Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Biomed Pharmacother ; 145: 112411, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34781149

ABSTRACT

Doxorubicin (DOX) is an effective anticancer drug. However, its use is hampered by the development of very mortal cardiomyopathy. Here, we investigate whether the co-administration of the antidepressant paroxetine (P), known to exert beneficial cardiovascular effects, would provide effective cardioprotection. Experiments were performed in male Wistar rats randomly assigned to control group (0.5 mL/kg 0.9% NaCl, i.v., n = 7), DOX group (DOX 5 mg /kg i.v., n = 23) and DOX+P group (DOX 5 mg/kg, i.v. plus P 10 mg/kg p.o. daily, beginning five days before DOX administration and during the follow-up period, n = 11). Rats' body weight and echocardiography parameters were monitored before and after drug/vehicle administration. Cardiac histology was performed post-mortem, as well as beta1-adrenergic receptor (ß1-AR), beta2-adrenergic receptor (ß2-AR), G protein-coupled receptor kinases type 2 (GRK2), type 3 (GRK3), beta-arrestin 1, and beta-arrestin 2 gene expression using RT-qPCR. DOX-treated rats exhibited bad general condition, adynamia, loss of body weight, and low survival. Echocardiography revealed two phenotypes: cardiomyopathy with left ventricular (LV) hypertrophy (DOX-HCM) and cardiomyopathy with LV dilation (DOX-DCM). In DOX-HCM rats only, there was an increased GRK2 and GRK3 gene expression and synthesis. DOX+P co-treated rats exhibited good general condition, normal spontaneous behaviour, gained weight over time, had increased survival, and preserved LV morphology and contractility. In these rats, gene expression and synthesis of GRK2 and GRK3 were decreased, while ß1-AR and ß2-AR were increased. Present results show for the first time that P effectively reduces DOX-induced cardiotoxicity and enhances survival.


Subject(s)
Cardiomyopathies/prevention & control , Cardiotoxicity/prevention & control , Doxorubicin/toxicity , Paroxetine/pharmacology , Animals , Antibiotics, Antineoplastic/toxicity , Cardiomyopathies/chemically induced , Cardiomyopathies/mortality , Cardiotonic Agents/pharmacology , Cardiotoxicity/etiology , Cardiotoxicity/mortality , Gene Expression Regulation/drug effects , Male , Rats , Rats, Wistar , Selective Serotonin Reuptake Inhibitors/pharmacology , Ventricular Remodeling/drug effects
2.
J Urol ; 207(4): 841-850, 2022 04.
Article in English | MEDLINE | ID: mdl-34854752

ABSTRACT

PURPOSE: Androgen deprivation therapy (ADT) includes bilateral orchiectomy or long-acting gonadotropin-releasing hormone (GnRH) agonists/antagonists. It remains controversial with respect to ADT associated cardiovascular outcomes. Hereby, we compared the risk of major adverse cardiovascular and cerebrovascular events (MACCEs) in patients with prostate cancer receiving either surgical castration or GnRH therapies. MATERIALS AND METHODS: Using the Taiwan Cancer Registry and Taiwan's National Health Insurance Research Database, we identified 8,413 patients receiving GnRH therapies compared with 694 receiving surgical castration from 2008 to 2017. The median followup duration was 3 years. RESULTS: The crude incidences of 3-year mortality and MACCEs were 19.90% vs 26.51% and 8.23% vs 8.65% in patients receiving GnRH therapies or surgical castration, respectively. After adjusting for age, cancer stage and comorbidities, despite no significant differences in MACCEs between groups there was a slight increase in the incidence of acute myocardial infarction (AMI) in patients receiving surgical castration compared with those receiving GnRH therapies. The mortality adjusted hazard ratios of MACCEs and AMI among patients receiving surgical castration were 1.11- and 1.8-fold higher than those receiving GnRH therapies. Notably, in subgroup analysis regarding cancer stage, patients with cancer stage IV showed the most significantly increasing risk of AMI in those receiving surgical castration compared with GnRH therapies. CONCLUSIONS: Collectively, we indicated an increased risk of AMI in patients with prostate cancer, especially in patients receiving surgical castration rather than those receiving GnRH therapies. Our findings highlight concerns regarding the cardiac safety of surgical castration compared with GnRH therapies.


Subject(s)
Androgen Antagonists/adverse effects , Antineoplastic Agents, Hormonal/adverse effects , Cardiotoxicity/etiology , Cardiovascular Diseases/etiology , Orchiectomy/adverse effects , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Cardiotoxicity/epidemiology , Cardiotoxicity/mortality , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/mortality , Humans , Male , Postoperative Complications/etiology , Risk Factors , Taiwan/epidemiology
3.
JNCI Cancer Spectr ; 5(4)2021 08.
Article in English | MEDLINE | ID: mdl-34350379

ABSTRACT

Cancer treatment-related cardiotoxicity (ie, heart failure, coronary artery disease, vascular diseases, arrhythmia) is a growing cancer survivorship concern within oncology practice; heart disease is the leading cause of noncancer death in cancer survivors and surpasses cancer as the leading cause of death for some cancers with higher survival rates. The issue of cardiotoxicity introduces a critical tradeoff that must be acknowledged and reconciled in clinical oncology practice: treating cancer aggressively and effectively in the present vs preventing future cardiotoxicity. Although many cancers must be treated as aggressively as possible, for others, multiple treatment options are available. Yet even when effective and less cardiotoxic treatments are available, they are not always chosen. Wariness to choose equally effective but less cardiotoxic treatment options may result in part from providers' and patients' reliance on "cognitive heuristics," or mental shortcuts that people (including, research shows, medical professionals) use to simplify complex judgments. These heuristics include delay discounting, availability and affect heuristics, and default bias. In the current commentary, we describe relevant research that illuminates how use of heuristics leads to biased medical decision making and translate how this research may apply when the tradeoff between aggressive cancer treatment and preventing future cardiotoxicity is considered. We discuss the implications of these biases in oncology practice, offer potential solutions to reduce bias, and call for future research in this area.


