340 results on '"Maurea, N"'
Search Results
202. Cisplatin resistance can be curtailed by blunting Bnip3-mediated mitochondrial autophagy.
- Author
-
Vianello C, Cocetta V, Catanzaro D, Dorn GW 2nd, De Milito A, Rizzolio F, Canzonieri V, Cecchin E, Roncato R, Toffoli G, Quagliariello V, Di Mauro A, Losito S, Maurea N, Scaffa C, Sales G, Scorrano L, Giacomello M, and Montopoli M
- Subjects
- Autophagy genetics, Carcinoma, Ovarian Epithelial drug therapy, Cell Line, Tumor, Drug Resistance, Neoplasm genetics, Female, Humans, Membrane Proteins metabolism, Mitochondria metabolism, Proto-Oncogene Proteins metabolism, Antineoplastic Agents therapeutic use, Bone Neoplasms drug therapy, Bone Neoplasms genetics, Bone Neoplasms metabolism, Cisplatin therapeutic use, Osteosarcoma drug therapy, Osteosarcoma genetics, Osteosarcoma metabolism, Ovarian Neoplasms drug therapy, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism
- Abstract
Cisplatin (CDDP) is commonly used to treat a multitude of tumors including sarcomas, ovarian and cervical cancers. Despite recent investigations allowed to improve chemotherapy effectiveness, the molecular mechanisms underlying the development of CDDP resistance remain a major goal in cancer research. Here, we show that mitochondrial morphology and autophagy are altered in different CDDP resistant cancer cell lines. In CDDP resistant osteosarcoma and ovarian carcinoma, mitochondria are fragmented and closely juxtaposed to the endoplasmic reticulum; rates of mitophagy are also increased. Specifically, levels of the mitophagy receptor BNIP3 are higher both in resistant cells and in ovarian cancer patient samples resistant to platinum-based treatments. Genetic BNIP3 silencing or pharmacological inhibition of autophagosome formation re-sensitizes these cells to CDDP. Our study identifies inhibition of BNIP3-driven mitophagy as a potential therapeutic strategy to counteract CDDP resistance in ovarian carcinoma and osteosarcoma., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
203. Case Series: Recovery of Chemotherapy-Related Acute Heart Failure by the Combined Use of Sacubitril Valsartan and Wearable Cardioverter Defibrillator: A Novel Winning Combination in Cardio-Oncology.
- Author
-
Canale ML, Coviello K, Solarino G, Del Meglio J, Simonetti F, Venturini E, Camerini A, Maurea N, Bisceglia I, Tessa C, and Casolo G
- Abstract
Effective anticancer treatments have dramatically improved the outcome of patients with cancer, but cardiac toxicity reduces their clinical efficacy in a non-negligible percentage of patients. Sacubitril/valsartan is a new paradigm in the treatment of chronic heart failure, with a reduced ejection fraction due to the enhancement of natriuretic peptides' properties when coupled with a blocking effect on the angiotensin II type 1 (AT1) receptors. As with other clinical conditions of heart failure with potentially reversible declines in cardiac function, a wearable cardioverter defibrillator (WCD) is a valid tool for protection against sudden death until recovery occurs. We report a case series of four patients with chemotherapy-related acute cardiac failure with severely reduced cardiac function. They were successfully treated with sacubitril/valsartan while being protected from malignant arrhythmias using a wearable cardioverter defibrillator until the recovery of cardiac function. Sacubitril/valsartan was confirmed to be effective in anthracycline-related cardiac toxicity and the wearable cardioverter defibrillator should be considered as a support tool even in the oncology patient., Competing Interests: This study received funding from Zoll (Pittsburgh, PA, USA) for open access option only. The funder was not involved in the study design, collection, analysis, interpretation of data, the writing of this article or the decision to submit it for publication. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Canale, Coviello, Solarino, Del Meglio, Simonetti, Venturini, Camerini, Maurea, Bisceglia, Tessa and Casolo.)
- Published
- 2022
- Full Text
- View/download PDF
204. Biocompatible, photo-responsive layer-by-layer polymer nanocapsules with an oil core: in vitro and in vivo study.
- Author
-
Di Cicco C, Vecchione R, Quagliariello V, Busato A, Tufano I, Bedini E, Gerosa M, Sbarbati A, Boschi F, Marzola P, Maurea N, and Netti PA
- Subjects
- Animals, Biocompatible Materials, Humans, Mice, Polymers, Curcumin, Nanocapsules, Neoplasms
- Abstract
In cancer therapy, stimulus-responsive drug delivery systems are of particular interest for reducing side effects in healthy tissues and improving drug selectivity in the tumoral ones. Here, a strategy for the preparation of a photo-responsive cross-linked trilayer deposited onto an oil-in-water nanoemulsion via a layer-by-layer technique is reported. The system is made of completely biocompatible materials such as soybean oil, egg lecithin and glycol chitosan, with heparin as the polymeric shell. The oil core is pre-loaded with curcumin as a model lipophilic active molecule with anti-tumoral properties. The trilayer cross-linkage is performed via a photoinitiator-free thiol-ene 'click' reaction. In particular, the system is implemented with an o -nitrobenzyl group functionalized with a thiol moiety which can perform both the thiol-ene 'click' reaction and the cleavage meant for controlled drug release at two different wavelengths, respectively. So the preparation and characterization of a photo-responsive natural nanocarrier (PNC) that is stable under physiological conditions owing to the thiol-ene cross-linkage are reported. PNC performance has been assessed in vitro on melanoma cells as well as in vivo on xenograft tumour-induced mice.
- Published
- 2022
- Full Text
- View/download PDF
205. Cardio-Oncology in the COVID Era (Co & Co): The Never Ending Story.
- Author
-
Bisceglia I, Canale ML, Gallucci G, Turazza FM, Lestuzzi C, Parrini I, Russo G, Maurea N, Quagliariello V, Oliva S, Di Fusco SA, Lucà F, Tarantini L, Trambaiolo P, Moreo A, Geraci G, Gabrielli D, Gulizia MM, Oliva F, and Colivicchi F
- Abstract
The pathophysiology of some non-communicable diseases (NCDs) such as hypertension, cardiovascular disease (CVD), diabetes, and cancer includes an alteration of the endothelial function. COVID-19 is a pulmonary and vascular disease with a negative impact on patients whose damaged endothelium is particularly vulnerable. The peculiar SARS-CoV-2-induced "endothelitis" triggers an intriguing immune-thrombosis that affects both the venous and arterial vascular beds. An increased liability for infection and an increased likelihood of a worse outcome have been observed during the pandemic in patients with active cancer and in cancer survivors. "Overlapping commonalities" between COVID-19 and Cardio-Oncology have been described that include shared phenotypes of cardiovascular toxicities such as left ventricular dysfunction, ischemic syndromes, conduction disturbances, myocarditis, pericarditis and right ventricular failure; shared pathophysiologic mechanisms such as inflammation, release of cytokines, the renin-angiotensin-aldosterone-pathway, coagulation abnormalities, microthrombosis and endothelial dysfunction. For these features and for the catalyst role of NCDs (mainly CVD and cancer), we should refer to COVID-19 as a "syndemic." Another challenging issue is the persistence of the symptoms, the so-called "long COVID" whose pathogenesis is still uncertain: it may be due to persistent multi-organ viral attacks or to an abnormal immune response. An intensive vaccination campaign is the most successful pharmacological weapon against SARS-CoV-2, but the increasing number of variants has reduced the efficacy of the vaccines in controlling SARS-CoV-2 infections. After a year of vaccinations we have also learned more about efficacy and side-effects of COVID-19 vaccines. An important byproduct of the COVID-19 pandemic has been the rapid expansion of telemedicine platforms across different care settings; this new modality of monitoring cancer patients may be useful even in a post pandemic era. In this paper we analyze the problems that the cardio-oncologists are facing in a pandemic scenario modified by the extensive vaccination campaign and add actionable recommendations derived from the ongoing studies and from the syndemic nature of the infection., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Bisceglia, Canale, Gallucci, Turazza, Lestuzzi, Parrini, Russo, Maurea, Quagliariello, Oliva, Di Fusco, Lucà, Tarantini, Trambaiolo, Moreo, Geraci, Gabrielli, Gulizia, Oliva and Colivicchi.)
- Published
- 2022
- Full Text
- View/download PDF
206. [ANMCO Position paper: Cardio-oncology in the COVID-19 era].
