville, TN, 37232, USA.
| | - Javid J Moslehi
- Divisions of Oncology, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, 777 Preston Research Bldg., Nashville, TN, 37232, USA.,Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Medicine, Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
16
|
Klee NS, McCarthy CG, Martinez-Quinones P, Webb RC. Out of the frying pan and into the fire: damage-associated molecular patterns and cardiovascular toxicity following cancer therapy. Ther Adv Cardiovasc Dis 2017; 11:297-317. [PMID: 28911261 PMCID: PMC5933669 DOI: 10.1177/1753944717729141] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 08/09/2017] [Indexed: 12/18/2022] Open
Abstract
Cardio-oncology is a new and rapidly expanding field that merges cancer and cardiovascular disease. Cardiovascular disease is an omnipresent side effect of cancer therapy; in fact, it is the second leading cause of death in cancer survivors after recurrent cancer. It has been well documented that many cancer chemotherapeutic agents cause cardiovascular toxicity. Nonetheless, the underlying cause of cancer therapy-induced cardiovascular toxicity is largely unknown. In this review, we discuss the potential role of damage-associated molecular patterns (DAMPs) as an underlying contributor to cancer therapy-induced cardiovascular toxicity. With an increasing number of cancer patients, as well as extended life expectancy, understanding the mechanisms underlying cancer therapy-induced cardiovascular disease is of the utmost importance to ensure that cancer is the only disease burden that cancer survivors have to endure.
Collapse
Affiliation(s)
- Nicole S. Klee
- Department of Physiology, Medical College of Georgia at Augusta University, 1120 15 Street, Augusta, GA 30912, USA
| | - Cameron G. McCarthy
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Patricia Martinez-Quinones
- Departments of Physiology and Surgery, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - R. Clinton Webb
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
17
|
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
|
18
|
Biomy R, Abdelshafy M, Abdelmonem A, Abu-Elenin H, Ghaly G. Effect of Chronic Hepatitis C Virus Treatment by Combination Therapy on Cardiovascular System. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2017; 11:1179546817713204. [PMID: 28804248 PMCID: PMC5484549 DOI: 10.1177/1179546817713204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2016] [Accepted: 04/16/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND The prevalence of hepatitis C virus (HCV) in Egypt is quite high, and the combined oral direct-acting antiviral agents (DAAs) may have impressive results. OBJECTIVE To assess the cardiovascular effects of DAAs in patients with HCV. METHODS A total of 170 patients with HCV were divided into 2 groups: first group (100 patients) received triple combination therapy (pegylated interferon alfa, sofosbuvir, and ribavirin, whereas the second group (70 patients) received dual combination therapy (sofosbuvir and simeprevir). Group 1 patients were followed up for 1 year more than 3 visits, whereas group 2 patients were followed up for 6 months more than 2 visits; and the end point of the study was the development of a major cardiovascular event (eg, congestive heart failure, echocardiographic evidence of left ventricular dysfunction, occurrence of significant arrhythmias, or acute coronary syndrome). The following parameters were accomplished: medical history and clinical examination, electrocardiogram, echo-Doppler study, and laboratory investigations. RESULTS No significant differences were found between the 2 study groups regarding demographic criteria. None of the both group patients had developed any major cardiac event. No significant changes were observed regarding ST-T wave abnormalities, arrhythmias, or QT interval. None of the both group patients developed echocardiographic regional wall motion abnormalities at baseline or at study end. Systolic function parameters showed minute nonsignificant changes over study visits. Diastolic function parameters showed nonsignificant changes between baseline and 6-month and 12-month visits. CONCLUSIONS The DAAs used in combination regimen with interferon or used orally in combination do not significantly affect the cardio-vascular system.