Subject(s)
Heart Diseases/etiology , Heuristics , Neoplasms/therapy , Vascular Diseases/etiology , Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Bias , Breast Neoplasms/drug therapy , Cancer Survivors , Cardiotoxicity/etiology , Cardiotoxicity/mortality , Cardiotoxicity/prevention & control , Cause of Death , Clinical Decision-Making , Guideline Adherence , Heart Diseases/mortality , Heart Diseases/prevention & control , Hodgkin Disease/drug therapy , Hodgkin Disease/radiotherapy , Humans , Neoplasms/mortality , Neoplasms/psychology , Radiotherapy/adverse effects , Trastuzumab/adverse effects , Vascular Diseases/mortality , Vascular Diseases/prevention & control
4.
Zhongguo Fei Ai Za Zhi ; 24(6): 394-403, 2021 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-34157799

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) such as antibodies against programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1), have shown remarkable efficacies in many subtypes of cancers. However, ICIs may also cause severe immune-related adverse events in the recipient patients. Recently, ICI-associated myocarditis have been reported in hundreds of patients worldwide, with a mortality rate of approximately 50% in these cases. This study aims to recapitulate the cardiotoxicity and explore the detoxicifying approaches to attenuate mortality caused by PD-1/PD-L1 inhibitors in healthy mice. METHODS: Six to eight-week-old C57BL/6 mice were inoculated with anti-PD-1 antibody (12.5 µg/g every 5 days for 6 injections), anti-PD-L1 antibody (10 µg/g once a week for 6 weeks), anti-PD-L1 antibody (with the same dosage described above) in combination with levothyroxine (0.25 µg/g, intraperitoneally injected half an hour before anti-PD-L1 antibody injection), or isotype control immunoglobulin IgG (10 µg/g once a week for 6 weeks). The ejection function of the hearts was detected by echocardiography, body temperature and blood pressure were detected by Mouse MonitorTM and non-invassive blood pressure minotor, and serum free thyroxine concentration was detected by The enzyme linked immunosorbent assay (ELISA). RESULTS: PD-L1 was expressed at different levels by the cardiomyocytes of the mice. The isotype control immunoglobulin and anti-PD-1 antibody did not cause death of the mice. The 12 mice receiving 3-6 injections of anti-PD-L1 antibody showed a significant increase in the heart-to-tibial ratio and cardiomyoctye degeneration, hyalinization and extravascular inflammatory cell infiltration. In addition, the serum thyroxine was mardedly decreased to 1/3 of that in the control group mice, and the blood pressure and body temperature were abnormally decreased in mice upon treatment with PD-L1 blockade. Eight of the 12 (66.7%) mice died from multiple intravenous injection of anti-PD-L1 antibody.Intraperitoneal injection of levothyroxine 30 min before the injection of anti-PD-L1 antibody significantly attenuated the mortality rate of the anti-PD-L1 antibody-treated mice. CONCLUSIONS: The anti-PD-L1 antibody is cardiotoxic and lethal, and levothyroxine is able to rescue the mice from this immune checkpoint inhibitor-caused mortality.


Subject(s)
Cardiotoxicity , Immune Checkpoint Inhibitors/adverse effects , Thyroxine , Animals , B7-H1 Antigen/antagonists & inhibitors , Cardiotoxicity/etiology , Cardiotoxicity/mortality , Cardiotoxicity/prevention & control , Humans , Mice , Mice, Inbred C57BL , Myocarditis/chemically induced , Myocarditis/mortality , Myocarditis/prevention & control , Programmed Cell Death 1 Receptor/metabolism , Thyroid Hormones/pharmacology , Thyroid Hormones/therapeutic use , Thyroxine/pharmacology , Thyroxine/therapeutic use
5.
Sci Rep ; 11(1): 2790, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33531527

ABSTRACT

Although radiation-induced cardiotoxicity has been addressed, its prognostic relevance to modern radiotherapy (RT) techniques is unclear. This study assessed the impact of adjuvant RT on heart-related deaths in patients with ductal carcinoma in situ. Patients who underwent adjuvant RT after breast-conserving surgery between 1988 and 2008 were identified from the Surveillance, Epidemiology, and End Results database. Kaplan‒Meier and competing risks analyses were conducted after propensity score-matching according to tumor laterality. A total of 41,526 propensity-matched patients were identified (n = 20,763 for either left- or right-sided tumor). In the analysis of the cumulative incidence of heart-related mortality events, there was a greater risk increment in the left-sided group over the first to second decades after RT in patients aged ≤ 50 years (P = 0.048). Competing risks analysis of the young patients showed that left-sided RT was associated with higher heart-related mortality rates (Grey's test, P = 0.049). The statistical significance remained after adjusting for other covariates (subdistribution hazard ratio 2.35; 95% confidence interval 1.09‒5.10). Regarding the intrinsic effect of modern RT techniques, further strategies to reduce heart-related risks are needed for young patients. Close surveillance within an earlier follow-up period should be considered for these patients in clinics.