- Author
-
Bisceglia I, Gabrielli D, Canale ML, Gallucci G, Parrini I, Turazza FM, Russo G, Maurea N, Quagliariello V, Lestuzzi C, Oliva S, Di Fusco SA, Lucà F, Tarantini L, Trambaiolo P, Gulizia MM, and Colivicchi F
- Subjects
- Humans, Pandemics, SARS-CoV-2, COVID-19, Myocardial Infarction, Neoplasms therapy
- Abstract
The COVID-19 pandemic and its impact on patients with cancer and cardiovascular disease have confirmed the particular vulnerability of this population. Indeed, not only a higher risk of contracting the infection has been reported, but also an increased occurrence of a more severe course and unfavorable outcome. Beyond the direct consequences of COVID-19, the pandemic has an enormous impact on global health systems. Screening programs and non-urgent tests have been postponed; clinical trials have suffered a setback. Similarly, in the area of cardiology care, a significant decline in ST-elevation myocardial infarction accesses and an increase in cases of late presenting heart attacks with increased mortality and complication rates have been reported. Health care systems must therefore get ready to tackle the "rebound effect" that will likely show a relative increase in the short and medium term incidence of diseases such as heart failure, myocardial infarction, arrhythmias and cardio- and cerebrovascular complications. Scientific societies are taking action to provide general guidance and recommendations aimed at mitigating the unfavorable outcomes of this pandemic emergency. Cardio-oncology, as an emerging discipline, is more flexible in modulating care pathways and represents a beacon of innovation in the development of multi-specialty patient management. In the era of the COVID-19 pandemic, cardio-oncology has rapidly modified its clinical care pathways and implemented flexible monitoring protocols that include targeted use of cardiac imaging, increased use of biomarkers, and telemedicine systems. The goal of these strategic adjustments is to minimize the risk of infection for providers and patients while maintaining standards of care for the treatment of oncologic and cardiovascular diseases. The aim of this position paper is to evaluate the impact of the COVID-19 pandemic on the management of cardio-oncologic patients with the-state-of-the-art knowledge about SARS-CoV-2 and COVID-19 in order to optimize medical strategies during and after the pandemic.
- Published
- 2021
- Full Text
- View/download PDF
207. Immune checkpoint inhibitors-associated pericardial disease: a systematic review of case reports.
- Author
-
Inno A, Maurea N, Metro G, Carbone A, Russo A, and Gori S
- Subjects
- Humans, Immune Checkpoint Inhibitors adverse effects, Pericardial Effusion complications
- Abstract
Treatment with immune checkpoint inhibitors (ICIs) can be complicated by cardiovascular toxicity, including pericardial disease. To date, no prospective studies specifically investigated the optimal treatment of ICI-associated pericardial disease, and the available evidence is based on case reports and series only. We performed a systematic review of case reports and series including 20 publications for a total of 28 cases of ICI-associated pericardial disease. In this review, pericardial disease was reversible in the majority of cases (75%), although 2 deaths were reported. The majority of cases were life-threatening (G4, 53.6%) or severe (G3, 21.4%), requiring pericardiocentesis. Higher rates of improvement were associated with administration of corticosteroids (86.7% vs 61.5%), presence of other immune-related adverse events (90.9% vs. 64.7%), and non-malignant effusions (86.7% vs 42.8%). ICIs were discontinued in the majority of cases and then restarted in 7 patients with no recurrence of pericardial disease. Based on these results, ICI-associated G3-G4 pericardial disease as well as G2 pericardial disease with moderate-severe effusion should be treated with ICIs discontinuation and high-dose steroids, also performing pericardiocentesis, pericardial drainage or pericardial window in case of cardiac tamponade. For G2 with small effusion or G1 pericardial disease, ICIs might be continued and colchicine or NSAIDs could be considered. For patients requiring ICIs discontinuation, a rechallenge with ICIs seems to be feasible after resolution or meaningful improvement of pericardial disease., (© 2021. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2021
- Full Text
- View/download PDF
208. ANMCO POSITION PAPER: cardio-oncology in the COVID era (CO and CO).
- Author
-
Bisceglia I, Gabrielli D, Canale ML, Gallucci G, Parrini I, Turazza FM, Russo G, Maurea N, Quagliariello V, Lestuzzi C, Oliva S, Di Fusco SA, Lucà F, Tarantini L, Trambaiolo P, Gulizia MM, and Colivicchi F
- Abstract
The COVID-19 pandemic and its impact on patients with cancer and cardiovascular disease have confirmed the particular vulnerability of these populations. Indeed, not only a higher risk of contracting the infection has been reported but also an increased occurrence of a more severe course and unfavourable outcome. Beyond the direct consequences of COVID-19 infection, the pandemic has an enormous impact on global health systems. Screening programmes and non-urgent tests have been postponed; clinical trials have suffered a setback. Similarly, in the area of cardiology care, a significant decline in STEMI accesses and an increase in cases of late presenting heart attacks with increased mortality and complication rates have been reported. Health care systems must therefore get ready to tackle the 'rebound effect' that will likely show a relative increase in the short- and medium-term incidence of diseases such as heart failure, myocardial infarction, arrhythmias, and cardio- and cerebrovascular complications. Scientific societies are taking action to provide general guidance and recommendations aimed at mitigating the unfavourable outcomes of this pandemic emergency. Cardio-oncology, as an emerging discipline, is more flexible in modulating care pathways and represents a beacon of innovation in the development of multi-specialty patient management. In the era of the COVID-19 pandemic, cardio-oncology has rapidly modified its clinical care pathways and implemented flexible monitoring protocols that include targeted use of cardiac imaging, increased use of biomarkers, and telemedicine systems. The goal of these strategic adjustments is to minimize the risk of infection for providers and patients while maintaining standards of care for the treatment of oncologic and cardiovascular diseases. The aim of this document is to evaluate the impact of the pandemic on the management of cardio-oncologic patients with the-state-of-the-art knowledge about severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and coronavirus disease (COVID-19) in order to optimize medical strategies during and after the pandemic., (Published on behalf of the European Society of Cardiology. © The Author(s) 2021.)
- Published
- 2021
- Full Text
- View/download PDF
209. The SGLT-2 inhibitor empagliflozin improves myocardial strain, reduces cardiac fibrosis and pro-inflammatory cytokines in non-diabetic mice treated with doxorubicin.
- Author
-
Quagliariello V, De Laurentiis M, Rea D, Barbieri A, Monti MG, Carbone A, Paccone A, Altucci L, Conte M, Canale ML, Botti G, and Maurea N
- Subjects
- Animals, Anti-Inflammatory Agents pharmacology, Antifibrotic Agents pharmacology, Apoptosis drug effects, Cardiotoxicity, Cell Line, Disease Models, Animal, Doxorubicin, Female, Ferroptosis drug effects, Fibrosis, Heart Diseases chemically induced, Heart Diseases metabolism, Heart Diseases pathology, Inflammasomes metabolism, Mice, Inbred C57BL, Myeloid Differentiation Factor 88 metabolism, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Signal Transduction, Mice, Benzhydryl Compounds pharmacology, Cytokines metabolism, Glucosides pharmacology, Heart Diseases drug therapy, Inflammation Mediators metabolism, Myocytes, Cardiac drug effects, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Ventricular Function, Left drug effects
- Abstract
Background: Empagliflozin (EMPA), a selective inhibitor of the sodium glucose co-transporter 2, reduced the risk of hospitalization for heart failure and cardiovascular death in type 2 diabetic patients in the EMPA-REG OUTCOME trial. Recent trials evidenced several cardio-renal benefits of EMPA in non-diabetic patients through the involvement of biochemical pathways that are still to be deeply analysed. We aimed to evaluate the effects of EMPA on myocardial strain of non-diabetic mice treated with doxorubicin (DOXO) through the analysis of NLRP3 inflammasome and MyD88-related pathways resulting in anti-apoptotic and anti-fibrotic effects., Methods: Preliminary cellular studies were performed on mouse cardiomyocytes (HL-1 cell line) exposed to doxorubicin alone or combined to EMPA. The following analysis were performed: determination of cell viability (through a modified MTT assay), study of intracellular ROS production, lipid peroxidation (quantifying intracellular malondialdehyde and 4-hydroxynonenal), intracellular Ca
2+ homeostasis. Moreover, pro-inflammatory studies were also performed: expression of NLRP3 inflammasome, MyD88 myddosome and p65/NF-κB associated to secretion of cytokines involved in cardiotoxicity (Interleukins 1β, 8, 6). C57Bl/6 mice were untreated (Sham, n = 6) or treated for 10 days with doxorubicin (DOXO, n = 6), EMPA (EMPA, n = 6) or doxorubicin combined to EMPA (DOXO-EMPA, n = 6). DOXO was injected intraperitoneally. Ferroptosis and xanthine oxidase were studied before and after treatments. Cardiac function studies, including EF, FS and radial/longitudinal strain were analysed through transthoracic echocardiography (Vevo 2100). Cardiac fibrosis and apoptosis were histologically studied through Picrosirius red and TUNEL assay, respectively and quantified through pro-collagen-1α1, MMP-9 and Caspase-3 expression. Tissue NLRP3, MyD88 and cytokines were also quantified before and after treatments through ELISA methods., Results: Cardiomyocytes exposed to doxorubicin increased the intracellular Ca2+ content and expression of several pro-inflammatory markers associated to cell death; co-incubation with EMPA reduced significantly the magnitude of the effects. In preclinical study, EMPA increased EF and FS compared to DOXO groups (p < 0.05), prevented the reduction of radial and longitudinal strain after 10 days of treatment with doxorubicin (RS) 30.3% in EMPA-DOXO vs 15.7% in DOXO mice; LS - 17% in EMPA-DOXO vs - 11.7% in DOXO mice (p < 0.001 for both). Significant reductions in ferroptosis, xanthine oxidase expression, cardiac fibrosis and apoptosis in EMPA associated to DOXO were also seen. A reduced expression of pro-inflammatory cytokines, NLRP3, MyD88 and NF-kB in heart, liver and kidneys was also seen in DOXO-EMPA group compared to DOXO (p < 0.001)., Conclusion: EMPA reduced ferroptosis, fibrosis, apoptosis and inflammation in doxorubicin-treated mice through the involvement of NLRP3 and MyD88-related pathways, resulting in significant improvements in cardiac functions. These findings provides the proof of concept for translational studies designed to reduce adverse cardiovascular outcomes in non-diabetic cancer patients treated with doxorubicin., (© 2021. The Author(s).)- Published
- 2021
- Full Text
- View/download PDF
210. Takotsubo Cardiomyopathy as Epiphenomenon of Cardiotoxicity in Patients With Cancer: A Meta-summary of Case Reports.
