1
|
Li C, Yan W, Gao M, Zhang Z, Zhao L, Ma J, Li X, Gao Y, Zhang D, Gao S. Knowledge, attitudes, and practice of physicians and pharmacists regarding the prevention and treatment of cardiovascular toxicity associated with cancer treatment. Sci Rep 2024; 14:20122. [PMID: 39209910 PMCID: PMC11362540 DOI: 10.1038/s41598-024-71015-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/23/2024] [Indexed: 09/04/2024] Open
Abstract
This study aimed to explore physicians' and pharmacists' knowledge, attitudes, and practice (KAP) regarding the prevention and treatment of cardiovascular toxicity associated with cancer treatment. A multicenter cross-sectional study included physicians and pharmacists between April 2023 and June 2023. The study included 918 participants (514 physicians and 404 pharmacists). The average scores of knowledge, attitudes, and practice were 11.6 ± 3.39, 24.7 ± 2.6, and 26.3 ± 6.8 points. Sufficient knowledge was significantly associated with age ≥ 41 years (odds ratio (OR) = 2.745, 95% confidence interval (CI) 1.086-6.941, P = 0.033), male (OR = 2.745, 95% CI 1.150-2.223, P = 0.005), bachelor's degree (OR = 0.084, 95% CI 0.013-0.533, P = 0.009), master's degree and above (OR = 0.096, 95% CI 0.015-0.609, P = 0.013), physician occupation (OR = 7.601, 95% CI 1.337-43.207, P = 0.022), pharmacy department (OR = 18.858, 95% CI 3.245-109.57, P = 0.001), oncology department (OR = 4.304, 95% CI 2.426-7.634, P < 0.001), cardiology department (OR = 3.001, 95% CI 1.387-6.492, P = 0.005), hospitals located in Eastern China (OR = 1.957, 95% CI 1.120-3.418, P = 0.018), and hospitals located in Western China (OR = 3.137, 95% CI 1.783-5.518, P < 0.001). Positive attitudes were significantly associated with a senior professional title (OR = 2.989, 95% CI 1.124-7.954, P = 0.028) and hospitals located in Eastern China (OR = 0.424, 95% CI 0.257-0.698, P = 0.001), Western China (OR = 0.231, 95% CI 0.136-0.394, P < 0.001), and Southern China (OR = 0.341, 95% CI 0.198-0.587, P < 0.001). Proactive practice was significantly associated with male (OR = 1.414, 95% CI 1.029-1.943, P = 0.033), senior professional title (OR = 3.838, 95% CI 1.176-12.524, P = 0.026), oncology department (OR = 3.827, 95% CI 2.336-6.272, P < 0.001), and cardiology department (OR = 2.428, 95% CI 1.263-4.669, P = 0.008). Both physicians and pharmacists had positive attitudes toward the prevention and treatment of cardiovascular toxicity associated with cancer treatment, while their knowledge and practice were not as proactive.
Collapse
Affiliation(s)
- Congxin Li
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China.
| | - Wei Yan
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Meiling Gao
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Zhihan Zhang
- Department of Nutrition, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050010, China
| | - Ling Zhao
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Jingtao Ma
- Department of Cardiovascular, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050010, China
| | - Xuejing Li
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Yuan Gao
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| | - Dongxia Zhang
- Department of Hepatology Infection, Bethune International Peace Hospital, Shijiazhuang, 050051, China
| | - Shan Gao
- Department of Pharmacy, Hebei Medical University Third Hospital, Shijiazhuang, 050051, China
| |
Collapse
|
2
|
Chen M, Xue J, Wang M, Yang J, Chen T. Cardiovascular Complications of Pan-Cancer Therapies: The Need for Cardio-Oncology. Cancers (Basel) 2023; 15:cancers15113055. [PMID: 37297017 DOI: 10.3390/cancers15113055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/28/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
It is more likely that a long-term survivor will have both cardiovascular disease and cancer on account of the progress in cancer therapy. Cardiotoxicity is a well-recognized and highly concerning adverse effect of cancer therapies. This side effect can manifest in a proportion of cancer patients and may lead to the discontinuation of potentially life-saving anticancer treatment regimens. Consequently, this discontinuation may adversely affect the patient's survival prognosis. There are various underlying mechanisms by which each anticancer treatment affects the cardiovascular system. Similarly, the incidence of cardiovascular events varies with different protocols for malignant tumors. In the future, comprehensive cardiovascular risk assessment and clinical monitoring should be considered for cancer treatments. Baseline cardiovascular evaluation risk should be emphasized prior to initiating clinical therapy in patients. Additionally, we highlight that there is a need for cardio-oncology to avoid or prevent cardiovascular side effects. Cardio-oncology service is based on identifying cardiotoxicity, developing strategies to reduce these toxicities, and minimizing long-term cardiotoxic effects.
Collapse
Affiliation(s)
- Mengjia Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jianing Xue
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Maoling Wang
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Junyao Yang
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China
- Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou 310058, China
| |
Collapse
|
3
|
Liang Z, He Y, Hu X. Cardio-Oncology: Mechanisms, Drug Combinations, and Reverse Cardio-Oncology. Int J Mol Sci 2022; 23:ijms231810617. [PMID: 36142538 PMCID: PMC9501315 DOI: 10.3390/ijms231810617] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/16/2022] Open
Abstract
Chemotherapy, radiotherapy, targeted therapy, and immunotherapy have brought hope to cancer patients. With the prolongation of survival of cancer patients and increased clinical experience, cancer-therapy-induced cardiovascular toxicity has attracted attention. The adverse effects of cancer therapy that can lead to life-threatening or induce long-term morbidity require rational approaches to prevention and treatment, which requires deeper understanding of the molecular biology underpinning the disease. In addition to the drugs used widely for cardio-protection, traditional Chinese medicine (TCM) formulations are also efficacious and can be expected to achieve “personalized treatment” from multiple perspectives. Moreover, the increased prevalence of cancer in patients with cardiovascular disease has spurred the development of “reverse cardio-oncology”, which underscores the urgency of collaboration between cardiologists and oncologists. This review summarizes the mechanisms by which cancer therapy induces cardiovascular toxicity, the combination of antineoplastic and cardioprotective drugs, and recent advances in reverse cardio-oncology.