Subject(s)
Breast Neoplasms/radiotherapy , Carcinoma, Intraductal, Noninfiltrating/radiotherapy , Cardiotoxicity , Radiotherapy, Adjuvant/adverse effects , Aged , Breast Neoplasms/surgery , Carcinoma, Intraductal, Noninfiltrating/surgery , Cardiotoxicity/etiology , Cardiotoxicity/mortality , Female , Humans , Mastectomy, Segmental/methods , Middle Aged , Retrospective Studies
6.
Clin Lung Cancer ; 21(2): 136-144.e1, 2020 03.
Article in English | MEDLINE | ID: mdl-31932217

ABSTRACT

INTRODUCTION: The cardiac radiation dose is an important predictor of cardiac toxicity and overall survival (OS) for patients with locally advanced non-small-cell lung cancer (NSCLC). However, radiation-induced cardiac toxicity among patients with early-stage NSCLC who have undergone stereotactic ablative radiotherapy (SABR) has been less well-characterized. Our objective was to assess the associations between cardiac radiation dosimetry and OS in patients with early-stage NSCLC undergoing SABR. MATERIALS AND METHODS: From 2009 to 2014, 153 patients with early-stage NSCLC had undergone SABR at a single institution. The maximum dose, mean dose, V10Gy, V25Gy, and V50Gy to 15 cardiac substructures and the whole heart were analyzed for their association with OS using the Kaplan-Meier method. An artificial neural network (ANN) analysis was performed to modulate confounding behaviors of dosimetric variables to predict for OS. RESULTS: A total of 112 patients were included in the present analysis. The right ventricle (RV) V10Gy most negatively predicted for OS, such that patients who had received a RV V10Gy dose < 4% had significantly longer OS than patients who had received a RV V10Gy does > 4% (5.3 years vs. 2.4 years). On ANN analysis, 74 input features, including cardiac dosimetry parameters, predicted for survival with a test accuracy of 64.7%. A repeat ANN analysis using dosimetry to dose neutral structure confirmed the predictive power of cardiac dosimetry. CONCLUSION: Cardiac dosimetry to subvolumes of the heart was associated with decreased OS in patients with early-stage NSCLC undergoing SABR. These data support the importance of minimizing the radiation dose to cardiac substructures. Further prioritizing the heart as an organ at risk might be warranted. Additionally, cardiac follow-up should be considered.


Subject(s)
Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/mortality , Cardiotoxicity/mortality , Lung Neoplasms/mortality , Neural Networks, Computer , Radiosurgery/adverse effects , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/surgery , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , Cardiotoxicity/diagnosis , Cardiotoxicity/etiology , Female , Follow-Up Studies , Humans , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Male , Middle Aged , Neoplasm Staging , Radiosurgery/mortality , Radiotherapy Dosage , Retrospective Studies , Survival Rate
7.
Asia Pac J Clin Oncol ; 16(1): 34-38, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31657878

ABSTRACT

AIMS: We sought to describe survival outcomes and toxicities of trastuzumab in real-world patients with HER2-positive, metastatic breast cancer (MBC) and compare these to a recent systematic review of clinical trials. METHODS: We searched the medical records of three Sydney cancer centers for patients with HER2-positive, MBC starting trastuzumab from January 2001 to March 2017. We recorded patient, tumor, and treatment characteristics; survival times from start of palliative trastuzumab; and rates of cardiac toxicity. Survival distribution was summarized using the following percentiles (represented scenario): 90th (worst-case), 75th (lower-typical), 25th (upper-typical), and 10th (best-case). Survival times were compared to recent review of HER2-positive MBC randomized trials. Factors associated with survival were assessed with Cox models. RESULTS: Characteristics of the 126 patients were: median age 53 years, ER positive cancer (50%), de-novo metastatic disease (23%), prior adjuvant trastuzumab (15%), liver metastases (37%), and brain metastases (23%). The median duration of first-line trastuzumab was 11 months (interquartile range, (IQR) 5-27). Survival times in months (vs the systematic review) were: 90th percentile 8 (9); 75th percentile 16 (19); and median 34 (33). Follow-up duration was insufficient to estimate the 25th and 10th percentiles, similar to the systematic review. Liver metastases were associated with shorter survival (HR = 1.74, 95% CI, 1.1-2.76, P = .02). Seventy percent of patients had a baseline cardiac assessment. Five patients (3.9%) developed symptomatic cardiac toxicity, similar to clinical trials. CONCLUSION: Survival and cardiac toxicity rates for women starting trastuzumab in routine practice were comparable to clinical trials. Oncologists can use clinical trial data as a reference point when explaining survival outcomes to women with HER2-positive MBC.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Breast Neoplasms/mortality , Cardiotoxicity/mortality , Practice Patterns, Physicians'/statistics & numerical data , Receptor, ErbB-2/metabolism , Trastuzumab/adverse effects , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cardiotoxicity/etiology , Cardiotoxicity/pathology , Female , Humans , Middle Aged , Survival Rate , Treatment Outcome
8.
Int J Cardiol ; 301: 163-166, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31711851