- Author
-
Carbone A, Bottino R, Russo V, D'Andrea A, Liccardo B, Maurea N, Quagliariello V, Cimmino G, and Golino P
- Subjects
- Adult, Aged, Aged, 80 and over, Cardiotoxicity, Female, Humans, Male, Middle Aged, Prevalence, Prognosis, Risk Assessment, Risk Factors, Shock, Cardiogenic epidemiology, Shock, Cardiogenic physiopathology, Shock, Cardiogenic therapy, Takotsubo Cardiomyopathy epidemiology, Takotsubo Cardiomyopathy physiopathology, Takotsubo Cardiomyopathy therapy, Young Adult, Antineoplastic Agents adverse effects, Neoplasms drug therapy, Shock, Cardiogenic chemically induced, Takotsubo Cardiomyopathy chemically induced, Ventricular Function, Left drug effects
- Abstract
Abstract: Many antitumoral drugs have been linked to takotsubo cardiomyopathy, with no clear pathogenetic mechanisms. Data about this condition are lacking in literature. The aim of this meta-summary is to summarize the characteristics of patients with antitumoral drug-induced takotsubo cardiomyopathy, described in case reports available in literature. We searched for published case reports in PubMed, Google Scholar, EMBASE, and Scopus from 2009 about stress cardiomyopathy and antiblastic drugs. We selected 41 case reports. All cases underwent chemotherapy/immunotherapy for different types of cancer. The median age was 58 years, and 61% of them were women. The most common comorbidities were hypertension (12.2%) and dyslipidemia (4.9%), but most of the population had no cardiological clinical history. Takotsubo cardiomyopathy is associated to the 5-fluorouracil (36.5%), capecitabine (9.7%), trastuzumab (9.7%), and immune check point inhibitor (9.7%) treatment. The median time of onset was 2 days (1-150). Cardiogenic shock was the first manifestation in 11 patients (26.8%). Left ventricle ejection fraction recovery was showed in 33 patients (89%) with mean ejection fraction 57.7 ± 7%, after a median of 30-day (4-300) follow-up. Patients with cancer experienced takotsubo cardiomyopathy within few days from the beginning of therapy, and the most of them normalized the heart function in few weeks. Cardiogenic shock showed high prevalence in this setting of patients. Larger studies are needed to better understand the pathological mechanisms of antiblastic drug-induced stress cardiomyopathy, to find risk factors associated and preventive strategies for limit this type of cardiotoxicities., Competing Interests: The authors report no conflicts of interest., (Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
211. Endocannabinoid system expression in ovarian epithelial tumors according to the dualistic model of ovarian carcinogenesis.
- Author
-
Ronchi A, Grauso F, Zito Marino F, Quagliariello V, Maurea N, Facchini G, Montopoli M, Franco R, Berretta M, and Messalli EM
- Subjects
- Carcinoma, Ovarian Epithelial pathology, Female, Humans, Middle Aged, Ovarian Neoplasms pathology, Receptor, Cannabinoid, CB1 analysis, Carcinoma, Ovarian Epithelial metabolism, Ovarian Neoplasms metabolism, Receptor, Cannabinoid, CB1 biosynthesis
- Abstract
Objective: Our study aimed to confirm the expression of the endocannabinoid system in the human epithelial ovarian tumors, assessing the immunohistochemical expression of Cannabinoid Receptor Type 1 and Fatty Acid Amide Hydrolase in benign, borderline and malignant tumors., Materials and Methods: Cannabinoid Receptor Type 1 and Fatty Acid Amide Hydrolase immunohistochemical expression was determined in 118 epithelial ovarian tumors sequentially treated during the last decade in our department: 36 benign, 34 borderline and 48 malignant neoplasms. Cannabinoid Receptor type 1 and Fatty Acid Amide Hydrolase expression resulted predominantly weak-moderate in the benign and borderline forms., Results: concerning malignant tumors, Cannabinoid Receptor Type 1 expression resulted predominantly moderate-strong in Type I tumors and negative-weak in Type II tumors. Fatty Acid Amide Hydrolase expression resulted, instead, independent by the tumor types. Furthermore, there was no significant difference in the Cannabinoid Receptor Type 1 and Fatty Acid Amide Hydrolase expression relatively to the tumoral stages., Conclusions: The present study confirmed a variable expression of the endocannabinoid system in human ovarian tumors. Cannabinoid Receptor Type 1 expression was significantly different in malignant epithelial ovarian tumors according to dualistic model of ovarian carcinogenesis. Thus, in the most aggressive types II ovarian tumors, Cannabinoid Receptor Type 1 expression resulted predominantly negative or weak.
- Published
- 2021
- Full Text
- View/download PDF
212. Portrait of Italian Cardio-Oncology: Results of a Nationwide Associazione Nazionale Medici Cardiologi Ospedalieri (ANMCO) Survey.
- Author
-
Canale ML, Turazza F, Lestuzzi C, Parrini I, Camerini A, Russo G, Colivicchi F, Gabrielli D, Gulizia MM, Oliva S, Tarantini L, Maurea N, Rigacci L, Petrolati S, Casolo G, and Bisceglia I
- Abstract
Aims: Cardio-oncology has achieved a pivotal role in science, but real world data on its clinical impact are still limited. Methods: A questionnaire was sent out to all cardio-oncology services across Italy ( n = 120). The questionnaire was made up of 28 questions divided into four blocks: (A) general information on hospitals and service, (B) the inner organization of cardio-oncology and its relationships with out-of-hospital cardiologists and general practitioners, (C) educational needs and referral guidelines, and (D) activities/specific workload. Results: Ninety-six out of 120 (80%) completed the questionnaire; 9.4% were cancer centers while 90.6% were general hospitals. A cardio-oncology team was present in 56% of the cancer centers and in 20% only of general hospitals, and a cardio-oncology pathway was active in 55% of cancer centers and in just 14% of the general hospitals. Relationships with out-of-hospital cardiologists and general practitioners were lacking. The guidelines of reference were ESC and ANMCO/AIOM. Patients receiving anthracycline chemotherapy underwent scheduled monitoring by means of echocardiography in 58% of cases. Routine use of cardiac damage biomarkers was overall low, ranging from 22 to 33% while the use of global longitudinal strain reached 44%. Conclusions: Italian cardio-oncology showed a growing influence on clinical practice but still has room for improvement. Cardio-oncology teams are still scarce, and the application of dedicated paths is poor. The need for specific training has been highlighted., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Canale, Turazza, Lestuzzi, Parrini, Camerini, Russo, Colivicchi, Gabrielli, Gulizia, Oliva, Tarantini, Maurea, Rigacci, Petrolati, Casolo and Bisceglia.)