Collapse
Affiliation(s)
- Reda Biomy
- Faculty of Medicine, Kafrelsheikh University, Kafr El-Sheikh, Egypt
| | | | | | | | - George Ghaly
- Al-Sahel Teaching Hospital, Cardiology Department Cairo, Egypt
| |
Collapse
|
19
|
Abdel-Wahab N, Alshawa A, Suarez-Almazor ME. Adverse Events in Cancer Immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 995:155-174. [PMID: 28321817 DOI: 10.1007/978-3-319-53156-4_8] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cancer immunotherapy has resulted in durable responses in patients with metastatic disease, unseen with traditional chemotherapy. Several therapies have been approved by the Food and Drug Administration for the treatment of various cancers, including: immune checkpoint inhibitors, cytokines - interleukin 2 (IL-2) and interferon alpha (IFN), and the cancer vaccine sipuleucel-T. These therapies upregulate the immune system to enhance antitumor responses. As a consequence, they can cause inflammatory and immune-related adverse events that can affect one or more organs, can be serious, and on occasion lifethreatening. The management of these adverse events is complex, and requires a multidisciplinary approach involving not only oncologists, but also other internal medicine specialists, to ensure prompt diagnosis and optimal management of these complications.
Collapse
Affiliation(s)
- Noha Abdel-Wahab
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
- Rheumatology and Rehabilitation Department, Assiut University Hospitals, Assiut, 71526, Egypt
| | - Anas Alshawa
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria E Suarez-Almazor
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA.
| |
Collapse
|
20
|
Page RL, O'Bryant CL, Cheng D, Dow TJ, Ky B, Stein CM, Spencer AP, Trupp RJ, Lindenfeld J. Drugs That May Cause or Exacerbate Heart Failure: A Scientific Statement From the American Heart Association. Circulation 2016; 134:e32-69. [PMID: 27400984 DOI: 10.1161/cir.0000000000000426] [Citation(s) in RCA: 268] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Heart failure is a common, costly, and debilitating syndrome that is associated with a highly complex drug regimen, a large number of comorbidities, and a large and often disparate number of healthcare providers. All of these factors conspire to increase the risk of heart failure exacerbation by direct myocardial toxicity, drug-drug interactions, or both. This scientific statement is designed to serve as a comprehensive and accessible source of drugs that may cause or exacerbate heart failure to assist healthcare providers in improving the quality of care for these patients.
Collapse
|
21
|
|
22
|
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: 0.3] [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
|
23
|
Types and predictors of interferon/ribavirin induced cardiac complications in the Egyptian patients with chronic hepatitis C virus. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jicc.2016.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
24
|
Chazova IE, Oshchepkova EV, Kantorova AY. [Comorbidity of cardiovascular diseases and cancers: Problems in the diagnosis of cardiotoxic effects of chemo- and radiation therapy]. TERAPEVT ARKH 2015; 87:4-10. [PMID: 26591546 DOI: 10.17116/terarkh20158794-10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cardiovascular diseases (CVD) and cancers are the leaders in their prevalence and the major causes of death in economically developed countries, determining their high sociomedical significance in society. Improvement of methods for the early diagnosis and treatment of cancers has contributed to increases in relapse-free survival and life expectancy in these patients. At the same time, the new problems have emerged particularly in the development of various cardiovascular events/diseases when treating cancer, which may predict worse prognosis in patients and be an independent cause of death. To search for new markers of cardiotoxicity at early stages and to develop effective methods for the prevention and personalized treatment of cancer and CVD are the problems that can be solved only by joint efforts of cardiologists and oncologists.
Collapse
Affiliation(s)
- I E Chazova
- Russian Cardiology Research-and-Production Complex, Ministry of Health of Russia, Moscow
| | - E V Oshchepkova
- Russian Cardiology Research-and-Production Complex, Ministry of Health of Russia, Moscow
| | - A Yu Kantorova
- Russian Cardiology Research-and-Production Complex, Ministry of Health of Russia, Moscow
| |
Collapse
|
25
|
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: 1.2] [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
|
26
|
Fung J. Era of direct acting antivirals in chronic hepatitis C: Who will benefit? World J Hepatol 2015; 7:2543-2550. [PMID: 26523206 PMCID: PMC4621468 DOI: 10.4254/wjh.v7.i24.2543] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 09/07/2015] [Accepted: 10/13/2015] [Indexed: 02/06/2023] Open
Abstract
In the era of highly effective direct acting antiviral (DAA) drugs for the treatment of chronic hepatitis C (CHC) infection, where eradication is almost ensured with minimal side effects, all hepatitis C carriers should benefit theoretically. In the real world setting however, only a small proportion will benefit at this time point due to the multiple barriers to accessing therapy. Given that universal treatment is unlikely, treatment with DAAs will likely be restricted to those with the highest health benefits, and for those who can afford the high expense of a treatment course. Those with the highest unmet needs include those who have failed previous interferon-based therapy or who are interferon-ineligible with evidence of active disease, those with advance liver disease, and those with recurrence of hepatitis C after liver transplantation. In the future, the focus should be on increasing access to treatment for those infected with CHC.