ABSTRACT

BACKGROUND: Limited information is available regarding the rate of long-term cardiovascular (CV) mortality in chronic myeloid leukaemia (CML) patients treated with second- and third-generation tyrosine kinase inhibitors (2ndG/3rdG TKIs) in the real-life practice. METHODS: We identified 656 consecutive CML patients treated with nilotinib, dasatinib, bosutinib and ponatinib. RESULTS: The 15-year CV-mortality free survival was 93 ±â€¯2.8%. Age ≥65 years (p = 0.005) and a positive history of CV disease (p = 0.04) were significantly associated with a lower CV-mortality free survival. CV disease accounted for 16.5% and 5% of potential years of life lost (PYLL) in male and female patients, respectively. The standard mortality ratio (SMR) following ischemic heart disease (IHD) was 3.9 in males and 3.8 in female patients, meaning an excess of IHD deaths observed, in comparison with the population of control. CONCLUSION: Prevention strategies based on CV risk factors, in particular in those patients with a previous history of CV disease, should be considered.


Subject(s)
Aniline Compounds , Cardiotoxicity , Cardiovascular Diseases , Dasatinib , Imidazoles , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Nitriles , Pyridazines , Pyrimidines , Quinolines , Aged , Aniline Compounds/administration & dosage , Aniline Compounds/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Cardiotoxicity/etiology , Cardiotoxicity/mortality , Cardiotoxicity/prevention & control , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/mortality , Cardiovascular Diseases/prevention & control , Dasatinib/administration & dosage , Dasatinib/adverse effects , Female , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Italy/epidemiology , Life Expectancy , Long Term Adverse Effects/chemically induced , Long Term Adverse Effects/mortality , Male , Mortality , Nitriles/administration & dosage , Nitriles/adverse effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Pyridazines/administration & dosage , Pyridazines/adverse effects , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Quinolines/administration & dosage , Quinolines/adverse effects , Risk Adjustment/methods
9.
Eur J Cancer ; 120: 1-9, 2019 10.
Article in English | MEDLINE | ID: mdl-31445454

ABSTRACT

BACKGROUND: We assessed long-term cardiac safety and efficacy in patients with human epidermal growth factor receptor 2-positive early breast cancer treated with a trastuzumab biosimilar (SB3) or its reference product, trastuzumab (TRZ), in a phase 3 study. METHODS: Patients who completed the phase 3 study could be enrolled in this extension study. The outcomes included the incidence of symptomatic congestive heart failure (CHF), asymptomatic significant left ventricular ejection fraction (LVEF) decrease, incidence of other cardiac events, event-free survival (EFS), and overall survival. In post hoc analysis, the Cox proportional hazards regression model was used to assess factors associated with EFS. RESULTS: A total of 367 patients were enrolled in the study (SB3, n = 186; TRZ, n = 181). The median follow-up duration from the main study enrolment was 40.8 and 40.5 months for SB3 and TRZ, respectively. During the two-year follow-up after adjuvant therapy, incidence of asymptomatic significant LVEF decrease was rare (SB3, n = 1; TRZ, n = 2), with all patients recovering with LVEF ≥ 50%, and no cases of symptomatic CHF or other cardiac events were reported. At 3 years, the EFS was 91.9% with SB3 and 85.2% with TRZ. The number of patients with events was 17 (9.1%) with SB3 and 31 (17.1%) with TRZ [hazard ratio: 0.47, 95% confidence interval: 0.26-0.87]. Antibody-dependent cell-mediated cytotoxicity (ADCC) activity and the breast pathologic complete response rate were the factors associated with EFS. CONCLUSION: Cardiotoxicity was rare in this extension study. EFS was higher with SB3 versus TRZ, with post hoc analysis suggesting that a downward drift in ADCC activity was a contributing factor. CLINICAL TRIAL REGISTRATION NUMBERS: NCT02771795 (EudraCT 2015-005663-17).


Subject(s)
Biosimilar Pharmaceuticals/therapeutic use , Breast Neoplasms/mortality , Carcinoma, Ductal, Breast/mortality , Carcinoma, Lobular/mortality , Neoadjuvant Therapy/mortality , Receptor, ErbB-2/metabolism , Trastuzumab/therapeutic use , Adult , Aged , Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/drug therapy , Carcinoma, Lobular/metabolism , Carcinoma, Lobular/pathology , Cardiotoxicity/mortality , Chemotherapy, Adjuvant , Cohort Studies , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Invasiveness , Prognosis , Stroke Volume , Survival Rate , Ventricular Function, Left
10.
G3 (Bethesda) ; 9(8): 2637-2646, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31263061