- Published
- 2021
- Full Text
- View/download PDF
213. Polydatin Reduces Cardiotoxicity and Enhances the Anticancer Effects of Sunitinib by Decreasing Pro-Oxidative Stress, Pro-Inflammatory Cytokines, and NLRP3 Inflammasome Expression.
- Author
-
Quagliariello V, Berretta M, Buccolo S, Iovine M, Paccone A, Cavalcanti E, Taibi R, Montopoli M, Botti G, and Maurea N
- Abstract
Renal cell carcinoma (RCC) represents the main renal tumors and are highly metastatic. Sunitinib, a recently-approved, multi-targeted Tyrosine Kinases Inhibitor (TKi), prolongs survival in patients with metastatic renal cell carcinoma and gastrointestinal stromal tumors, however a dose related cardiotoxicity was well described. Polydatin (3,4',5-trihydroxystilbene-3-β-d-glucoside) is a monocrystalline compound isolated from Polygonum cuspidatum with consolidated anti-oxidant and anti-inflammatory properties, however no studies investigated on its putative cardioprotective and chemosensitizing properties during incubation with sunitinib. We investigated on the effects of polydatin on the oxidative stress, NLRP3 inflammasome and Myd88 expression, highlighting on the production of cytokines and chemokines (IL-1β, IL-6, IL-8, CXCL-12 and TGF-β) during treatment with sunitinib. Exposure of cardiomyocytes and cardiomyoblasts (AC-16 and H9C2 cell lines) and human renal adenocarcinoma cells (769-P and A498) to polydatin combined to plasma-relevant concentrations of sunitinib reduces significantly iROS, MDA and LTB4 compared to only sunitinib-treated cells (P<0.001). In renal cancer cells and cardiomyocytes polydatin reduces expression of pro-inflammatory cytokines and chemokines involved in myocardial damages and chemoresistance and down-regulates the signaling pathway of NLRP3 inflammasome, MyD88 and NF-κB. Data of the present study, although in vitro , indicate that polydatin, besides reducing oxidative stress, reduces key chemokines involved in cancer cell survival, chemoresistance and cardiac damages of sunitinib through downregulation of NLRP3-MyD88 pathway, applying as a potential nutraceutical agent in preclinical studies of preventive cardio-oncology., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Quagliariello, Berretta, Buccolo, Iovine, Paccone, Cavalcanti, Taibi, Montopoli, Botti and Maurea.)
- Published
- 2021
- Full Text
- View/download PDF
214. Reasons why COVID-19 survivors should follow dietary World Cancer Research Fund/American Institute for Cancer Research (WCRF/AICR) recommendations: from hyper-inflammation to cardiac dysfunctions.
- Author
-
Quagliariello V, D'Aiuto G, Iaffaioli RV, Berretta M, Buccolo S, Iovine M, Paccone A, Cerrone F, Bonanno S, Nunnari G, Laganà N, Botti G, and Maurea N
- Subjects
- Alcohol Drinking, Body Weight, COVID-19 complications, COVID-19 virology, Carbonated Beverages, Cytokines metabolism, Guidelines as Topic, Humans, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Neoplasms complications, Prognosis, Red Meat, Risk Factors, SARS-CoV-2 isolation & purification, Survivors, COVID-19 pathology, Diet, Neoplasms pathology
- Abstract
The World Cancer Research Fund and American Institute for Cancer Research (WCRF/AICR) advise cancer survivors to follow their lifestyle recommendations for cancer prevention. Recent research indicates that a proper diet could exerts beneficial metabolic and immune effects in humans through the involvement of several, not yet properly known, metabolic pathways. Here, we argue that following WCRF/AICR recommendations could be a strategy to prevent cardiovascular outcomes [fulminant myocarditis, heart failure, venous thromboembolism (VTE)] and acute respiratory distress syndrome (ARDS) in patients during follow-up post COVID-19 infection. We discuss the metabolic effects of a WCRF/AICR based diet, highlighting on the involved cardio-metabolic pathways related on NLRP3 inflammasome-cytokines axis aimed to improve prognosis of COVID-19, especially in patients with cancer.
- Published
- 2021
- Full Text
- View/download PDF
215. Multiple Effects of Ascorbic Acid against Chronic Diseases: Updated Evidence from Preclinical and Clinical Studies.
- Author
-
Berretta M, Quagliariello V, Maurea N, Di Francia R, Sharifi S, Facchini G, Rinaldi L, Piezzo M, Manuela C, Nunnari G, and Montopoli M
- Abstract
Severe disease commonly manifests as a systemic inflammatory process. Inflammation is associated withthe enhanced production of reactive oxygen and nitrogen species and with a marked reduction in the plasma concentrations of protective antioxidant molecules. This imbalance gives rise to oxidative stress, which is greater in patients with more severe conditions such as sepsis, cancer, cardiovascular disease, acute respiratory distress syndrome, and burns. In these patients, oxidative stress can trigger cell, tissue, and organ damage, thus increasing morbidity and mortality. Ascorbic acid (ASC) is a key nutrient thatserves as an antioxidant and a cofactor for numerous enzymatic reactions. However, humans, unlike most mammals, are unable to synthesize it. Consequently, ASC must be obtained through dietary sources, especially fresh fruit and vegetables. The value of administering exogenous micronutrients, to reestablish antioxidant concentrations in patients with severe disease, has been recognized for decades. Despite the suggestion that ASC supplementation may reduce oxidative stress and prevent several chronic conditions, few large, randomized clinical trials have tested it in patients with severe illness. This article reviews the recent literature on the pharmacological profile of ASC and the role of its supplementation in critically ill patients.
- Published
- 2020
- Full Text
- View/download PDF
216. SARS-CoV-2 Infection and Cardioncology: From Cardiometabolic Risk Factors to Outcomes in Cancer Patients.
- Author
-
Quagliariello V, Bonelli A, Caronna A, Conforti G, Iovine M, Carbone A, Berretta M, Botti G, and Maurea N
- Abstract
The coronavirus disease-2019 (COVID-19) is a highly transmissible viral illness caused by SARS-CoV-2, which has been defined by the World Health Organization as a pandemic, considering its remarkable transmission speed worldwide. SARS-CoV-2 interacts with angiotensin-converting enzyme 2 and TMPRSS2, which is a serine protease both expressed in lungs, the gastro-intestinal tract, and cardiac myocytes. Patients with COVID-19 experienced adverse cardiac events (hypertension, venous thromboembolism, arrhythmia, myocardial injury, fulminant myocarditis), and patients with previous cardiovascular disease have a higher risk of death. Cancer patients are extremely vulnerable with a high risk of viral infection and more negative prognosis than healthy people, and the magnitude of effects depends on the type of cancer, recent chemotherapy, radiotherapy, or surgery and other concomitant comorbidities (diabetes, cardiovascular diseases, metabolic syndrome). Patients with active cancer or those treated with cardiotoxic therapies may have heart damages exacerbated by SARS-CoV-2 infection than non-cancer patients. We highlight the cardiovascular side effects of COVID-19 focusing on the main outcomes in cancer patients in updated perspective and retrospective studies. We focus on the main cardio-metabolic risk factors in non-cancer and cancer patients and provide recommendations aimed to reduce cardiovascular events, morbidity, and mortality.
- Published
- 2020
- Full Text
- View/download PDF
217. NLRP3 as Putative Marker of Ipilimumab-Induced Cardiotoxicity in the Presence of Hyperglycemia in Estrogen-Responsive and Triple-Negative Breast Cancer Cells.