Collapse
|
27
|
Tortorella G, Piccin A, Tieghi A, Marcheselli L, Steurer M, Gastl G, Codeluppi K, Fama A, Santoro U, Birtolo C, Gugliotta G, Cortelazzo S, Gugliotta L. Anagrelide treatment and cardiovascular monitoring in essential thrombocythemia. A prospective observational study. Leuk Res 2015; 39:592-8. [PMID: 25850727 DOI: 10.1016/j.leukres.2015.03.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 10/23/2022]
Abstract
In this prospective observational single-center study, 55 patients with essential thrombocythemia who were candidates for second line treatment with anagrelide (ANA) received a preliminary cardiovascular (CV) clinical, instrumental and biochemical evaluation (CV history and symptoms, CV risk factors, blood pressure, heart rate, ECG and ECHO-cardio parameters, Troponin I, NT-proBNP). After this in-depth CV screening, 54 out of 55 patients were deemed to be fit for ANA treatment. Thirty-eight of the 55 patients received ANA treatment for a median of 36 months (range 3-48), and were monitored using the same CV evaluation. Fourteen of these 38 patients manifested CV adverse events (10 palpitation, 4 edema, 2 arterial hypertension, 2 acute myocardial infarction) that were not predicted by the in-depth CV evaluation, and that led to ANA withdrawal in only one case (non-cardiac refractory edema). In conclusion, the planned in-depth CV evaluation did not appear to be necessary in ET patients to evaluate their suitability for ANA treatment, and, moreover, was not able to predict the occurrence of CV adverse events during ANA treatment. Nevertheless, the CV adverse events (mostly palpitations and edema) were easily managed by the hematologists, and required the cardiologist involvement in very few selected cases.
Collapse
Affiliation(s)
- Giovanni Tortorella
- Cardiology Unit, Azienda Ospedaliera Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - Andrea Piccin
- Hematology Department, San Maurizio Regional Hospital, Bolzano, South Tyrol, Italy
| | - Alessia Tieghi
- Hematology Department, Azienda Ospedaliera Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - Luigi Marcheselli
- Clinical and Public Health Medicine Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Michael Steurer
- Hematology and Oncology Department, Medical University of Innsbruck, Innsbruck, Austria
| | - Günther Gastl
- Hematology and Oncology Department, Medical University of Innsbruck, Innsbruck, Austria
| | - Katia Codeluppi
- Hematology Department, Azienda Ospedaliera Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - Angelo Fama
- Hematology Department, Azienda Ospedaliera Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - Umberto Santoro
- Statistics Department, University of Bologna, Bologna, Italy
| | - Chiara Birtolo
- Internal Medicine Department, S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Gabriele Gugliotta
- Hematology Institute "L. e A. Seragnoli", S. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Sergio Cortelazzo
- Hemato-Oncology Department, Institute Humanitas Gavazzeni, Bergamo, Italy
| | - Luigi Gugliotta
- Hematology Institute "L. e A. Seragnoli", S. Orsola-Malpighi University Hospital, Bologna, Italy.