Collapse
|
4
|
Analysis of Epidemiological Characteristics of New Cardiovascular Diseases in Cancer Patients with Cardiovascular Disease. JOURNAL OF ONCOLOGY 2022; 2022:5157398. [PMID: 36090898 PMCID: PMC9452938 DOI: 10.1155/2022/5157398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022]
Abstract
In cancer patients, a cardiovascular disease (CVD) is a prevalent occurrence. When a patient has both heart disease and cancer, the treatment can be complicated because treatment for one condition can have an adverse effect on the outcome of the other. A cardiovascular disease that involves heart failures, coronary artery disease (CAD), stroke, pericardial diseases, arrhythmias, and valve and vascular dysfunction is a serious worry for long-term cancer patients. Because preclinical research is limited, it is critical to comprehend the pathophysiology of CVD as a consequence of anticancerous therapies while taking into account the developing and expanding heart. As a result, in this research, we look at the epidemiological characteristics of cancer patients who also have cardiovascular illness. Low-dose chest computed tomography, cardiac CT, and cardiac magnetic resonance imaging (MRI) are used to acquire the data and perform the screening. Chemotherapeutic drugs such as anthracyclines and trastuzumab are used to treat the condition. Univariate analysis is used to examine risk factors and predict cardiovascular damage. Sensitivity, specificity, positive predictive value, negative predictive value, life expectancy, left ventricular ejection fraction (LVEF), and longitudinal strain are among the metrics examined.
Collapse
|
5
|
The Therapeutic Potential of Carnosine as an Antidote against Drug-Induced Cardiotoxicity and Neurotoxicity: Focus on Nrf2 Pathway. Molecules 2022; 27:molecules27144452. [PMID: 35889325 PMCID: PMC9324774 DOI: 10.3390/molecules27144452] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
Different drug classes such as antineoplastic drugs (anthracyclines, cyclophosphamide, 5-fluorouracil, taxanes, tyrosine kinase inhibitors), antiretroviral drugs, antipsychotic, and immunosuppressant drugs are known to induce cardiotoxic and neurotoxic effects. Recent studies have demonstrated that the impairment of the nuclear factor erythroid 2–related factor 2 (Nrf2) pathway is a primary event in the pathophysiology of drug-induced cardiotoxicity and neurotoxicity. The Nrf2 pathway regulates the expression of different genes whose products are involved in antioxidant and inflammatory responses and the detoxification of toxic species. Cardiotoxic drugs, such as the anthracycline doxorubicin, or neurotoxic drugs, such as paclitaxel, suppress or impair the Nrf2 pathway, whereas the rescue of this pathway counteracts both the oxidative stress and inflammation that are related to drug-induced cardiotoxicity and neurotoxicity. Therefore Nrf2 represents a novel pharmacological target to develop new antidotes in the field of clinical toxicology. Interestingly, carnosine (β-alanyl-l-histidine), an endogenous dipeptide that is characterized by strong antioxidant, anti-inflammatory, and neuroprotective properties is able to rescue/activate the Nrf2 pathway, as demonstrated by different preclinical studies and preliminary clinical evidence. Starting from these new data, in the present review, we examined the evidence on the therapeutic potential of carnosine as an endogenous antidote that is able to rescue the Nrf2 pathway and then counteract drug-induced cardiotoxicity and neurotoxicity.
Collapse
|
6
|
Rembiałkowska N, Novickij V, Baczyńska D, Dubińska-Magiera M, Saczko J, Rudno-Rudzińska J, Maciejewska M, Kulbacka J. Micro- and Nanosecond Pulses Used in Doxorubicin Electrochemotherapy in Human Breast and Colon Cancer Cells with Drug Resistance. Molecules 2022; 27:molecules27072052. [PMID: 35408450 PMCID: PMC9000361 DOI: 10.3390/molecules27072052] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022] Open
Abstract
(1) Background: Pulsed electric field (PEF) techniques are commonly used to support the delivery of various molecules. A PEF seems a promising method for low permeability drugs or when cells demonstrate therapy resistance and the cell membrane becomes an impermeable barrier. (2) Methods: In this study, we have used doxorubicin-resistant and sensitive models of human breast cancer (MCF-7/DX, MCF-7/WT) and colon cancer cells (LoVo, LoVoDX). The study aimed to investigate the susceptibility of the cells to doxorubicin (DOX) and electric fields in the 20–900 ns pulse duration range. The viability assay was utilized to evaluate the PEF protocols’ efficacy. Cell confluency and reduced glutathione were measured after PEF protocols. (3) Results: The obtained results showed that PEFs significantly supported doxorubicin delivery and cytotoxicity after 48 and 72 h. The 60 kV/cm ultrashort pulses × 20 ns × 400 had the most significant cytotoxic anticancer effect. The increase in DOX concentration provokes a decrease in cell viability, affected cell confluency, and reduced GSSH when combined with the ESOPE (European Standard Operating Procedures of Electrochemotherapy) protocol. Additionally, reactive oxygen species after PEF and PEF-DOX were detected. (4) Conclusions: Ultrashort electric pulses with low DOX content or ESOPE with higher DOX content seem the most promising in colon and breast cancer treatment.
Collapse
Affiliation(s)
- Nina Rembiałkowska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (N.R.); (D.B.); (J.S.)
| | - Vitalij Novickij
- Institute of High Magnetic Fields, Vilnius Gediminas Technical University, LT-03227 Vilnius, Lithuania;
| | - Dagmara Baczyńska
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (N.R.); (D.B.); (J.S.)
| | - Magda Dubińska-Magiera
- Department of Animal Developmental Biology, Faculty of Biological Science, University of Wroclaw, Sienkiewicza 21, 50-335 Wroclaw, Poland;
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (N.R.); (D.B.); (J.S.)
| | - Julia Rudno-Rudzińska
- Department of General and Oncological Surgery, Medical University Hospital, Borowska 211, 50-556 Wroclaw, Poland;
| | - Magdalena Maciejewska
- Laboratory of Experimental Anticancer Therapy, Hirszfeld Institute of Immunology and Experimental Therapy, Rudolfa Weigla 12, 53-114 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland; (N.R.); (D.B.); (J.S.)