ABSTRACT

Anthracyclines cause progressive cardiotoxicity whose ultimate severity is individual to the patient. Genetic determinants contributing to this variation are difficult to study using current mouse models. Our objective was to determine whether a spectrum of anthracycline induced cardiac disease can be elicited across 10 Collaborative Cross mouse strains given the same dose of doxorubicin. Mice from ten distinct strains were given 5 mg/kg of doxorubicin intravenously once weekly for 5 weeks (total 25 mg/kg). Mice were killed at acute or chronic timepoints. Body weight was assessed weekly, followed by terminal complete blood count, pathology and a panel of biomarkers. Linear models were fit to assess effects of treatment, sex, and sex-by-treatment interactions for each timepoint. Impaired growth and cardiac pathology occurred across all strains. Severity of these varied by strain and sex, with greater severity in males. Cardiac troponin I and myosin light chain 3 demonstrated strain- and sex-specific elevations in the acute phase with subsequent decline despite ongoing progression of cardiac disease. Acute phase cardiac troponin I levels predicted the ultimate severity of cardiac pathology poorly, whereas myosin light chain 3 levels predicted the extent of chronic cardiac injury in males. Strain- and sex-dependent renal toxicity was evident. Regenerative anemia manifested during the acute period. We confirm that variable susceptibility to doxorubicin-induced cardiotoxicity observed in humans can be modeled in a panel of CC strains. In addition, we identified a potential predictive biomarker in males. CC strains provide reproducible models to explore mechanisms contributing to individual susceptibility in humans.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiotoxicity/etiology , Doxorubicin/adverse effects , Animals , Antibiotics, Antineoplastic/therapeutic use , Biomarkers , Biopsy , Cardiotoxicity/mortality , Crosses, Genetic , Disease Models, Animal , Doxorubicin/therapeutic use , Female , Fibrosis , Heart Diseases/diagnosis , Heart Diseases/etiology , Humans , Male , Mice
11.
Immunotherapy ; 11(8): 725-735, 2019 06.
Article in English | MEDLINE | ID: mdl-31088241

ABSTRACT

Background: With antiprogrammed death receptor-1 (anti-PD-L1) therapy, a recent meta-analysis reported higher incidence of cutaneous, endocrine and gastrointestinal complications especially with dual anti-PD-L1 immunotherapy (IMM). Methods: Our primary outcome was assessment of all cardiotoxicity grades in IMM compared with different treatments, thus a systemic review and a meta-analysis on randomized clinical trials (RCTs) were done. Results: We included 11 RCTs with 6574 patients (3234 patients in IMM arm vs 3340 patients in the other arm). Three non-small-cell lung cancer RCTs, seven melanoma RCTs and only one prostatic cancer RCT met the inclusion criteria. There were five RCTs that compared monoimmunotherapy to chemotherapy "(n = 2631 patients)". No difference exists in all cardiotoxicity grades or high-grade cardiotoxicity (p > 0.05). Lung cancer exhibited a higher response rate and lower mortality in IMM. Conclusion: There was no reported statistically significant cardiotoxicity associated with anti-PD/PD-L1 use. Lung cancer subgroups showed better response and survival rates.


Subject(s)
B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung , Immunotherapy , Lung Neoplasms , Melanoma , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Cardiotoxicity/immunology , Cardiotoxicity/mortality , Cardiotoxicity/pathology , Cardiotoxicity/prevention & control , Female , Humans , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Melanoma/immunology , Melanoma/mortality , Melanoma/pathology , Melanoma/therapy , Randomized Controlled Trials as Topic
12.
Acta Oncol ; 58(6): 872-879, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30831041

ABSTRACT

Background: There is an unmet need for markers predicting the outcome of patients with advanced soft tissue sarcoma (STS) treated with pazopanib. Since toxicity might be related to the anti-tumor activity of the drug, the aim of this study was to determine whether pazopanib-induced proteinuria, hypothyroidism and cardiotoxicity grade 3-4 were associated with outcome. Methods: The combined results of the EORTC 62043 and 62072 trials were retrospectively assessed and used in a landmark analysis to evaluate the effect of the toxicities on progression-free survival (PFS) and overall survival (OS), using the Kaplan-Meier method and Cox regression models. Results: Of the 333 eligible patients, 259 patients were included in the analyses, for which a landmark time point of 60 days after randomization/registration was selected. Proteinuria occurred in 25.1%, hypothyroidism in 22.0% and cardiotoxicity grade 3-4 in 5.8% of the patients (any grade in 41.7%). There was no effect of the occurrence of proteinuria (6-months PFS 35.4% for patients with vs. 38.3% for patients without proteinuria, HR 1.01, p = .953), hypothyroidism (41.2% vs. 36.5%, HR 0.82, p = .210) or cardiotoxicity grade 3-4 (26.7% vs. 38.2%, HR 0.97, p = .897) on PFS. Nor was there an effect of proteinuria (6-months OS 63.2% for patients with vs. 74.4% for patients without proteinuria, HR 1.22, p = .196), hypothyroidism (76.2% vs. 70.5%, HR 0.75, p = .093) or cardiotoxicity grade 3-4 (80.0% vs. 77.2%, HR 0.93, p = .801) on OS. Conclusion: There was no association between the occurrence of pazopanib-induced proteinuria, hypothyroidism and cardiotoxicity and outcome. Therefore, these toxicities cannot be used as predictors for pazopanib activity in patients with advanced STS.


Subject(s)
Angiogenesis Inhibitors/adverse effects , Cardiotoxicity/mortality , Hypothyroidism/mortality , Proteinuria/mortality , Pyrimidines/adverse effects , Sarcoma/drug therapy , Sulfonamides/adverse effects , Cardiotoxicity/etiology , Cardiotoxicity/pathology , Female , Follow-Up Studies , Humans , Hypothyroidism/chemically induced , Hypothyroidism/pathology , Indazoles , Male , Middle Aged , Prognosis , Proteinuria/chemically induced , Proteinuria/pathology , Retrospective Studies , Sarcoma/pathology , Survival Rate
13.
Int J Radiat Oncol Biol Phys ; 104(3): 582-589, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30630029