| | | |
Collapse
|
28
|
Tamargo J, Caballero R, Delpón E. Cancer Chemotherapy and Cardiac Arrhythmias: A Review. Drug Saf 2015; 38:129-52. [DOI: 10.1007/s40264-014-0258-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
29
|
Ando M, Takahashi Y, Nishikawa M, Takakura Y. [Control of spatiotemporal distribution of interferon γ by genetically fusing functional peptides]. YAKUGAKU ZASSHI 2014. [PMID: 23208047 DOI: 10.1248/yakushi.12-00235-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Type II interferon (IFNγ) is a representative Th1 cytokine and it possesses a variety of functions, including immune regulation, antiviral and antitumor activity. Because of its multifunctional nature, IFNγ is expected to be applied to the treatment of autoimmune diseases, cancer and viral infection. Although IFNγ has therapeutic potential for such diseases, the clinical use of IFNγ has been limited due to its short in vivo half-life and serious adverse effects. In contrast, gene delivery of IFNγ is an alternative approach to increasing the retention time of IFNγ. To extend transgene expression after plasmid DNA (pDNA) gene transfer, we designed and developed pDNA with varying numbers of CpG motifs. CpG-reduced pDNA resulted in more durable transgene expression than its CpG replete counterpart in mice. Comparison of the effect of promoter/enhancer elements on transgene expression showed that ROSA26 promoter-mediated IFNγ expression was safe because of the lack of an initial surge after hydrodynamic gene transfer. We also designed an IFNγ-mouse serum albumin (MSA) fusion protein, IFNγ-MSA. Gene transfer of this fusion protein resulted in a sustained concentration of IFNγ fusion protein in mouse serum, and inhibited tumor metastasis in mice. These results provide experimental evidence that IFNγ gene therapy can be a useful treatment for a variety of diseases.
Collapse
Affiliation(s)
- Mitsuru Ando
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | |
Collapse
|
30
|
Hakim FA, Singh S, Pandit A, Alegria JR, Camoriano J, Stanton ML, Mookadam F. Interferon-α and pericardial injury: a case report and literature review. HEART ASIA 2014; 6:48-53. [PMID: 27326167 DOI: 10.1136/heartasia-2013-010488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/19/2014] [Accepted: 03/14/2014] [Indexed: 11/03/2022]
Abstract
Interferon- α (IFN-α) alone or in combination with other chemotherapeutic agents has been used in the management of many malignant and non-malignant conditions. Pericarditis with or without pericardial effusion has been reported with IFN-α therapy, and available literature is limited to case reports. Pericardial constriction after interferon use has not been described in the published literature to date. We performed a systematic review of literature to address the demographic features, clinical presentation, diagnosis, treatment and outcome of interferon-related pericardial injury.
Collapse
Affiliation(s)
- Fayaz A Hakim
- Department of Cardiovascular Diseases, Scottsdale, Arizona , USA
| | | | - Anil Pandit
- Department of Cardiovascular Diseases, Scottsdale, Arizona , USA
| | - Jorge R Alegria
- Department of Cardiovascular Diseases, Scottsdale, Arizona , USA
| | | | - Melissa L Stanton
- Laboratory Medicine and pathology , Mayo Clinic , Scottsdale, Arizona , USA
| | - Farouk Mookadam
- Department of Cardiovascular Diseases, Scottsdale, Arizona , USA
| |
Collapse
|
31
|
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; 18:110-3. [PMID: 24120833 PMCID: PMC9425206 DOI: 10.1016/j.bjid.2013.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 05/23/2013] [Indexed: 01/20/2023] Open
|
32
|
|
33
|
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: 32] [Impact Index Per Article: 2.9] [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
|
34
|
Christian JB, Finkle JK, Ky B, Douglas PS, Gutstein DE, Hockings PD, Lainee P, Lenihan DJ, Mason JW, Sager PT, Todaro TG, Hicks KA, Kane RC, Ko HS, Lindenfeld J, Michelson EL, Milligan J, Munley JY, Raichlen JS, Shahlaee A, Strnadova C, Ye B, Turner JR. Cardiac imaging approaches to evaluate drug-induced myocardial dysfunction. Am Heart J 2012. [PMID: 23194484 DOI: 10.1016/j.ahj.2012.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The ability to make informed benefit-risk assessments for potentially cardiotoxic new compounds is of considerable interest and importance at the public health, drug development, and individual patient levels. Cardiac imaging approaches in the evaluation of drug-induced myocardial dysfunction will likely play an increasing role. However, the optimal choice of myocardial imaging modality and the recommended frequency of monitoring are undefined. These decisions are complicated by the array of imaging techniques, which have varying sensitivities, specificities, availabilities, local expertise, safety, and costs, and by the variable time-course of tissue damage, functional myocardial depression, or recovery of function. This White Paper summarizes scientific discussions of members of the Cardiac Safety Research Consortium on the main factors to consider when selecting nonclinical and clinical cardiac function imaging techniques in drug development. We focus on 3 commonly used imaging modalities in the evaluation of cardiac function: echocardiography, magnetic resonance imaging, and radionuclide (nuclear) imaging and highlight areas for future research.