- Correspondence:
| |
Collapse
|
7
|
Chen D, Kelly C, Haw TJ, Lombard JM, Nordman IIC, Croft AJ, Ngo DTM, Sverdlov AL. Heart Failure in Breast Cancer Survivors: Focus on Early Detection and Novel Biomarkers. Curr Heart Fail Rep 2021; 18:362-377. [PMID: 34731413 DOI: 10.1007/s11897-021-00535-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2021] [Indexed: 01/17/2023]
Abstract
PURPOSE OF REVIEW Breast cancer survival rate has greatly improved in the last two decades due to the emergence of next-generation anti-cancer agents. However, cardiotoxicity remains a significant adverse effect arising from traditional and emerging chemotherapies as well as targeted therapies for breast cancer patients. In this review, we will discuss cardiotoxicities of both traditional and emerging therapies for breast cancer. We will discuss current practices to detect cardiotoxicity of these therapies with the focus on new and emerging biomarkers. We will then focus on 'omics approaches, especially the use of epigenetics to discover novel biomarkers and therapeutics to mitigate cardiotoxicity. RECENT FINDINGS Significant cardiotoxicities of conventional chemotherapies remain and new and unpredictable new forms of cardiac and/or vascular toxicity emerge with the surge in novel and targeted therapies. Yet, there is no clear guidance on detection of cardiotoxicity, except for significant left ventricular systolic dysfunction, and even then, there is no uniform definition of what constitutes cardiotoxicity. The gold standard for detection of cardiotoxicity involves a serial echocardiography in conjunction with blood-based biomarkers to detect early subclinical cardiac dysfunction. However, the ability of these tests to detect early disease remains limited and not all forms of toxicity are detectable with these modalities. There is an unprecedented need to discover novel biomarkers that are sensitive and specific for early detection of subclinical cardiotoxicity. In that space, novel echocardiographic techniques, such as strain, are becoming more common-place and new biomarkers, discovered by epigenetic approaches, seem to become promising alternatives or adjuncts to conventional non-specific cardiac biomarkers.
Collapse
Affiliation(s)
- Dongqing Chen
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia
| | - Conagh Kelly
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia
| | - Tatt Jhong Haw
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia.,Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Janine M Lombard
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Ina I C Nordman
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Amanda J Croft
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia
| | - Doan T M Ngo
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia. .,School of Biomedical Science and Pharmacy, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.
| | - Aaron L Sverdlov
- Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle, NSW, Callaghan, Australia. .,Cardio-Oncology & Cardiometabolic Research Group, College of Health, Medicine and Wellbeing, Hunter Medical Research Institute & University of Newcastle Calvary Mater Newcastle, NSW, Waratah, Australia. .,Cardiovascular Department, John Hunter Hospital, Hunter New England Local Health District, NSW, New Lambton Heights, Australia. .,School of Medicine and Public Health, College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia.
| |
Collapse
|
8
|
Boovarahan SR, Kurian GA. Preconditioning the rat heart with 5-azacytidine attenuates myocardial ischemia/reperfusion injury via PI3K/GSK3β and mitochondrial K ATP signaling axis. J Biochem Mol Toxicol 2021; 35:e22911. [PMID: 34462995 DOI: 10.1002/jbt.22911] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 07/22/2021] [Accepted: 08/24/2021] [Indexed: 12/18/2022]
Abstract
5-Azacytidine is well known for its clinical usage in cancer treatments. The present study investigates the role of 5-azacytidine as a cardioprotective agent to ameliorate ischemia/reperfusion (I/R) injury. The cardioprotective effect of 5-azacytidine was evaluated in three experimental models: in vitro, ex vivo, and in vivo. The cardioprotective effect was evaluated via cell viability, hemodynamic indices, infarct size measurement, and assessment of histopathology, oxidative stress, and mitochondrial function. The experiments were repeated in the presence of PI3K/GSK3β and mitochondrial KATP (mtKATP ) cardioprotective signaling pathway inhibitors to understand the underlying mechanism. 5-Azacytidine improved the cell viability by 29% in I/R-challenged H9C2 cells. Both isolated rat heart and LAD ligation model confirmed the infarct sparing effect of 5-azacytidine against I/R. It also provided a beneficial effect by normalizing the altered hemodynamics, reducing the infarct size and cardiac injury markers, reversing the perturbation of mitochondria, reduced oxidative stress, and improved the pPI3K and pAKT protein expression from I/R. In addition, it also augmented the activation of PI3K/AKT and mtKATP signaling pathway, confirmed by using wortmannin (PI3K inhibitor), SB216763 (GSK3β inhibitor), and glibenclamide (mtKATP channel closer). The effectiveness of 5-azacytidine as a cardioprotective agent is attributed to its activation of the PI3K/GSK3β and mtKATP channel signaling axis, thereby preserving mitochondrial function and reducing oxidative stress.
Collapse
Affiliation(s)
- Sri Rahavi Boovarahan
- School of Chemical and Biotechnology, Vascular Biology Lab, SASTRA Deemed University, Thanjavur, Tamilnadu, India
| | - Gino A Kurian
- School of Chemical and Biotechnology, Vascular Biology Lab, SASTRA Deemed University, Thanjavur, Tamilnadu, India
| |
Collapse
|
9
|
Abstract
BACKGROUND Immunotherapy is a significant breakthrough in cancer therapy in the last decade. Immunotherapy is better tolerated compared with chemotherapy. However, it does have side effects, and one of the rare and serious side effects of immunotherapy is cardiotoxicity. Cardiotoxicity has been described with other cancer-related treatments such as chemotherapy and targeted therapy. A high index of suspicion is required, and prompt management with immunosuppression needs to be instituted as soon as possible to prevent fatal outcomes. AREAS OF UNCERTAINTY Research is still ongoing to identify biomarkers that will help us to choose the patients, who will respond well to immunotherapy. Tumor-infiltrating lymphocytes, tumor PD-L1 expression, and tumor mutational burden explored as potential biomarkers. There are no predictive biomarkers to identify patients who are at higher risk of severe cardiotoxicity. Both cardiologists and oncologists should be aware of cardiac toxicity from immune checkpoint inhibitors. CONCLUSION All patients who are starting immune checkpoint inhibitors should undergo baseline cardiac risk factor assessment with referral to a cardiologist in a patient with multiple risk factors or previous history of cardiovascular disease. Cardiac immune-related adverse events are higher in patients taking combination therapy with anti-CTLA-4/anti-PD-1 agents compared with monotherapy. Patients with known cardiac comorbidities require a higher level of vigilance to monitor for cardiac toxicity because nonspecific symptoms can lead to rapid clinical deterioration and a higher rate of mortality when treated with checkpoint inhibitors.