ABSTRACT

PURPOSE: Some recent studies have suggested a relationship between cardiac dose and mortality in non-small cell lung cancer (NSCLC), but others have reported conflicting data. The goal of this study was to conduct a systematic review and meta-analysis to provide an evidence-based estimate of the relationship between cardiac dose and mortality in these patients. METHODS AND MATERIALS: A systematic review of MEDLINE (PubMed) and Embase databases (inception to January 2018) was performed according to PRISMA guidelines. Studies that evaluated cardiac dosimetric factors in patients with NSCLC and included outcomes of cardiac events, cardiac mortality, and/or overall survival were identified. RESULTS: From 5614 patients across 22 studies, a total of 214 cardiac dosimetric parameters (94 unique) were assessed as possible predictors of cardiac toxicity or death. Assessed predictors included general (eg, mean heart dose [MHD]), threshold-based (eg, heart V5), and anatomic-based (eg, atria, ventricles) dosimetric factors. The most commonly analyzed parameters were MHD, heart V5, and V30. Most studies did not make corrections for multiplicity of testing. For overall survival, V5 was found to be significant on multivariable analysis (MVA) in 1 of 11 studies and V30 in 2 of 12 studies; MHD was not significant in any of 8 studies. For cardiac events, V5 was found to be significant on multivariable analysis in 1 of 2 studies, V30 in 1 of 3 studies, and MHD in 2 of 4 studies. A meta-analysis of the data could not be performed because most negative studies did not report effect estimates. CONCLUSIONS: Consistent heart dose-volume parameters associated with overall survival of patients with NSCLC were not identified. Multiplicity of testing is a major issue and likely inflates the overall risk of type I errors in the literature. Future studies should specify predictors a priori, correct for multiplicity of testing, and report effect estimates for nonsignificant variables.


Subject(s)
Carcinoma, Non-Small-Cell Lung/radiotherapy , Heart/radiation effects , Lung Neoplasms/radiotherapy , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/mortality , Cardiotoxicity/mortality , Female , Humans , Lung Neoplasms/mortality , Male , Middle Aged , Multivariate Analysis , Radiation Dosage , Radiation Exposure
14.
Heart Fail Rev ; 24(3): 325-333, 2019 05.
Article in English | MEDLINE | ID: mdl-30523513

ABSTRACT

Some randomized controlled trials (RCTs) have tested the efficacy of beta-blockers as prophylactic agents on cancer therapy-induced cardiotoxicity; however, the quality of this evidence remains undetermined. This systematic review and meta-analysis study aims to evaluate the prophylactic effects of beta-blockers, especially carvedilol, on chemotherapy-induced cardiotoxicity. RCTs were identified by searching the MEDLINE (PubMed), Embase (OvidSP), Cochrane CENTRAL (OvidSP), etc., until December 2017. Inclusion criteria were randomized clinical trial and adult cancer patients started beta-blockers before chemotherapy. We evaluated the mean differences (MD) by fixed- or random-effects model and the odds ratio by Peto's method. Primary outcome was the left ventricular ejection fraction (LVEF) of patients after chemotherapy, and secondary outcomes were all-cause mortality, clinically overt cardiotoxicity, and other echocardiographic measurements. In total, we included six RCTs that used carvedilol as a prophylactic agent in patients receiving chemotherapy. The LVEF was not significantly distinct between those using carvedilol and placebo after chemotherapy (MD, 1.74; 95% confidence interval (CI), - 0.18 to 3.66; P = 0.08). The incidence of clinically overt cardiotoxicity was lower in the carvedilol group compared with the control group (Peto OR, 0.42; 95% CI, 0.20-0.89; P = 0.02). Furthermore, after chemotherapy, the LV end-diastolic diameter did not increase in the carvedilol group compared with the placebo group (MD, - 1.41; 95% CI, - 2.32 to - 0.50; P = 0.002). The prophylactic use of carvedilol exerted no impact on the early asymptomatic LVEF decrease but seemed to attenuate the frequency of clinically overt cardiotoxicity and prevent ventricular remodeling.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Cardiotoxicity/epidemiology , Cardiotoxicity/prevention & control , Carvedilol/therapeutic use , Protective Agents/therapeutic use , Adult , Aged , Antineoplastic Agents/adverse effects , Cardiotoxicity/etiology , Cardiotoxicity/mortality , Echocardiography , Female , Humans , Incidence , Male , Middle Aged , Stroke Volume , Treatment Outcome , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Young Adult
15.
Xenobiotica ; 49(9): 1116-1125, 2019 Sep.
Article in English | MEDLINE | ID: mdl-29985077

ABSTRACT

We aimed to investigate the drug-drug interaction (DDI) between doxorubicin (DOX) and Dioscorea bulbifera L. (DB) solution in mice, and to explore the effect of P-glycoprotein (P-gp) on this type of DDI. The toxicity of DOX in the liver, kidneys, and heart was assessed with alanine aminotransferase (ALT), aspartate aminotransferase (AST), creatinine (Cr), urea nitrogen (BUN), creatine kinase MB (CK-MB), creatine kinase (CK) and histopathology. High-performance liquid chromatography tandem mass spectrometry (HPLC-MS/MS) was used to determine the concentrations of DOX in the serum, liver, kidneys and heart. Immunohistochemistry and western blots were used to determine the expression levels of P-gp in these tissues. Our results demonstrated that, after co-administration of DOX and DB, survival was significantly decreased compared with either administration of DOX or DB alone, or water. Co-administration of DOX and DB induced elevated levels of toxicity in the heart and kidneys, but not the liver, compared with DOX alone. We conclude that concurrent treatment with DOX and DB results in increased levels of toxicity due to the accumulation of DOX in the body. Delayed excretion of DOX is associated with inhibition of P-gp in liver and kidneys.