Collapse
|
35
|
Almawardy R, Elhammady W, Mousa N, Abotaleb S. Is combination therapy for chronic hepatitis C toxic for cardiac function? HEPATITIS MONTHLY 2012; 12:e6254. [PMID: 23105944 PMCID: PMC3477652 DOI: 10.5812/hepatmon.6254] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Revised: 06/14/2012] [Accepted: 07/27/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Many types of cardiovascular complications such as; cardiac arrhythmias, impaired cardiac function, myocardial ischemia and decreased left ventricular function, have been attributed to interferon therapy. OBJECTIVES The aim of this study was to evaluate the effects of combination therapy pegylated interferon and ribavirin on left ventricular systolic and diastolic functions in patients with a chronic hepatitis C infection. PATIENTS AND METHODS A total of 120 patients, eligible for hepatitis C virus (HCV) treatment with pegylated interferon and ribavirin, were included in this study. All patients underwent a full cardiovascular baseline examination including; detailed medical history, thorough clinical examination, 12 lead electrocardiogram (ECG), and echocardiography. A cardiac evaluation was performed at the beginning and six months after starting combination therapy. RESULTS No significant changes regarding cardiac symptoms including; shortness of breath, cough, palpitations, chest pain and hypertension, were found during or six months after starting the combined therapy. ECG findings showed statistically non-significant decreases in the QT interval, while corrected QT showed statistically non-significant increases six months after beginning combined therapy, when compared to their values before treatment. Also with regard to the echocardiography findings, there was no statistically significant difference found between any of the echocardiography parameters six months after starting combined therapy compared to their values before treatment. CONCLUSIONS The results of our study suggest that, combination therapy does not cause a significant deterioration in cardiac function in patients with a chronic hepatitis C infection, and it may be used safely in patients without cardiac disease.
Collapse
Affiliation(s)
| | | | - Nasser Mousa
- Tropical Department, Mansoura University, Mansoura, Egypt
- Corresponding author: Nasser Mousa, Tropical Medicine Department, Mansoura University, Mansoura, Egypt.Tel.: +20-573606440, Fax: E-mail:
| | | |
Collapse
|
36
|
Gormley RH, Kovarik CL. Human papillomavirus-related genital disease in the immunocompromised host: Part II. J Am Acad Dermatol 2012; 66:883.e1-17; quiz 899-900. [PMID: 22583721 DOI: 10.1016/j.jaad.2010.12.049] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 12/19/2010] [Indexed: 12/17/2022]
Abstract
Human papillomavirus is responsible for common condyloma acuminata and a number of premalignant and malignant anogenital lesions. The immunocompromised population is at particular risk because of a higher incidence of malignant transformation. Lesions in this population may prove refractory to standard treatment. This is part II of a two-part review that will discuss the treatment of condyloma acuminata and vaginal, vulvar, penile, and anal intraepithelial neoplasias. This article will provide an updated review of available treatments, with a focus on recent advances and the challenges faced in successfully treating human papillomavirus lesions in the immunocompromised host.