Collapse
|
10
|
Life after Cell Death-Survival and Survivorship Following Chemotherapy. Cancers (Basel) 2021; 13:cancers13122942. [PMID: 34208331 PMCID: PMC8231100 DOI: 10.3390/cancers13122942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Treatment of aggressive cancers often relies on chemotherapy. This treatment has improved survival rates, but while effective at killing cancer cells, inevitably it also kills or alters the function of others. While many of the known effects are transient and resolve after treatment, as survival rates increase, so does our understanding of the long-term health costs that accompany cancer survivors. Here we provide an overview of common long-term morbidities known to be caused by conventional chemotherapy, including the risk of relapse, but more importantly, the cost of quality of life experienced, especially by those who have cancer in early life. We aim to highlight the importance of the development of targeted therapies to replace the use of conventional chemotherapy, but also that of treating the patients along with the disease to enable not only longer but also healthier life after cancer. Abstract To prevent cancer cells replacing and outnumbering their functional somatic counterparts, the most effective solution is their removal. Classical treatments rely on surgical excision, chemical or physical damage to the cancer cells by conventional interventions such as chemo- and radiotherapy, to eliminate or reduce tumour burden. Cancer treatment has in the last two decades seen the advent of increasingly sophisticated therapeutic regimens aimed at selectively targeting cancer cells whilst sparing the remaining cells from severe loss of viability or function. These include small molecule inhibitors, monoclonal antibodies and a myriad of compounds that affect metabolism, angiogenesis or immunotherapy. Our increased knowledge of specific cancer types, stratified diagnoses, genetic and molecular profiling, and more refined treatment practices have improved overall survival in a significant number of patients. Increased survival, however, has also increased the incidence of associated challenges of chemotherapy-induced morbidity, with some pathologies developing several years after termination of treatment. Long-term care of cancer survivors must therefore become a focus in itself, such that along with prolonging life expectancy, treatments allow for improved quality of life.
Collapse
|
11
|
Micheletti PL, Carla-da-Silva J, Scandolara TB, Kern R, Alves VD, Malanowski J, Victorino VJ, Herrera ACSA, Rech D, Souza JAO, Simão ANC, Panis C, Dichi I. Proinflammatory circulating markers: new players for evaluating asymptomatic acute cardiovascular toxicity in breast cancer treatment. J Chemother 2021; 33:106-115. [PMID: 33480330 DOI: 10.1080/1120009x.2021.1873632] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
PURPOSE This study aimed to evaluate markers of cardiac damage (total CK, CKMB and CRP), inflammatory markers (free iron, homocysteine and TNF-α) as well as lipidogram in breast cancer patients undergoing acute cycles of doxorubicin (DOX), paclitaxel (PTX) or trastuzumab (TZ) and to verify if there is an association between these markers and the toxicity of the chemotherapeutic treatment. Methods: Included in the study were 120 breast cancer patients and 50 healthy controls. All analyzes were performed on automated systems. For the statistical analysis, each group was compared with the controls according to their normality by Student's t-test and Mann-Whitney test. Results: Our results showed that DOX treatment led to increased hsCRP (4.80 ± 1.23 mg/dL, p = 0.0005), triglycerides (187.6 ± 25.06, p = 0.0231), TNF-α (42.31 ± 17.96 pg/mL, p = 0.01) and Fe levels (138.8 ± 18.6 μg/dL, p = 0.0193). In the meantime, PTX induced changes in CK-MB (8.78 ± 4.2 U/L, p = 0.0361), hsCRP (7.12 ± 1.87 mg/dL, p = 0.0006), cholesterol (201.7 ± 19.54, p = 0.05), triglycerides (201.7 ± 19.54, p = 0.0277), TNF-α (38.27 ± 9.12 pg/mL, p = 0.023), homocysteine (10.95 ± 0, 86 μmol/L, p = 0.005), and free iron (113 ± 18 6 μg/dL, p = 0.045) while TZ augmented CK-MB (6.9 ± 1.97 U/L, p < 0.00), hsPCR (3.12 ± 0.68 mg/dL, p = 0.095), cholesterol (218.3 ± 16.79, p = 0.0317), triglycerides (218.3 ± 16.79, p = 0.0127), TNF-α (89.6 ± 12.11, p = 0.032), homocysteine (9.95 ± 1.15 μmol/L, p = 0.0396), free iron (120.5 ± 4.64 μg/dl, p = 0.0058) as well. Conclusions: Our data demonstrated the existence of a proinflammatory net triggered by breast cancer chemotherapy that could increase cardiomyocytes permeability and allow the leakage of circulating proteins as CK-MB and induce the production of hsCRP.
Collapse
Affiliation(s)
- P L Micheletti
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Estadual de Londrina, Paraná, Brazil
| | - J Carla-da-Silva
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - T B Scandolara
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - R Kern
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - V D Alves
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - J Malanowski
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - V J Victorino
- Instituto Federal do Rio de Janeiro, Campus Engenheiro Paulo de Frontin, Rio de Janeiro, Brazil
| | - A C S A Herrera
- Pontifícia Universidade Católica de Londrina, Paraná, Brazil
| | - D Rech
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil.,Hospital de Câncer de Francisco Beltrão, Ceonc, Paraná, Brazil
| | - J A O Souza
- Hospital de Câncer de Francisco Beltrão, Ceonc, Paraná, Brazil
| | - A N C Simão
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Estadual de Londrina, Paraná, Brazil
| | - C Panis
- Laboratório de Biologia de Tumores, Universidade Estadual do Oeste do Paraná, Paraná, Brazil
| | - I Dichi
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Estadual de Londrina, Paraná, Brazil
| |
Collapse
|
12
|
Iqbal M, Victory V, Astuti A, Febrianora M, Karwiky G, Achmad C, Akbar MR. Cardiotoxicity by Anthracycline Regimen Chemotherapy Prolonged T Peak to T End Interval. Cardiol Res 2020; 11:305-310. [PMID: 32849965 PMCID: PMC7430896 DOI: 10.14740/cr1052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 06/12/2020] [Indexed: 11/21/2022] Open
Abstract
Background Myocardial necrosis may occur due to anthracycline (doxorubicin/adriamycin) chemotherapy usage. Furthermore, myocardial necrosis can affect the heterogeneity of heart conduction system and lead to repolarization abnormalities. The aim of this study was to investigate the effect of cardiotoxicity caused by anthracycline to repolarization abnormalities measured by T peak to T end (TpTe) interval. Methods This was a single center prospective cohort study with linear regression from October 2018 to May 2019. The subjects of the study were breast cancer patients after completing administration of chemotherapy with fluorouracil, adriamycin and cyclophosphamide (FAC) regimen (containing anthracycline) for 6 months. Myocardial necrosis was assessed by high sensitive (hs)-troponin I, and the heterogeneity of repolarization was measured by TpTe interval. Results This study involved 25 breast cancer patients after chemotherapy in the 6-month FAC regimen. The mean age is 46 ± 7 years, and the cumulative dose of anthracycline is 591 ± 52 mg/m2. The mean level of hs-troponin I is 90.5 ± 44.7 ng/L and the TpTe interval is 108.2 ± 10 ms. The results of linear regression analysis showed a positive correlation between hs-troponin I and TpTe interval (r: 0.421, P: 0.036) after controlling for one confounding variable (cumulative dose of anthracycline). Conclusions Cardiotoxicity caused by accumulative dose of anthracycline may lead to myocardial necrosis which was shown by elevated hs-troponin I levels. This process may lead to heterogeneity conduction system that affect the repolarization phase of cardiac cycle which was shown by increased TpTe interval.