- Author
-
Quagliariello V, De Laurentiis M, Cocco S, Rea G, Bonelli A, Caronna A, Lombari MC, Conforti G, Berretta M, Botti G, and Maurea N
- Subjects
- Antineoplastic Agents, Immunological adverse effects, Benzhydryl Compounds pharmacology, CTLA-4 Antigen metabolism, Cardiotoxicity metabolism, Cell Line, Tumor, Female, Glucose metabolism, Glucose pharmacology, Glucosides pharmacology, Humans, Hyperglycemia etiology, Leukotriene B4 metabolism, Myeloid Differentiation Factor 88 metabolism, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Sodium-Glucose Transporter 2 Inhibitors pharmacology, Transcription Factor RelA metabolism, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology, Biomarkers, Pharmacological metabolism, Cardiotoxicity etiology, Ipilimumab adverse effects, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Triple Negative Breast Neoplasms drug therapy
- Abstract
Hyperglycemia, obesity and metabolic syndrome are negative prognostic factors in breast cancer patients. Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, achieving unprecedented efficacy in multiple malignancies. However, ICIs are associated with immune-related adverse events involving cardiotoxicity. We aimed to study if hyperglycemia could affect ipilimumab-induced anticancer efficacy and enhance its cardiotoxicity. Human cardiomyocytes and estrogen-responsive and triple-negative breast cancer cells (MCF-7 and MDA-MB-231 cell lines) were exposed to ipilimumab under high glucose (25 mM); low glucose (5.5 mM); high glucose and co-administration of SGLT-2 inhibitor (empagliflozin); shifting from high glucose to low glucose. Study of cell viability and the expression of new putative biomarkers of cardiotoxicity and resistance to ICIs (NLRP3, MyD88, cytokines) were quantified through ELISA (Cayman Chemical) methods. Hyperglycemia during treatment with ipilimumab increased cardiotoxicity and reduced mortality of breast cancer cells in a manner that is sensitive to NLRP3. Notably, treatment with ipilimumab and empagliflozin under high glucose or shifting from high glucose to low glucose reduced significantly the magnitude of the effects, increasing responsiveness to ipilimumab and reducing cardiotoxicity. To our knowledge, this is the first evidence that hyperglycemia exacerbates ipilimumab-induced cardiotoxicity and decreases its anticancer efficacy in MCF-7 and MDA-MB-231 cells. This study sets the stage for further tests on other breast cancer cell lines and primary cardiomyocytes and for preclinical trials in mice aimed to decrease glucose through nutritional interventions or administration of gliflozines during treatment with ipilimumab.
- Published
- 2020
- Full Text
- View/download PDF
218. Evidences of CTLA-4 and PD-1 Blocking Agents-Induced Cardiotoxicity in Cellular and Preclinical Models.
- Author
-
Quagliariello V, Passariello M, Rea D, Barbieri A, Iovine M, Bonelli A, Caronna A, Botti G, De Lorenzo C, and Maurea N
- Abstract
Background: Several strategies based on immune checkpoint inhibitors (ICIs) have been developed for cancer therapy, opening to advantages in cancer outcomes. However, several ICI-induced side effects have emerged in these patients, especially a rare but clinically significant cardiotoxicity with high rate of mortality. We studied the cytotoxic and pro-inflammatory properties of Ipilimumab and Nivolumab, the underlying pathways and cytokine storm involved., Methods: Co-cultures of human cardiomyocytes and lymphocytes were exposed to Ipilimumab or Nivolumab; cell viability and expression of leukotrienes, NLRP3, MyD88, and p65/NF-kB were performed. C57 mice were treated with Ipilimumab (15 mg/kg); analysis of fractional shortening, ejection fraction, radial and longitudinal strain were made before and after treatments through 2D-echocardiography. Expression of NLRP3, MyD88, p65/NF-kB, and 12 cytokines were analyzed in murine myocardium., Results: Nivolumab and Ipilimumab exert effective anticancer, but also significant cardiotoxic effects in co-cultures of lymphocytes and tumor or cardiac cells. Both ICIs increased NLRP3, MyD88, and p65/NF-kB expression compared to untreated cells, however, the most pro-inflammatory and cardiotoxic effects were seen after exposure to Ipilimumab. Mice treated with Ipilimumab showed a significant decrease in fractional shortening and radial strain with respect to untreated mice, coupled with a significant increase in myocardial expression of NLRP3, MyD88, and several interleukins., Conclusions: Nivolumab and Ipilimumab exert cytotoxic effects mediated by the NLRP3/IL-1β and MyD88 pathways, leading to pro-inflammatory cytokine storm in heart tissue.
- Published
- 2020
- Full Text
- View/download PDF
219. Can Beta-2-Adrenergic Pathway Be a New Target to Combat SARS-CoV-2 Hyperinflammatory Syndrome?-Lessons Learned From Cancer.
- Author
-
Barbieri A, Robinson N, Palma G, Maurea N, Desiderio V, and Botti G
- Subjects
- Betacoronavirus drug effects, COVID-19, Cytokine Release Syndrome pathology, Humans, Inflammation drug therapy, Inflammation immunology, Inflammation pathology, Neoplasms drug therapy, Neoplasms pathology, Pandemics, Respiratory Distress Syndrome pathology, Respiratory Distress Syndrome virology, SARS-CoV-2, Th17 Cells immunology, Adrenergic beta-2 Receptor Antagonists therapeutic use, Coronavirus Infections drug therapy, Coronavirus Infections pathology, Cytokine Release Syndrome drug therapy, Pneumonia, Viral drug therapy, Pneumonia, Viral pathology, Receptors, Adrenergic, beta-2 drug effects, Respiratory Distress Syndrome drug therapy
- Abstract
SARS-CoV-2 infection is a new threat to global public health in the 21
st century (2020), which has now rapidly spread around the globe causing severe pneumonia often linked to Acute Respiratory Distress Syndrome (ARDS) and hyperinflammatory syndrome. SARS-CoV-2 is highly contagious through saliva droplets. The structural analysis suggests that the virus enters human cells through the ligation of the spike protein to angiotensin-converting enzyme 2 (ACE2 ). The progression of Covid-19 has been divided into three main stages: stage I-viral response, stage II-pulmonary phase, and stage III-hyperinflammation phase. Once the patients enter stage III, it will likely need ventilation and it becomes difficult to manage. Thus, it will be of paramount importance to find therapies to prevent or slow down the progression of the disease toward stage III. The key event leading to hyperinflammation seems to be the activation of Th-17 immunity response and Cytokine storm. B2 -adrenergic receptors (B2 ARs) are expressed on airways and on all the immune cells such as macrophages, dendritic cells, B and T lymphocytes. Blocking (B2 AR) has been proven, also in clinical settings, to reduce Th-17 response and negatively modulate inflammatory cytokines including IL-6 while increasing IFN γ . Non-selective beta-blockers are currently used to treat several diseases and have been proven to reduce stress-induced inflammation and reduce anxiety. For these reasons, we speculate that targeting B2 AR in the early phase of Covid-19 might be beneficial to prevent hyperinflammation., (Copyright © 2020 Barbieri, Robinson, Palma, Maurea, Desiderio and Botti.)- Published
- 2020
- Full Text
- View/download PDF
220. [HCF-ANMCO/AICPR/GIEC/ITAHFA/SICOA/SICP/SIMG/SIT Cardiological Societies Council Consensus document: Anticoagulant therapy in venous thromboembolism and atrial fibrillation of the patient with cancer. Current knowledge and new evidence].
- Author
-
Gulizia Chairperson MM, Parrini Co-Chairperson I, Colivicchi Co-Chairperson F, Bisceglia I, Caiazza F, Gensini GF, Mureddu GF, Santomauro M, Ageno W, Ambrosetti M, Aspromonte N, Barni S, Bellocci F, Caldarola P, Carletti M, De Luca L, Di Fusco SA, Di Lenarda A, Di Nisio M, Domenicucci S, Enea I, Francese GM, Lestuzzi C, Lucà F, Maurea N, Nassiacos D, Pedretti RFE, Pusineri E, Roscio G, Rossini R, Russo A, Volterrani M, and Gabrielli Co-Chairperson D
- Subjects
- Administration, Oral, Anticoagulants adverse effects, Antithrombins administration & dosage, Antithrombins adverse effects, Female, Hemorrhage chemically induced, Heparin, Low-Molecular-Weight administration & dosage, Heparin, Low-Molecular-Weight adverse effects, Humans, Male, Pulmonary Embolism prevention & control, Risk Factors, Anticoagulants administration & dosage, Atrial Fibrillation complications, Cardiology, Consensus, Neoplasms complications, Societies, Medical, Venous Thromboembolism prevention & control
- Abstract
Venous thromboembolism (VTE), including pulmonary embolism and deep venous thrombosis, either symptomatic or incidental, is a common complication in the history of cancer disease. The risk of VTE is 4-7-fold higher in oncology patients, and it represents the second leading cause of death, after cancer itself. In cancer patients, compared with the general population, VTE therapy is associated with higher rates of recurrent thrombosis and/or major bleeding. The need for treatment of VTE in patients with cancer is a challenge for the clinician because of the multiplicity of types of cancer, the disease stage and the imbricated cancer treatment. Historically, in cancer patients, low molecular weight heparins have been preferred for treatment of VTE. More recently, in large randomized clinical trials, direct oral anticoagulants (DOACs) demonstrated to reduce the risk of VTE. However, in the "real life", uncertainties remain on the use of DOACs, especially for the bleeding risk in patients with gastrointestinal cancers and the potential drug-to-drug interactions with specific anticancer therapies.In cancer patients, atrial fibrillation can arise as a perioperative complication or for the side effect of some chemotherapy agents, as well as a consequence of some associated risk factors, including cancer itself. The current clinical scores for predicting thrombotic events (CHA2DS2-VASc) or for predicting bleeding (HAS-BLED), used to guide antithrombotic therapy in the general population, have not yet been validated in cancer patients. Encouraging data for DOAC prescription in patients with atrial fibrillation and cancer are emerging: recent post-hoc analysis showed safety and efficacy of DOACs for the prevention of embolic events compared to warfarin in cancer patients. Currently, anticoagulant therapy of cancer patients should be individualized with multidisciplinary follow-up and frequent reassessment. This consensus document represents an advanced state of the art on the subject and provides useful notes on clinical practice.