Collapse
Affiliation(s)
- Rachel H Gormley
- Department of Dermatology, Division of Infectious Diseases, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
37
|
Garib JR, Garcia GF, Teixeira R, Lima e Silva FDC. Dyspnoea in patients with chronic hepatitis C treated with pegylated interferon and ribavirin. ACTA ACUST UNITED AC 2011; 43:625-31. [DOI: 10.3109/00365548.2011.574150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
|
38
|
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
|
39
|
Rauw J, Ahmed S, Petrella T. Pericardial effusion and tamponade following interferon alpha treatment for locally advanced melanoma. Med Oncol 2011; 29:1304-7. [DOI: 10.1007/s12032-011-9935-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Accepted: 03/28/2011] [Indexed: 11/28/2022]
|
40
|
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
|
41
|
[Complete atrio-ventricular block: a rare complication of interferon alpha therapy]. Presse Med 2011; 40:316-8. [PMID: 21242049 DOI: 10.1016/j.lpm.2010.10.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Revised: 10/09/2010] [Accepted: 10/15/2010] [Indexed: 11/21/2022] Open
|
42
|
Hong RA, Iimura T, Sumida KN, Eager RM. Cardio-oncology/onco-cardiology. Clin Cardiol 2010; 33:733-7. [PMID: 21184556 PMCID: PMC6653579 DOI: 10.1002/clc.20823] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Accepted: 06/24/2010] [Indexed: 02/06/2023] Open
Abstract
An understanding of onco-cardiology or cardio-oncology is critical for the effective care of cancer patients. Virtually all antineoplastic agents are associated with cardiotoxicity, which can be divided into 5 categories: direct cytotoxic effects of chemotherapy and associated cardiac systolic dysfunction, cardiac ischemia, arrhythmias, pericarditis, and chemotherapy-induced repolarization abnormalities. Radiation therapy can also lead to coronary artery disease and fibrotic changes to the valves, pericardium, and myocardium. All patients being considered for chemotherapy, especially those who have prior cardiac history, should undergo detailed cardiovascular evaluation to optimize the treatment. Serial assessment of left ventricular systolic function and cardiac biomarkers might also be considered in selected patient populations. Cardiotoxic effects of chemotherapy might be decreased by the concurrent use of angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, or beta-blockers. Antiplatelet or anticoagulation therapy might be considered in patients with a potential hypercoagulable state associated with chemotherapy or cancer. Open dialogue between both cardiologists and oncologists will be required for optimal patient care.
Collapse
Affiliation(s)
- Robert A. Hong
- Internal Medicine Residency Program, The Queen's Medical Center, Honolulu, Hawaii
| | - Takeshi Iimura
- Internal Medicine Residency Program, The Queen's Medical Center, Honolulu, Hawaii
| | - Kenneth N. Sumida
- Internal Medicine Residency Program, The Queen's Medical Center, Honolulu, Hawaii
| | - Robert M. Eager
- Internal Medicine Residency Program, The Queen's Medical Center, Honolulu, Hawaii
| |
Collapse
|
43
|
Schutz FAB, Je Y, Azzi GR, Nguyen PL, Choueiri TK. Bevacizumab increases the risk of arterial ischemia: a large study in cancer patients with a focus on different subgroup outcomes. Ann Oncol 2010; 22:1404-1412. [PMID: 21115602 DOI: 10.1093/annonc/mdq587] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Bevacizumab, a humanized monoclonal antibody targeting the vascular endothelial growth factor, is a therapeutic agent used in a variety of neoplasms. We did a meta-analysis of randomized controlled trials to fully characterize the arterial thromboembolic events (ATEs) risk with bevacizumab in certain patients' subgroups. MATERIALS AND METHODS We carried out a literature search on Medline for randomized trial reported from January 1966 to December 2009. Abstracts presented at the American Society of Clinical Oncology held between 2004 and 2009 were also searched for relevant clinical trials. Summary incidence, relative risks (RRs) and 95% confidence intervals (CIs) were calculated using random-effects or fixed-effects models based on the heterogeneity of included studies. RESULTS A total of 13,026 patients from 20 randomized trials were included in the meta-analysis. Overall RR for ATE with bevacizumab-based therapy versus controls was 1.46 (95% CI 1.11-1.93, P = 0.007). On subgroup analysis, no significant risk differences were found based on the type of malignancy, type of clinical trial (phase II or III trials), type of publication (full papers versus presentations), high- versus low-dose bevacizumab and early versus advanced disease trials. When stratified by concomitant therapies, we found that gemcitabine-based regimens had a significant lower ATE risk compared with non-gemcitabine regimens (P = 0.01). CONCLUSIONS Bevacizumab treatment is associated with a significant increase in the risk of arterial thrombosis. Our results seem to be generalizable to the vast majority of patients receiving bevacizumab in multiple settings.