Collapse
Affiliation(s)
- Mohammad Iqbal
- Department of Cardiology and Vascular Medicine, Hasan Sadikin General Hospital, Jalan Eyckman 38, Bandung 40161, Indonesia
| | - Viky Victory
- Department of Cardiology and Vascular Medicine, Hasan Sadikin General Hospital, Jalan Eyckman 38, Bandung 40161, Indonesia
| | - Astri Astuti
- Department of Cardiology and Vascular Medicine, Hasan Sadikin General Hospital, Jalan Eyckman 38, Bandung 40161, Indonesia
| | - Mega Febrianora
- Department of Cardiology and Vascular Medicine, Hasan Sadikin General Hospital, Jalan Eyckman 38, Bandung 40161, Indonesia
| | - Giky Karwiky
- Department of Cardiology and Vascular Medicine, Hasan Sadikin General Hospital, Jalan Eyckman 38, Bandung 40161, Indonesia
| | - Chaerul Achmad
- Department of Cardiology and Vascular Medicine, Hasan Sadikin General Hospital, Jalan Eyckman 38, Bandung 40161, Indonesia
| | - Mohammad Rizki Akbar
- Department of Cardiology and Vascular Medicine, Hasan Sadikin General Hospital, Jalan Eyckman 38, Bandung 40161, Indonesia
| |
Collapse
|
13
|
An EJ, Kim Y, Lee SH, Ko HM, Chung WS, Jang HJ. Anti-Cancer Potential of Oxialis obtriangulata in Pancreatic Cancer Cell through Regulation of the ERK/Src/STAT3-Mediated Pathway. Molecules 2020; 25:molecules25102301. [PMID: 32422890 PMCID: PMC7288118 DOI: 10.3390/molecules25102301] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 01/09/2023] Open
Abstract
As a plant medicine, Oxalidaceae has been used to treat various diseases in Korea. However, there is little data on the anti-cancer efficacy of Oxalidaceae, particularly O. obtriangulata. This study aimed to investigate the anti-cancer effect of O. obtriangulata methanol extract (OOE) and its regulatory actions on pancreatic carcinoma. OOE showed anti-proliferative effects and induced cell death in the colony formation and cell viability assays, respectively. The Fluorescence-activated cell sorting (FACS) data confirmed that OOE significantly induced cell cycle accumulation at the G2/M phase and apoptotic effects. Additionally, OOE inhibited the activated ERK (extracellular-signal-regulated kinase)/Src (Proto-oncogene tyrosine-protein kinase Src)/STAT3 (signal transducers and activators of transcription 3) pathways including nuclear translocation of STAT3. Furthermore, suppression of Ki67, PARP(Poly ADP-ribose polymerase), caspase-3, P27(Cyclin-dependent kinase inhibitor 1B), and c-Myc as well as the STAT3 target genes CDK(cyclin-dependent kinase)1, CDK2, Cyclin B1, VEGF-1(vascular endothelial growth factor-1), MMP-9(Matrix metallopeptidase 9), and Survivin by OOE was observed in BxPC3. We speculate that these molecular actions might support an anti-cancer effect of OOE. In this study, we demonstrated that OOE may be a promising anti-cancer material and may serve as a natural therapy and alternative remedy for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Eun-Jin An
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Yumi Kim
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Seung-Hyeon Lee
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Hyun Min Ko
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
| | - Won-Seok Chung
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Correspondence: (W.-S.C.); (H.-J.J.)
| | - Hyeung-Jin Jang
- College of Korean Medicine, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (E.-J.A.); (Y.K.); (S.-H.L.); (H.M.K.)
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Korea
- College of Korean Medicine and College of Pharmacy, Kyung Hee University, 26, Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Correspondence: (W.-S.C.); (H.-J.J.)
| |
Collapse
|
14
|
Current Concepts in Pharmacometabolomics, Biomarker Discovery, and Precision Medicine. Metabolites 2020; 10:metabo10040129. [PMID: 32230776 PMCID: PMC7241083 DOI: 10.3390/metabo10040129] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Abstract
Pharmacometabolomics (PMx) studies use information contained in metabolic profiles (or metabolome) to inform about how a subject will respond to drug treatment. Genome, gut microbiome, sex, nutrition, age, stress, health status, and other factors can impact the metabolic profile of an individual. Some of these factors are known to influence the individual response to pharmaceutical compounds. An individual’s metabolic profile has been referred to as his or her “metabotype.” As such, metabolomic profiles obtained prior to, during, or after drug treatment could provide insights about drug mechanism of action and variation of response to treatment. Furthermore, there are several types of PMx studies that are used to discover and inform patterns associated with varied drug responses (i.e., responders vs. non-responders; slow or fast metabolizers). The PMx efforts could simultaneously provide information related to an individual’s pharmacokinetic response during clinical trials and be used to predict patient response to drugs making pharmacometabolomic clinical research valuable for precision medicine. PMx biomarkers can also be discovered and validated during FDA clinical trials. Using biomarkers during medical development is described in US Law under the 21st Century Cures Act. Information on how to submit biomarkers to the FDA and their context of use is defined herein.
Collapse
|
15
|
Liu Y, Wu W. Cardiovascular immune-related adverse events: Evaluation, diagnosis and management. Asia Pac J Clin Oncol 2020; 16:232-240. [PMID: 32129935 PMCID: PMC7496884 DOI: 10.1111/ajco.13326] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/28/2020] [Indexed: 12/15/2022]
Abstract
Cardiotoxicities are associated with immune checkpoint inhibitor (ICI) therapy. Recent case series and retrospective studies have shown that cardiac immune‐related adverse events (irAEs) are rare but potentially fatal complications of immunotherapy, with various underlying risk factors such as combinations of different ICIs. High mortality rates and overreactive inflammation have been observed with ICI‐associated cardiotoxicities, highlighting the necessity of baseline and serial evaluations and the identification and management of cardiac irAEs as early as possible. The clinical presentations of irAEs range from asymptomatic cardiac biomarker elevation, myocarditis and pericardial diseases to heart failure and mild to fatal arrhythmia. Troponin measurement and electrocardiogram are sensitive initial tests, whereas cardiac magnetic resonance imaging and endomyocardial biopsy are both gold standard components of the diagnostic criteria. Close monitoring and timely consultation with a cardiologist are important for the diagnosis of ICI‐related cardiotoxicities, with decisions of stopping or rechallenging ICIs and strategies to manage heart injuries. Treatment principles are made according to risk stratifications. The first‐line medication is glucocorticoids of various doses, and the second‐line immunosuppression includes intravenous immunoglobin, antithymocyte globulin and other immunosuppressants, which are recommended in life‐threatening cases or in cases of resistance/no response to steroids.