Subject(s)
Cardiotoxicity/etiology , Dioscorea/chemistry , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Kidney/drug effects , Plant Extracts/adverse effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Cardiotoxicity/metabolism , Cardiotoxicity/mortality , Herb-Drug Interactions , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Male , Mice , Mortality , Tissue Distribution
16.
Br J Haematol ; 183(5): 717-726, 2018 12.
Article in English | MEDLINE | ID: mdl-30406945

ABSTRACT

Cardiotoxicity is a known risk of anthracycline treatment. However, the relative contribution of anthracyclines to the development of congestive heart failure (CHF), when included in a poly-chemotherapy regimen, is unclear. We examined cardiotoxicity in adult patients with diffuse large B-cell lymphoma and follicular lymphoma undergoing first-line immunochemotherapy from 2000-2012. In total, 2440 patients without previous heart disease were identified from the Danish Lymphoma Registry, of which 1994 (81·7%) were treated with anthracycline-containing chemotherapy [R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine, prednisolone) or R-CHOEP (R-CHOP + etoposide)] and 446 (18·3%) were treated without anthracyclines (reference group). Compared to the reference group, the adjusted hazard ratio of CHF after 3-5 cycles of R-CHOP/CHOEP was 5·0 [95% confidence interval (CI) 1·4; 18·5], 6 cycles 6·8 (95% CI 2·0; 23·3) and >6 cycles 13·4 (95% CI 4·0; 45·0). The cumulative 5-year risk of CHF with all-cause mortality as competing risk was 4·6% after 3-5 cycles of R-CHOP/CHOEP, 4·5% after 6 and 7·9% after more than 6 cycles. Cumulative 5-year risk for patients treated without anthracyclines was 0·8%. Using anthracyclines in first-line lymphoma treatment increases risk of CHF in patients without previous history of heart disease. In particular, treatment with >6 cycles of R-CHOP/CHOEP is associated with a significant increase in CHF rate.


Subject(s)
Anthracyclines/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Arrhythmias, Cardiac/chemically induced , Heart Failure/chemically induced , Adolescent , Adult , Aged , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Arrhythmias, Cardiac/mortality , Cardiotoxicity/etiology , Cardiotoxicity/mortality , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Denmark/epidemiology , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Heart Failure/mortality , Humans , Immunotherapy/adverse effects , Lymphoma, Follicular/drug therapy , Lymphoma, Follicular/mortality , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/mortality , Male , Middle Aged , Prednisone/administration & dosage , Prednisone/adverse effects , Registries , Retrospective Studies , Risk Factors , Rituximab , Vincristine/administration & dosage , Vincristine/adverse effects , Young Adult
17.
Isr Med Assoc J ; 20(8): 486-490, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30084573

ABSTRACT

BACKGROUND: Cardiac damage caused by oncological therapy may manifest early or many years after the exposure. OBJECTIVES: To determine the differences between sub-acute and late-onset cardiotoxicity in left ventricular ejection fraction (LVEF) recovery as well as long-term prognosis. METHODS: We studied 91 patients diagnosed with impaired systolic function and previous exposure to oncological therapy. The study population was divided according to sub-acute (from 2 weeks to ≤ 1 year) and late-onset (> 1 year) presentation cardiotoxicity. Recovery of LVEF of at least 50% was defined as the primary end point and total mortality was the secondary end point. RESULTS: Fifty-three (58%) patients were classified as sub-acute, while 38 (42%) were defined as late-onset cardiotoxicity. Baseline clinical characteristics were similar in the two groups. The mean LVEF at presentation was significantly lower among patients in the late-onset vs. sub-acute group (28% vs. 37%, respectively, P < 0.001). Independent predictors of LVEF recovery were trastuzumab therapy and a higher baseline LVEF. Although long-term mortality rates were similar in the groups with sub-acute and late-onset cardiotoxicity, improvement of LVEF was independently associated with reduced mortality. CONCLUSIONS: Our findings suggest that early detection and treatment of oncological cardiotoxicity play an important role in LVEF recovery and long-term prognosis.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxicity/epidemiology , Heart/drug effects , Trastuzumab/adverse effects , Ventricular Function, Left/drug effects , Cardiotoxicity/etiology , Cardiotoxicity/mortality , Female , Heart/physiopathology , Humans , Israel/epidemiology , Male , Middle Aged , Prognosis , Recovery of Function/drug effects , Time Factors
18.
Ugeskr Laeger ; 180(6)2018 02 05.
Article in Danish | MEDLINE | ID: mdl-29429477

ABSTRACT

This review summarizes the cardiovascular non-inferiority trials of novel antidiabetic drugs performed since 2008, when regulatory agencies started mandating thorough examination of their cardiovascular safety. So far, eight randomized trials on three different drug classes have been completed. Sodium-glucose cotransporter-2 inhibitors and glucagon-like peptide-1 receptor agonists may reduce cardiovascular risk and possibly mortality, while dipeptidyl dipeptidase-4 inhibitors may increase the risk of heart failure. A brief discussion of potential mechanisms and clinical implications is provided.