- Published
- 2020
- Full Text
- View/download PDF
221. Randomized phase II study of valproic acid in combination with bevacizumab and oxaliplatin/fluoropyrimidine regimens in patients with RAS -mutated metastatic colorectal cancer: the REVOLUTION study protocol.
- Author
-
Avallone A, Piccirillo MC, Di Gennaro E, Romano C, Calabrese F, Roca MS, Tatangelo F, Granata V, Cassata A, Cavalcanti E, Maurea N, Maiolino P, Silvestro L, De Stefano A, Giuliani F, Rosati G, Tamburini E, Aprea P, Vicario V, Nappi A, Vitagliano C, Casaretti R, Leone A, Petrillo A, Botti G, Delrio P, Izzo F, Perrone F, and Budillon A
- Abstract
Background: Despite effective treatments, metastatic colorectal cancer (mCRC) prognosis is still poor, mostly in RAS -mutated tumors, thus suggesting the need for novel combinatorial therapies. Epigenetic alterations play an important role in initiation and progression of cancers, including CRC. Histone-deacetylase inhibitors (HDACi) have shown activity in combination with chemotherapy in the treatment of solid tumors. Owing to its HDACi activity and its safe use for epileptic disorders, valproic acid (VPA) is a good candidate for anticancer therapy that we have largely explored preclinically translating our findings in currently ongoing clinical studies. We have shown in CRC models that HDACi, including VPA, induces synergistic antitumor effects in combination with fluoropyrimidines. Furthermore, unpublished results from our group demonstrated that VPA induces differentiation and sensitization of CRC stem cells to oxaliplatin. Moreover, preclinical and clinical data suggest that HDACi may prevent/reverse anti-angiogenic resistance., Methods/design: A randomized, open-label, two-arm, multicenter phase-II study will be performed to explore whether the addition of VPA to first line bevacizumab/oxaliplatin/fluoropyrimidine regimens (mFOLFOX-6/mOXXEL) might improve progression-free survival (PFS) in RAS -mutated mCRC patients. A sample size of 200 patients was calculated under the hypothesis that the addition of VPA to chemotherapy/bevacizumab can improve PFS from 9 to 12 months, with one-sided alpha of 0.20 and a power of 0.80. Secondary endpoints are overall survival, objective response rate, metastases resection rate, toxicity, and quality of life. Moreover, the study will explore several prognostic and predictive biomarkers on blood samples, primary tumors, and on resected metastases., Discussion: The "Revolution" study aims to improve the treatment efficacy of RAS -mutated mCRC through an attractive strategy evaluating the combination of VPA with standard cancer treatment. Correlative studies could identify novel biomarkers and could add new insight in the mechanism of interaction between VPA, fluoropyrimidine, oxaliplatin, and bevacizumab., Trial Registration: EudraCT: 2018-001414-15; ClinicalTrials.gov identifier: NCT04310176., Competing Interests: Conflict of interest statement: The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article., (© The Author(s), 2020.)
- Published
- 2020
- Full Text
- View/download PDF
222. Incidence of Pericardial Effusion in Patients with Advanced Non-Small Cell Lung Cancer Receiving Immunotherapy.
- Author
-
Canale ML, Camerini A, Casolo G, Lilli A, Bisceglia I, Parrini I, Lestuzzi C, Del Meglio J, Puccetti C, Camerini L, Amoroso D, and Maurea N
- Subjects
- Adult, Aged, Aged, 80 and over, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Carcinoma, Non-Small-Cell Lung epidemiology, Female, Humans, Immune Checkpoint Inhibitors therapeutic use, Immunotherapy methods, Incidence, Lung Neoplasms epidemiology, Male, Middle Aged, Nivolumab therapeutic use, Pericardial Effusion epidemiology, Retrospective Studies, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents, Immunological adverse effects, Carcinoma, Non-Small-Cell Lung drug therapy, Immune Checkpoint Inhibitors adverse effects, Lung Neoplasms drug therapy, Nivolumab adverse effects, Pericardial Effusion chemically induced
- Abstract
Introduction: Cardiovascular toxicity of immunotherapy represents an underreported but potentially fatal side effect. A relatively high incidence of pericardial disease has been noticed in patients with non-small cell lung cancer (NSCLC)., Methods: We retrospectively analyzed a population of patients with advanced NSCLC receiving immune checkpoint inhibitors (ICIs) looking for the presence of pericardial effusion at baseline or during treatment. The study population was compared with a control group treated with chemotherapy. All patients were checked for the presence of concomitant pleural effusion., Results: We identify 60 patients (36 male/24 female, median age 70 years [range 43-81]). Prevalent histology was adenocarcinoma (65%) followed by squamous cell carcinoma (28%) and large cell or not otherwise specified (NOS) carcinoma (7%). Treatment consisted of nivolumab 3 mg/kg every 14 days (52 cases; 45 as second-line and 7 as third-line treatment) or pembrolizumab 200 mg (8 cases; all first-line treatment) for a total of 302 cycles delivered. Four out of 60 patients (6.7%) developed pericardial effusion during treatment, in two cases (3.3%) without concomitant pleural effusion, compared to 2 out of 60 (3.3%) in the control group in one case without concomitant pleural effusion (1.6%). Median time of onset was 40 days. Myocarditis was not observed., Conclusion: Our findings confirm pericardial effusion as a relatively frequent side effect of immunotherapy in NSCLC. Clinicians should be aware of this specific toxicity in patients with metastatic NSCLC receiving immunotherapy and refer to a cardiologist for a multidisciplinary approach.
- Published
- 2020
- Full Text
- View/download PDF
223. Resveratrol in Cancer Patients: From Bench to Bedside.
- Author
-
Berretta M, Bignucolo A, Di Francia R, Comello F, Facchini G, Ceccarelli M, Iaffaioli RV, Quagliariello V, and Maurea N
- Subjects
- Animals, Antineoplastic Agents, Phytogenic chemistry, Cardiovascular Diseases drug therapy, Chemical Phenomena, Clinical Studies as Topic, Drug Evaluation, Preclinical, Drug Interactions, Humans, Structure-Activity Relationship, Translational Research, Biomedical, Treatment Outcome, Antineoplastic Agents, Phytogenic pharmacology, Antineoplastic Agents, Phytogenic therapeutic use, Neoplasms drug therapy, Resveratrol pharmacology, Resveratrol therapeutic use
- Abstract
Resveratrol (3,5,4'-trihydroxystilbene) is a natural phytoalexin that accumulates in several vegetables and fruits like nuts, grapes, apples, red fruits, black olives, capers, red rice as well as red wines. Being both an extremely reactive molecule and capable to interact with cytoplasmic and nuclear proteins in human cells, resveratrol has been studied over the years as complementary and alternative medicine (CAM) for the therapy of cancer, metabolic and cardiovascular diseases like myocardial ischemia, myocarditis, cardiac hypertrophy and heart failure. This review will describe the main biological targets, cardiovascular outcomes, physico-chemical and pharmacokinetic properties of resveratrol in preclinical and clinical models implementing its potential use in cancer patients.
- Published
- 2020
- Full Text
- View/download PDF
224. Boswellic acid has anti-inflammatory effects and enhances the anticancer activities of Temozolomide and Afatinib, an irreversible ErbB family blocker, in human glioblastoma cells.