Collapse
Affiliation(s)
- Yingxian Liu
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Wei Wu
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
16
|
Mohammad Khanlou E, Atashbar S, Kahrizi F, Shokouhi Sabet N, Salimi A. Bevacizumab as a monoclonal antibody inhibits mitochondrial complex II in isolated rat heart mitochondria: ameliorative effect of ellagic acid. Drug Chem Toxicol 2020; 45:456-463. [PMID: 31973585 DOI: 10.1080/01480545.2020.1715423] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug-induced cardiotoxicity usually manifests as heart failure or left ventricular systolic dysfunction. Left ventricular dysfunction is a rarely reported side effect of bevacizumab (BEV) with an incidence of 1.2%, and this occurs irrespective of the route of administration. In this study, we focused on an analysis of BEV effects on mitochondrial complexes activities and protective effect of ellagic acid (EA) against BEV-induced mitochondria toxicity. Rat heart mitochondria were isolated using differential centrifugation form wistar rats. Using biochemical and flowcytometry assays we evaluated mitochondrial complexes activity, succinate dehydrogenases (SDH), mitochondrial swelling, reactive oxygen species (ROS) formation and mitochondrial membrane potential (MMP) in isolated mitochondria. We observed only decreased activity of complexes II after exposure with BEV (50 and 100 µg/ml). The inhibition of complex II is paralleled by the decreased MMP, mitochondrial swelling, and ROS formation. Also, we showed that EA (10-100 µM) as an antioxidant and natural agent significantly decreases mitochondrial toxicity induced by BEV. Together, for the first time, this preliminary study has demonstrated a significant decrease in activity of complexes II after exposure with BEV and proved the protective effects of EA in alleviating BEV-mediated mitochondria toxicity.
Collapse
Affiliation(s)
- Elham Mohammad Khanlou
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.,
| | - Saman Atashbar
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.,
| | - Farzad Kahrizi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Damghan Islamic Azad University, Damghan, Iran
| | - Nima Shokouhi Sabet
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran.,
| | - Ahmad Salimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
17
|
Exosome Treatment Enhances Anti-Inflammatory M2 Macrophages and Reduces Inflammation-Induced Pyroptosis in Doxorubicin-Induced Cardiomyopathy. Cells 2019; 8:cells8101224. [PMID: 31600901 PMCID: PMC6830113 DOI: 10.3390/cells8101224] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/18/2019] [Accepted: 09/26/2019] [Indexed: 12/20/2022] Open
Abstract
Doxorubicin (Dox) is an effective antineoplastic agent used to treat cancers, but its use is limited as Dox induces adverse cardiotoxic effects. Dox-induced cardiotoxicity (DIC) can lead to heart failure and death. There is no study that investigates whether embryonic stem cell-derived exosomes (ES-Exos) in DIC can attenuate inflammation-induced pyroptosis, pro-inflammatory M1 macrophages, inflammatory cell signaling, and adverse cardiac remodeling. For this purpose, we transplanted ES-Exos and compared with ES-cells (ESCs) to examine pyroptosis, inflammation, cell signaling, adverse cardiac remodeling, and their influence on DIC induced cardiac dysfunction. Therefore, we used C57BL/6J mice ages 10 ± 2 weeks and divided them into four groups (n = 6–8/group): Control, Dox, Dox + ESCs, and Dox + ES-Exos. Our data shows that the Dox treatment significantly increased expression of inflammasome markers (TLR4 and NLRP3), pyroptotic markers (caspase-1, IL1-β, and IL-18), cell signaling proteins (MyD88, p-P38, and p-JNK), pro-inflammatory M1 macrophages, and TNF-α cytokine. This increased pyroptosis, inflammation, and cell signaling proteins were inhibited with ES-Exos or ESCs. Moreover, ES-Exos or ESCs increased M2 macrophages and anti-inflammatory cytokine, IL-10. Additionally, ES-Exos or ESCs treatment inhibited significantly cytoplasmic vacuolization, myofibril loss, hypertrophy, and improved heart function. In conclusion, for the first time we demonstrated that Dox-induced pyroptosis and cardiac remodeling are ameliorated by ES-Exos or ESCs.
Collapse
|
18
|
Kleckner AS, Kleckner IR, Kamen CS, Tejani MA, Janelsins MC, Morrow GR, Peppone LJ. Opportunities for cannabis in supportive care in cancer. Ther Adv Med Oncol 2019; 11:1758835919866362. [PMID: 31413731 PMCID: PMC6676264 DOI: 10.1177/1758835919866362] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/03/2019] [Indexed: 12/17/2022] Open
Abstract
Cannabis has the potential to modulate some of the most common and debilitating symptoms of cancer and its treatments, including nausea and vomiting, loss of appetite, and pain. However, the dearth of scientific evidence for the effectiveness of cannabis in treating these symptoms in patients with cancer poses a challenge to clinicians in discussing this option with their patients. A review was performed using keywords related to cannabis and important symptoms of cancer and its treatments. Literature was qualitatively reviewed from preclinical models to clinical trials in the fields of cancer, human immunodeficiency virus (HIV), multiple sclerosis, inflammatory bowel disease, post-traumatic stress disorder (PTSD), and others, to prudently inform the use of cannabis in supportive and palliative care in cancer. There is a reasonable amount of evidence to consider cannabis for nausea and vomiting, loss of appetite, and pain as a supplement to first-line treatments. There is promising evidence to treat chemotherapy-induced peripheral neuropathy, gastrointestinal distress, and sleep disorders, but the literature is thus far too limited to recommend cannabis for these symptoms. Scant, yet more controversial, evidence exists in regard to cannabis for cancer- and treatment-related cognitive impairment, anxiety, depression, and fatigue. Adverse effects of cannabis are documented but tend to be mild. Cannabis has multifaceted potential bioactive benefits that appear to outweigh its risks in many situations. Further research is required to elucidate its mechanisms of action and efficacy and to optimize cannabis preparations and doses for specific populations affected by cancer.