Collapse
Affiliation(s)
- F A B Schutz
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | - Y Je
- Department of Nutrition, Harvard School of Public Health, Boston, USA
| | - G R Azzi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | - P L Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School
| | - T K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School.
| |
Collapse
|
44
|
Higashiyama M, Hokari R, Kurihara C, Ueda T, Nakamura M, Komoto S, Okada Y, Watanabe C, Kawaguchi A, Nagao S, Miura S. Interferon-α increases monocyte migration via platelet-monocyte interaction in murine intestinal microvessels. Clin Exp Immunol 2010; 162:156-62. [PMID: 20659125 DOI: 10.1111/j.1365-2249.2010.04222.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The aim of this study was to investigate the effect of interferon (IFN)-α on recruitment of platelets and monocytes within the murine small intestinal venular endothelium. Monocytes were isolated from bone marrow of C57B6 mice. Platelets were collected from murine blood. Rolling and adhesion to submucosal microvessels in the small intestine were examined under an intravital fluorescence microscope after injection of fluorescein-labelled monocytes or platelets. In some mice, IFN-α (5×10(5) U/kg) was administered intraperitoneally. After treatment with an antibody against P-selectin, changes in monocyte and platelet migration were also investigated. Changes in monocyte migration under the condition of thrombocytopenia were also investigated. Platelets and monocytes interacted with murine intestinal microvessels, although only few platelets and monocytes showed migration behaviour. Intraperitoneal injection of IFN-α enhanced the migration of both platelets and monocytes in the intestinal microvessels. Pretreatment with anti-P-selectin attenuated the increase in migration of platelets and monocytes induced by administration of IFN-α. Thrombocytopenia decreased the rolling ratio of monocytes, suggesting that the effect of IFN-α on migration was P-selectin-dependent, derived from both the endothelium of microvessels and platelets. The results of this study suggest that IFN-α acts as a potent proinflammatory agent via its stimulatory effect on the endothelium-platelet-monocyte interaction in intestinal microvessels by a P-selectin-dependent mechanism.
Collapse
Affiliation(s)
- M Higashiyama
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
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: 0.6] [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
|
46
|
Pagnoux C, Guillevin L. Churg-Strauss syndrome: evidence for disease subtypes? Curr Opin Rheumatol 2010; 22:21-8. [PMID: 19851111 DOI: 10.1097/bor.0b013e328333390b] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Churg-Strauss syndrome (CSS) is a rare systemic small-vessel necrotizing vasculitis. Its main clinical characteristics, some potentially life-threatening, are now well known, as are its usual successive phases, from allergic rhinitis to asthma, and finally vasculitis. Conversely, physiopathogenetic mechanisms are not completely elucidated and clearly multiple, thereby suggesting the existence of different disease subtypes. RECENT FINDINGS Almost 40% of CSS patients have circulating antineutrophil cytoplasm autoantibodies (ANCAs), mostly directed against myeloperoxidase. ANCA-positive patients suffer more frequently from renal disease, peripheral nervous system involvement and/or alveolar hemorrhage, whereas frequent cardiac involvement, lung infiltrates and/or systemic manifestations are more common in those who are ANCA-negative. However, their respective global outcomes do not clearly differ. Patients might also be categorized according to other, more subtle clinical, radiological and/or biological parameters, for example, cardiac magnetic resonance imaging abnormalities or genetic background. SUMMARY Because of its practical and therapeutic repercussions, the priority remains the prompt, relatively easy identification of the most severely affected patients at CSS diagnosis, before searching for and trying to classify subsets. Large, collaborative studies are needed to determine whether other subgroups might be associated with outcomes and warrant different, and possibly new, therapeutic strategies.
Collapse
Affiliation(s)
- Christian Pagnoux
- Department of Internal Medicine, French Vasculitis Study Group, National Referral Center for Necrotizing Vasculitides and Systemic Scleroderma, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Université Paris-Descartes, Paris, France.
| | | |
Collapse
|
47
|
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
|
48
|
|
49
|
Gaillard N, Mania A, Brunel H, Blanc F, Arquizan C. Interferon-Alfa- and Erythropoetin-Associated Cerebral Vasoconstriction. Cephalalgia 2009; 29:1340-3. [DOI: 10.1111/j.1468-2982.2009.01875.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | - H Brunel
- Service de Neuroradiologie, Hôpital Universitaire Gui de Chauliac
| | - F Blanc
- Service de Médecine Interne et d' Hépatologie, Hôpital Universitaire Saint Eloi, Montpellier, France
| | | |
Collapse
|
50
|
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: 1.0] [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
|