Subject(s)
Cardiovascular Diseases , Hypoglycemic Agents , Cardiotoxicity/etiology , Cardiotoxicity/mortality , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/mortality , Cardiovascular Diseases/prevention & control , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacokinetics , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Glucagon-Like Peptide-1 Receptor/agonists , Heart Failure/chemically induced , Heart Failure/mortality , Heart Failure/prevention & control , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Myocardial Infarction/chemically induced , Myocardial Infarction/mortality , Myocardial Infarction/prevention & control , Risk Factors , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Sodium-Glucose Transporter 2 Inhibitors/pharmacokinetics , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Stroke/chemically induced , Stroke/mortality , Stroke/prevention & control
19.
J Pediatr Hematol Oncol ; 40(6): 417-425, 2018 08.
Article in English | MEDLINE | ID: mdl-29432315

ABSTRACT

Cardiotoxicity is a dose-limiting and potentially lethal complication of anthracycline administration. Previous studies failed to determine definitive toxic doses or cardioprotective factors. Current dosing strategies may utilize unnecessarily high anthracycline doses, such that survival benefit may not outweigh increased toxicity rates. A systematic review of randomized controlled trials and prospective/retrospective studies investigating anthracycline treatment in pediatric solid tumors was performed from PubMed/MEDLINE and Cochrane databases. Generalized linear models mapping survival, cardiotoxicity, and cardiotoxicity-free survival adjusted for male-to-female ratio, follow-up time, and concomitant chemotherapeutic drugs or cardioprotective agents (dexrazoxane) were generated using R. Survival rose linearly with increasing cumulative anthracycline dose whereas cardiotoxicity demonstrated exponential increases both without (dose, >200 mg/m) and with (dose, >400 mg/m) dexrazoxane. Maximum cardiotoxicity-free survival was 268.2 mg/m without and 431.8 mg/m with dexrazoxane. Despite increasing cardiotoxicity-free dose by >150 mg/m, dexrazoxane minimally improved projected survival (71.9% vs. 75.4%). Cardiotoxicity increased linearly as a function of follow-up time with rates doubling from 5 to 20 years, without evidence of plateau. On the basis of our model, current dosing regimens-doxorubicin doses >375 mg/m without dexrazoxane-overvalue increased anthracycline administration and may contribute to devastating cardiotoxicity. The linear increase of cardiotoxicity without evidence of plateau confirms the necessity for lifelong cardiac monitoring.


Subject(s)
Anthracyclines/adverse effects , Cardiotoxicity , Dexrazoxane/therapeutic use , Neoplasms , Adolescent , Anthracyclines/therapeutic use , Cardiotoxicity/mortality , Cardiotoxicity/prevention & control , Child , Child, Preschool , Dexrazoxane/adverse effects , Disease-Free Survival , Female , Humans , Infant , Male , Neoplasms/drug therapy , Neoplasms/mortality , Survival Rate
20.
Eur Heart J Cardiovasc Imaging ; 19(5): 544-552, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29106497

ABSTRACT

Aims: Anthracycline treatment may cause myocyte loss and expansion of the myocardial extracellular volume (ECV) fraction by oedema and fibrosis. We tested the hypotheses that adjuvant treatment for early breast cancer with the anthracycline epirubicin is dose dependently associated with increased ECV fraction and total ECV, as well as reduced total myocardial cellular volume, and that these changes could be prevented by concomitant angiotensin or beta-adrenergic blockade. Methods and results: PRevention of cArdiac Dysfunction during Adjuvant breast cancer therapy (PRADA) was a 2 × 2 factorial, placebo-controlled, double-blinded trial of candesartan and metoprolol. Sixty-nine women had valid ECV measurements. ECV fraction, total ECV, and total cellular volume were measured by cardiovascular magnetic resonance before and at the completion of anthracycline therapy. ECV fraction increased from 27.5 ± 2.7% to 28.6 ± 2.9% (P = 0.002). A cumulative doxorubicin equivalent dose of 268 mg/m2 was associated with greater increase in ECV fraction than doses <268 mg/m2 (mean change 3.4% [95% confidence interval (CI) 1.2, 5.5] vs. 0.7% [95% CI 0.0, 1.5], P = 0.006), as well as greater increase in total ECV (1.9 mL [95% CI 0.4, 3.5] vs. 0.1 mL [95% CI -0.6, 0.8], P = 0.04). In patients receiving candesartan, total cellular volume decreased (-3.5 mL [95% CI - 4.7, -2.2], P < 0.001) while in patients not receiving candesartan, it remained unchanged (P = 0.45; between group difference P = 0.003). Conclusions: Anthracycline therapy is associated with dose-dependent increase in ECV fraction and total ECV. Concomitant treatment with candesartan reduces left ventricular total cellular volume.


Subject(s)
Anthracyclines/adverse effects , Benzimidazoles/adverse effects , Breast Neoplasms/drug therapy , Cardiotoxicity/diagnostic imaging , Magnetic Resonance Imaging, Cine/methods , Tetrazoles/adverse effects , Adult , Aged , Anthracyclines/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/therapeutic use , Biphenyl Compounds , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Cardiotoxicity/mortality , Cardiotoxicity/physiopathology , Chemotherapy, Adjuvant , Cyclophosphamide/adverse effects , Cyclophosphamide/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Epirubicin/adverse effects , Epirubicin/therapeutic use , Female , Fluorouracil/adverse effects , Fluorouracil/therapeutic use , Hospitals, University , Humans , Kaplan-Meier Estimate , Mastectomy/methods , Middle Aged , Norway , Prognosis , Prospective Studies , Risk Assessment , Statistics, Nonparametric , Stroke Volume/drug effects , Survival Analysis , Tetrazoles/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...