Collapse
Affiliation(s)
- Amber S Kleckner
- Cancer Control and Survivorship, University of Rochester Medical Center, CU 420658, 265 Crittenden Blvd., Rochester, NY 14642, USA
| | - Ian R Kleckner
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| | - Charles S Kamen
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| | - Mohamedtaki A Tejani
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Michelle C Janelsins
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| | - Gary R Morrow
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| | - Luke J Peppone
- Cancer Control and Survivorship, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
19
|
Potential Protective Effect of Achillea fragrantissima against Adriamycin-Induced Cardiotoxicity in Rats via an Antioxidant and Anti-Inflammatory Pathway. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5269074. [PMID: 31317032 PMCID: PMC6601502 DOI: 10.1155/2019/5269074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 04/24/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Adriamycin (Adr) is a cytotoxic anthracycline agent that is utilized to manage many types of tumors, but its clinical use is undesirable due to severe cardiotoxicity. The present study aimed to investigate the cardioprotective effect of Achillea fragrantissima (A. fragrantissima) against Adr-induced cardiotoxicity through the antioxidant and anti-inflammatory metabolic pathways. A single dose of Adr was injected in rats to induce cardiotoxicity. Rats are divided into 5 groups, control, A. fragrantissima 800, Adr, A. fragrantissima 400 + Adr, and A. fragrantissima 800 + Adr. 72 h after Adr administration, electrocardiographic (ECG) study was performed for all rats. Serum and hearts were then collected for biochemical and histopathological studies. A. fragrantissima ameliorated Adr-induced ST-segment elevation. It reduced Adr-induced elevation in lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), thiobarbituric acid reactive substance (TBARS), tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and IL-6. It also protected against Adr-induced histopathological changes. Pretreatment with the extract increased heart tissue contents of glutathione peroxidase (GSH-PX) and reduced glutathione (GSH). Phytochemical analysis of the extract revealed that it is rich in phenolic and flavonoid active constituents. The results of this study revealed that A. fragrantissima extract ameliorates Adr-induced cardiotoxicity via an antioxidant and anti-inflammatory mechanisms. Further studies are warranted in order to recognize the precise active constituents of this natural extract which are responsible for the antioxidant and anti-inflammatory actions.
Collapse
|
20
|
Cardiac MRI: a Promising Diagnostic Tool to Detect Cancer Therapeutics–Related Cardiac Dysfunction. CURRENT CARDIOVASCULAR IMAGING REPORTS 2019. [DOI: 10.1007/s12410-019-9489-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
Serra W, Fantin A, Longo C, Rabia G, De Rosa F, Plenteda C, Re F, Crisafulli E, Chetta A. Carfilzomib: A Tale of a Heartbreaking Moment: Case Report and Concise Review of the Literature. Cardiovasc Hematol Disord Drug Targets 2018; 19:253-258. [PMID: 30516116 DOI: 10.2174/1871529x19666181205100705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 11/12/2018] [Accepted: 11/28/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Carfilzomib, a proteasome inhibitor, known as a therapeutical option for people who have already received one or more previous treatments for multiple myeloma, has well known cardiac and systemic adverse effects. OBJECTIVE There is evidence supporting that adverse effects are dose dependent, yet there is no known patient phenotype characterized by worse associated consequences, nor are there widely accepted monitoring protocols. RESULTS In this article we describe two patients with cardiovascular adverse events related to carfilzomib treatment and their clinical course. Our goal was to present two cases of daily practice, which highlighted the complexity of their management and led to underline how baseline evaluation and close follow-up with echocardiography and cardiac biomarkers, including natriuretic peptides, remain an important tool for the cardiotoxicity surveillance. CONCLUSION These reflections should lead to further studies in order to identify high risk patients for cardiovascular adverse event and clarify the real incidence of cardiotoxicity of this drug and adequate follow-up timing. Finally further research is needed to evaluate strategies for prevention and attenuation of cardiovascular complications of cancer therapy.
Collapse
Affiliation(s)
- W Serra
- Cardiology Unit, Department of Surgery, University of Parma, Parma, Italy
| | - A Fantin
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - C Longo
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - G Rabia
- Cardiology Unit, Department of Surgery, University of Parma, Parma, Italy
| | - F De Rosa
- Cardiology Unit, Department of Surgery, University of Parma, Parma, Italy
| | - C Plenteda
- Emathologic Department, University of Parma, Parma, Italy
| | - F Re
- Emathologic Department, University of Parma, Parma, Italy
| | - E Crisafulli
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - A Chetta
- Respiratory Disease and Lung Function Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
22
|
Liu B, An T, Li M, Yi Z, Li C, Sun X, Guan X, Li L, Wang Y, Zhang Y, Xu B, Ma F, Zeng Y. The association between early-onset cardiac events caused by neoadjuvant or adjuvant chemotherapy in triple-negative breast cancer patients and some novel autophagy-related polymorphisms in their genomic DNA: a real-world study. Cancer Commun (Lond) 2018; 38:71. [PMID: 30514381 PMCID: PMC6280434 DOI: 10.1186/s40880-018-0343-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 11/22/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND An increasing number of cancer patients die of cardiovascular diseases. The cardiotoxicity of chemotherapy is particularly important in triple-negative breast cancer (TNBC) with limited therapeutic options. Cardiac autophagy is an important mechanism of cardiotoxicity. This research was aimed to investigate the cardiotoxicity of chemotherapy in TNBC, screen the susceptible population, and determine the relationship between cardiotoxicity and autophagy-related polymorphisms. METHODS From a total of 2450 stage I-III TNBC patients, 147 met the inclusion criteria and finally recruited. Electrocardiography (ECG) was performed before most chemotherapy cycles, and echocardiography (UCG) was performed according to clinical needs. All ECG and UCG records were re-interpreted by cardiologists at the National Center for Cardiovascular Disease, Fuwai Hospital. According to the National Center for Biotechnology Information and the Catalog of Somatic Mutations in Cancer database, we selected 25 single nucleotide polymorphisms (SNPs) related to autophagy and genotyped the 147 TNBC patients. Paired-sample T tests, Chi squared tests, and logistic regression models were employed for the analysis. RESULTS Only 46 (31.3%) patients had normal ECG records after every chemotherapy cycle. Among the 16 patients who underwent UCG, 2 (12.5%) had a reversible decrease of left ventricular ejection fraction. The use of anthracyclines and excessive alcohol consumption were risk factors of ECG abnormalities. With the continuation of chemotherapy, heart rate gradually increased. Anthracyclines were associated with QRS duration abnormalities (P = 0.043). After genotyping for 25 autophagy-related SNPs, we found that the G allele of autophagy-related 13 (ATG13) rs10838611 was significantly associated with ECG abnormalities (odds ratio = 2.258, 95% confidence interval = 1.318-3.869; P = 0.003). CONCLUSION ECG abnormalities caused by chemotherapy are common in the real world. Autophagy-related SNPs are associated with chemotherapy-induced cardiotoxicity, thereby providing new evidence for autophagy as a cause of chemotherapy-induced cardiac damage.