- Author
-
Barbarisi M, Barbarisi A, De Sena G, Armenia E, Aurilio C, Libutti M, Iaffaioli RV, Botti G, Maurea N, and Quagliariello V
- Subjects
- Anti-Inflammatory Agents pharmacology, Antioxidants pharmacology, Boswellia chemistry, Cardiotonic Agents pharmacology, Cell Line, Tumor, Chemokine CXCL12 metabolism, ErbB Receptors antagonists & inhibitors, Glioblastoma drug therapy, Humans, Interleukin-6 metabolism, Interleukin-8 metabolism, Leukotriene B4 metabolism, Lipid Peroxidation, Myocytes, Cardiac drug effects, Reactive Oxygen Species metabolism, Transcription Factor RelA metabolism, Afatinib pharmacology, Antineoplastic Agents, Phytogenic pharmacology, Glioblastoma pathology, Temozolomide pharmacology, Triterpenes pharmacology
- Published
- 2019
- Full Text
- View/download PDF
225. Oil Core-PEG Shell Nanocarriers for In Vivo MRI Imaging.
- Author
-
Calcagno V, Vecchione R, Quagliariello V, Marzola P, Busato A, Giustetto P, Profeta M, Gargiulo S, Cicco CD, Yu H, Cassani M, Maurea N, Mancini M, Pellegrino T, and Netti PA
- Subjects
- HT29 Cells, Humans, Indocyanine Green chemistry, Indocyanine Green pharmacology, MCF-7 Cells, Contrast Media chemistry, Contrast Media pharmacology, Drug Carriers chemistry, Ferric Compounds chemistry, Ferric Compounds pharmacology, Magnetic Resonance Imaging, Nanoparticles chemistry, Oils chemistry, Oils pharmacology, Polyethylene Glycols chemistry, Polyethylene Glycols pharmacology
- Abstract
Oil-in-water emulsions represent a promising carrier for in vivo imaging because of the possibility to convey poorly water-soluble species. To promote accumulation at the tumor site and prolong circulation time, reduction of carrier size and surface PEGylation plays a fundamental role. In this work a novel, simple method to design an oil-core/PEG-shell nanocarrier is reported. A PEG-shell is grown around a monodisperse oil-in-water nanoemulsion with a one-pot method, using the radical polymerization of poly(ethylene glycol)diacrylate. PEG polymerization is triggered by UV, obtaining a PEG-shell with tunable thickness. This core-shell nanosystem combines the eluding feature of the PEG with the ability to confine high payloads of lipophilic species. Indeed, the core is successfully loaded with a lipophilic contrast agent, namely super paramagnetic iron oxide nanocubes. Interestingly, it is demonstrated an in vitro and an in vivo MRI response of the nanocapsules. Additionally, when the nanosystem loaded with nanocubes is mixed with a fluorescent contrast agent, indo-cyanine green, a relevant in vitro photoacoustic effect is observed. Moreover, viability and cellular uptake studies show no significant cell cytotoxicity. These results, together with the choice of low cost materials and the scale up production, make this nanocarrier a potential platform for in vivo imaging., (© 2019 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2019
- Full Text
- View/download PDF
226. Costs of clinical trials with anticancer biological agents in an Oncologic Italian Cancer Center using the activity-based costing methodology.
- Author
-
Pascarella G, Capasso A, Nardone A, Triassi M, Pignata S, Arenare L, Ascierto P, Curvietto M, Maiolino P, D'Aniello R, Montanino A, Laudato F, De Feo G, Botti G, Perrone F, Petrillo A, Cavalcanti E, Lastoria S, Maurea N, and Morabito A
- Subjects
- Antineoplastic Agents, Immunological economics, Antineoplastic Agents, Immunological therapeutic use, Clinical Trials, Phase III as Topic economics, Drug Costs, Female, Humans, Italy, Male, Multicenter Studies as Topic economics, Antineoplastic Agents economics, Antineoplastic Agents therapeutic use, Health Care Costs, Neoplasms drug therapy, Neoplasms economics, Randomized Controlled Trials as Topic economics
- Abstract
Aim: The aim of the present study was to assess the estimated "per patient" total cost for a single Oncologic Italian Cancer Center participating in a multicenter clinical trial with new anticancer biological agents using the activity-based costing (ABC) methodology., Methodology: Nine randomized phase 3 clinical trials employing biological agents at the National Cancer Institute of Napoli, Italy, were analyzed to indentify "per patient" costs of each trial, according to the ABC methodology. The average consumption of resources for a patient completing the entire planned treatment was estimated for each trial. Through interviews of the personnel (doctors, nurses and technicians) and by analyses of the clinical trials protocols, the main activities of the 9 clinical trials were identified and, for each trial, the complete health care pathway of the patients and the treatment programmes were minutely reconstructed. Drug costs were not included because provided by Sponsors., Principal Findings: The average costs of the pre-study, treatment, monitoring, follow-up, audit, and administrative activities accounted for 2.357, 4.783, 700, 372, 1.263, and 9 Euro, respectively. The average total cost estimated for all "per patient" activities, including overhead costs, was 11.379 Euro. Staff costs accounted for € 5.988, while costs of diagnostic test accounted for 3.494 Euro. Clinical trials with immunotherapeutic drugs accounted for higher costs (+601 Euro as oncological staff costs, +1.318 Euro as intermediate services cost and +384 Euro as overheads)., Conclusions: The average total cost estimated for all "per patient" activities of a clinical trial with new anticancer biological agents was 11.379 Euro using the ABC methodology., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
227. Cardioprotective Effects of Nanoemulsions Loaded with Anti-Inflammatory Nutraceuticals against Doxorubicin-Induced Cardiotoxicity.
- Author
-
Quagliariello V, Vecchione R, Coppola C, Di Cicco C, De Capua A, Piscopo G, Paciello R, Narciso V, Formisano C, Taglialatela-Scafati O, Iaffaioli RV, Botti G, Netti PA, and Maurea N
- Subjects
- Animals, Antioxidants pharmacology, Cardiotoxicity, Cell Line, Cell Survival drug effects, Cytokines metabolism, Cytoprotection, Drug Compounding, Emulsions, Heart Diseases chemically induced, Heart Diseases metabolism, Heart Diseases pathology, Inflammation Mediators metabolism, Lipid Peroxidation drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Oxidative Stress drug effects, Rats, Anti-Inflammatory Agents pharmacology, Curcumin pharmacology, Dietary Supplements, Doxorubicin toxicity, Drug Carriers, Heart Diseases prevention & control, Lycopene pharmacology, Myocytes, Cardiac drug effects, Nanoparticles
- Abstract
Doxorubicin is a highly active antineoplastic agent, but its clinical use is limited because of its cardiotoxicity. Although nutraceuticals endowed with anti-inflammatory properties exert cardioprotective activity, their bioavailability and stability are inconsistent. In an attempt to address this issue, we evaluated whether bioavailable nanoemulsions loaded with nutraceuticals (curcumin and fresh and dry tomato extracts rich in lycopene) protect cardiomyoblasts (H9C2 cells) from doxorubicin-induced toxicity. Nanoemulsions were produced with a high-pressure homogenizer. H9C2 cells were incubated with nanoemulsions loaded with different nutraceuticals alone or in combination with doxorubicin. Cell viability was evaluated with a modified MTT method. The levels of the lipid peroxidation products malondialdehyde (MDA) and 4-hydroxy-2-butanone (4-HNA), and of the cardiotoxic-related interleukins IL-6, IL-8, IL-1β and IL-10, tumor necrosis factor-alpha (TNF-α), and nitric oxide were analyzed in cardiomyoblasts. The hydrodynamic size of nanoemulsions was around 100 nm. Cell viability enhancement was 35⁻40% higher in cardiomyoblasts treated with nanoemulsion + doxorubicin than in cardiomyoblasts treated with doxorubicin alone. Nanoemulsions also protected against oxidative stress as witnessed by a reduction of MDA and 4-HNA. Notably, nanoemulsions inhibited the release of IL-6, IL-8, IL-1β, TNF-α and nitric oxide by around 35⁻40% and increased IL-10 production by 25⁻27% versus cells not treated with emulsions. Of the nutraceuticals evaluated, lycopene-rich nanoemulsions had the best cardioprotective profile. In conclusion, nanoemulsions loaded with the nutraceuticals described herein protect against cardiotoxicity, by reducing inflammation and lipid oxidative stress. These results set the stage for studies in preclinical models.
- Published
- 2018
- Full Text
- View/download PDF
228. [Edoxaban in patients with atrial fibrillation and cancer].