Collapse
Affiliation(s)
- Binliang Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Tao An
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, P. R. China
| | - Meiying Li
- Shandong Cancer Hospital and Institute, Shandong University, Jinan, 250117, Shandong, P. R. China
| | - Zongbi Yi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Chunxiao Li
- State Key Laboratory of Molecular Oncology, Cancer Institute/Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Xiaoying Sun
- Department of Medical Oncology, Cancer Hospital of Huanxing, Beijing, 100065, P. R. China
| | - Xiuwen Guan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Lixi Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Yanfeng Wang
- Department of Comprehensive Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Yuhui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, P. R. China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, P. R. China.
| | - Yixin Zeng
- Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, P. R. China. .,Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, P. R. China.
| |
Collapse
|
23
|
Kansy B, Lang S. [Immunotherapy - The New Era of Oncology]. Laryngorhinootologie 2018; 97:S3-S47. [PMID: 29905353 PMCID: PMC6541097 DOI: 10.1055/s-0043-121594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
In the field of immunotherapy, essential progress was achieved over the past years partially demonstrating long lasting therapeutic responses in different tumor entities. A better understanding of the interactions between the tumor and the immune system as well as the integration of immunotherapeutic approaches into clinical routine were the foundations for this development. The different approaches intervene on multiple levels of the immune response and directly or indirectly mount the patient‘s own immune defense against tumor cells. Immunotherapeutic approaches are represented by cytokine therapies, vaccinations, the use of oncolytic viruses and monoclonal antibody therapies as well as adoptive cell transfer strategies.
Collapse
Affiliation(s)
- Benjamin Kansy
- Klinik für Hals-Nasen-Ohrenheilkunde, Kopf- und Halschirurgie, Universitätsklinikum Essen, Universität Duisburg-Essen
| | - Stephan Lang
- Klinik für Hals-Nasen-Ohrenheilkunde, Kopf- und Halschirurgie, Universitätsklinikum Essen, Universität Duisburg-Essen
| |
Collapse
|
24
|
Yang X, Liu N, Li X, Yang Y, Wang X, Li L, Jiang L, Gao Y, Tang H, Tang Y, Xing Y, Shang H. A Review on the Effect of Traditional Chinese Medicine Against Anthracycline-Induced Cardiac Toxicity. Front Pharmacol 2018; 9:444. [PMID: 29867456 PMCID: PMC5963334 DOI: 10.3389/fphar.2018.00444] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 04/16/2018] [Indexed: 01/29/2023] Open
Abstract
Anthracyclines are effective agents generally used to treat solid-tumor and hematologic malignancies. The use of anthracyclines for over 40 years has improved cancer survival statistics. Nevertheless, the clinical utility of anthracyclines is limited by its dose-dependent cardiotoxicity that adversely affects 10-30% of patients. Anthracycline-induced cardiotoxicity may be classified as acute/subacute or chronic/late toxicity and leads to devastating adverse effects resulting in poor quality of life, morbidity, and premature mortality. Traditional Chinese medicine has a history of over 2,000 years, involving both unique theories and substantial experience. Several studies have investigated the potential of natural products to decrease the cardiotoxic effects of chemotherapeutic agents on healthy cells, without negatively affecting their antineoplastic activity. This article discusses the mechanism of anthracycline-induced cardiotoxicity, and summarizes traditional Chinese medicine treatment for anthracycline-induced heart failure (HF), cardiac arrhythmia, cardiomyopathy, and myocardial ischemia in recent years, in order to provide a reference for the clinical prevention and treatment of cardiac toxicity.
Collapse
Affiliation(s)
- Xinyu Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Nian Liu
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Xinye Li
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yihan Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China.,Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xiaofeng Wang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Linling Li
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Le Jiang
- Department of Cardiology, Beijing Anzhen Hospital of the Capital University of Medical Sciences, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Hebin Tang
- Department of Pharmacology, School of Pharmaceutical Sciences, South-Central University for Nationalities, Wuhan, China
| | - Yong Tang
- Department of Pancreatic Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yanwei Xing
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing, China
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
25
|
Xiong C, Wu YZ, Zhang Y, Wu ZX, Chen XY, Jiang P, Guo HC, Xie KR, Wang KX, Su SW. Protective effect of berberine on acute cardiomyopathy associated with doxorubicin treatment. Oncol Lett 2018; 15:5721-5729. [PMID: 29552206 PMCID: PMC5840547 DOI: 10.3892/ol.2018.8020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 12/22/2017] [Indexed: 12/27/2022] Open
Abstract
Doxorubicin (DOX) is a potent and broad-spectrum anthracycline chemotherapeutic agent, but dose-dependent cardiotoxic side effects limit its clinical application. This toxicity is closely associated with the generation of reactive oxygen species (ROS) radical during DOX metabolism. The present study investigated the effects of Berberine (Ber) on DOX-induced acute cardiac injury in a rat model and analysed its mechanism in cardiomyocytes in vitro. Serum creatine kinase (CK), creatine kinase isoenzyme (CK-MB) and malondialdehyde (MDA) levels were significantly increased in the DOX group compared with the control group. This increase was accompanied by cardiac histopathological injury and a decrease in cardiomyocyte superoxide dismutase (SOD) and catalase (CAT). CK, CK-MB and MDA levels decreased and SOD and CAT levels increased in the Ber-treated group compared to the DOX group. Ber ameliorated the DOX-induced increase in cytosolic calcium concentration ([Ca2+]i), attenuated mitochondrial Ca2+ overload and restored the DOX-induced loss of mitochondrial membrane potential in vitro. These results demonstrated that Ber exhibited protective effects against DOX-induced heart tissue free radical injury, potentially via the inhibition of intracellular Ca2+ elevation and attenuation of mitochondrial dysfunction.
Collapse
Affiliation(s)
- Chen Xiong
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Key Laboratory of Pharmacology and Toxicology for New Drugs, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Yan-Zhao Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yu Zhang
- Center for Reproductive Medicine, Family Planning Scientific and Technical Institution of Hebei Province, Shijiazhuang, Hebei 050000, P.R. China
| | - Zi-Xiao Wu
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Key Laboratory of Pharmacology and Toxicology for New Drugs, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Xue-Yan Chen
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Key Laboratory of Pharmacology and Toxicology for New Drugs, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Ping Jiang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Key Laboratory of Pharmacology and Toxicology for New Drugs, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Hui-Cai Guo
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Key Laboratory of Pharmacology and Toxicology for New Drugs, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Ke-Rang Xie
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Key Laboratory of Pharmacology and Toxicology for New Drugs, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Ke-Xin Wang
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Key Laboratory of Pharmacology and Toxicology for New Drugs, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Su-Wen Su
- Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China.,Key Laboratory of Pharmacology and Toxicology for New Drugs, Department of Pharmacology, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|