- Author
-
Maurea N and Riva L
- Subjects
- Anticoagulants administration & dosage, Anticoagulants adverse effects, Anticoagulants pharmacology, Atrial Fibrillation complications, Atrial Fibrillation etiology, Cancer Survivors, Cardiovascular Diseases epidemiology, Cardiovascular Diseases etiology, Embolism etiology, Embolism prevention & control, Factor Xa Inhibitors administration & dosage, Factor Xa Inhibitors adverse effects, Hemorrhage chemically induced, Hemorrhage epidemiology, Humans, International Normalized Ratio, Pyridines adverse effects, Stroke etiology, Stroke prevention & control, Thiazoles adverse effects, Warfarin administration & dosage, Warfarin adverse effects, Atrial Fibrillation drug therapy, Neoplasms complications, Pyridines administration & dosage, Thiazoles administration & dosage
- Abstract
Advances in cancer therapy have led to a significant improvement of survival in most types of malignancies over the past few decades. As a result, there is a growing population of cancer survivors, expected to reach 18 million people in 2030 in the US and a similar number in Europe. Interestingly, cancer survivor studies have shown that although about half of these patients eventually die of cancer, one third of them actually die of cardiovascular disease. Arrhythmias represent a significant part of cardiovascular complications and atrial fibrillation is the main arrhythmia occurring in cancer patients.Antithrombotic therapy is a challenge: the optimal international normalized ratio (INR) level in patients on therapy with vitamin K antagonists is achieved in only 12% of them; in these patients, direct oral anticoagulants seem to be effective and safe for the prevention of stroke and systemic embolic events compared to warfarin and have similar risk of major bleeding. Among the trials, ENGAGE AF-TIMI 48 provides more data on the efficacy and safety of edoxaban in cancer patients.
- Published
- 2018
- Full Text
- View/download PDF
229. [Venous thromboembolism and atrial fibrillation in patients with cancer].
- Author
-
Maurea N and Riva L
- Subjects
- Atrial Fibrillation etiology, Humans, Neoplasms therapy, Risk Factors, Venous Thromboembolism etiology, Atrial Fibrillation epidemiology, Neoplasms complications, Venous Thromboembolism epidemiology
- Published
- 2018
- Full Text
- View/download PDF
230. Ranolazine Attenuates Trastuzumab-Induced Heart Dysfunction by Modulating ROS Production.
- Author
-
Riccio G, Antonucci S, Coppola C, D'Avino C, Piscopo G, Fiore D, Maurea C, Russo M, Rea D, Arra C, Condorelli G, Di Lisa F, Tocchetti CG, De Lorenzo C, and Maurea N
- Abstract
The ErbB2 blocker trastuzumab improves survival in oncologic patients, but can cause cardiotoxicity. The late Na+ current inhibitor ranolazine has been shown to counter experimental HF, including doxorubicin cardiotoxicity (a condition characterized by derangements in redox balance), by lowering the levels of reactive oxygen species (ROS). Since ErbB2 can modulate ROS signaling, we tested whether trastuzumab cardiotoxicity could be blunted by ranolazine via redox-mediated mechanisms. Trastuzumab decreased fractional shortening and ejection fraction in mice, but ranolazine prevented heart dysfunction when co-administered with trastuzumab. Trastuzumab cardiotoxicity was accompanied by elevations in natriuretic peptides and matrix metalloproteinase 2 (MMP2) mRNAs, which were not elevated with co-treatment with ranolazine. Trastuzumab also increased cleavage of caspase-3, indicating activation of the proapoptotic machinery. Again, ranolazine prevented this activation. Interestingly, Neonatal Rat Ventricular Myocytes (NRVMs), labeled with MitoTracker Red and treated with trastuzumab, showed only a small increase in ROS compared to baseline conditions. We then stressed trastuzumab-treated cells with the beta-agonist isoproterenol to increase workload, and we observed a significant increase of probe fluorescence, compared with cells treated with isoproterenol alone, reflecting induction of oxidative stress. These effects were blunted by ranolazine, supporting a role for I Na inhibition in the regulation of redox balance also in trastuzumab cardiotoxicity.
- Published
- 2018
- Full Text
- View/download PDF
231. Management of QT prolongation induced by anti-cancer drugs: Target therapy and old agents. Different algorithms for different drugs.
- Author
-
Coppola C, Rienzo A, Piscopo G, Barbieri A, Arra C, and Maurea N
- Subjects
- Algorithms, Humans, Antineoplastic Agents adverse effects, Arrhythmias, Cardiac chemically induced, Cardiotoxicity etiology, Drug-Related Side Effects and Adverse Reactions etiology
- Abstract
The side effects of anticancer drugs still play a critical role in survival and quality of life. Although the recent progresses of cancer therapies have significantly improved the prognosis of oncologic patients, side effects of antineoplastic treatments are still responsible for the increased mortality of cancer survivors. Cardiovascular toxicity is the most dangerous adverse effect induced by anticancer therapies. A survey conducted by the National Health and Nutrition Examination, showed that 1807 cancer survivors followed up for seven years: 51% died of cancer and 33% of heart disease (Vejpongsa and Yeh, 2014). Moreover, the risk of cardiotoxicity persists even with the targeted therapy, the newer type of cancer treatment, due to the presence of on-target and off-target effects related to this new class of drugs. The potential cardiovascular toxicity of anticancer agents includes: QT prolongation, arrhythmias, myocardial ischemia, stroke, hypertension (HTN), thromboembolism, left ventricular dysfunction and heart failure (HF). Compared to other cardiovascular disorders, the interest in QT prolongation and its complications is fairly recent. However, oncologists have to deal with it and to evaluate the risk-benefit ratio before starting the treatment or during the same. Electrolyte abnormalities, low levels of serum potassium and several drugs may favour the acquired QT prolongation. Treatment of marked QT prolongation includes cardiac monitoring, caution in the use or suspension of cancer drugs and correction of electrolyte abnormalities (hypokalaemia, hypomagnesaemia, hypocalcaemia). Syndrome of QT prolongation can be associated with potentially fatal cardiac arrhythmias and its treatment consists of intravenous administration of magnesium sulphate and the use of electrical cardioversion., (Copyright © 2017 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
232. Enhanced Drug Delivery into Cell Cytosol via Glycoprotein H-Derived Peptide Conjugated Nanoemulsions.
- Author
-
Fotticchia T, Vecchione R, Scognamiglio PL, Guarnieri D, Calcagno V, Di Natale C, Attanasio C, De Gregorio M, Di Cicco C, Quagliariello V, Maurea N, Barbieri A, Arra C, Raiola L, Iaffaioli RV, and Netti PA
- Subjects
- Curcumin chemistry, Cytosol chemistry, Drug Carriers chemistry, Emulsions chemistry, Emulsions metabolism, Human Umbilical Vein Endothelial Cells chemistry, Human Umbilical Vein Endothelial Cells cytology, Humans, Molecular Structure, Peptides chemistry, Viral Proteins chemistry, Curcumin pharmacokinetics, Cytosol metabolism, Drug Delivery Systems, Human Umbilical Vein Endothelial Cells metabolism, Nanoparticles chemistry, Peptides metabolism, Viral Proteins metabolism
- Abstract
The key role of nanocarriers in improving the pharmacological properties of commonly used drugs is recognized worldwide. It is also known that in the development of new effective nanocarriers the use of targeting moieties integrated on their surface is essential. Herein, we propose a nanocarrier based on an oil in water nanoemulsion coated with a membranotropic peptide derived from the glycoprotein H of Herpes simplex virus 1, known as gH625, in order to reduce endolysosomal accumulation and to enhance cytosolic localization. In addition, we show an enhanced anti-inflammatory activity of curcumin, a bioactive compound isolated from the Curcuma longa plant, when loaded into our engineered nanocarriers. This effect is a consequence of a higher uptake combined with a high curcumin preservation exerted by the active nanocapsules compared to control ones. When loaded into our nanocapsules, indeed, curcumin molecules are directly internalized into the cytosol rather than into lysosomes. Further, in order to extend the in vitro experimental setting with a more complex model and to explore the possibility to use our nanocarriers for further biological applications, we tested their performance in a 3D sprouting angiogenesis model. Finally, we show promising preliminary in vivo results by assessing the anti-inflammatory properties of the proposed nanocarrier.
- Published
- 2017
- Full Text
- View/download PDF
233. Intracardiac metastasis originated from chondrosarcoma.
- Author
-
Maurea N, Ragone G, Coppola C, Caronna A, Tocchetti CG, Agozzino L, Apice G, and Iaffaioli RV
- Subjects
- Adolescent, Bone Neoplasms therapy, Chondrosarcoma diagnostic imaging, Chondrosarcoma therapy, Diagnosis, Differential, Disease Progression, Echocardiography, Fatal Outcome, Heart Atria pathology, Heart Neoplasms diagnostic imaging, Heart Neoplasms therapy, Humans, Male, Predictive Value of Tests, Treatment Outcome, Bone Neoplasms pathology, Chondrosarcoma secondary, Heart Neoplasms secondary, Scapula pathology
- Abstract
Primary cardiac tumors are extremely rare. By comparison, metastatic involvement of the heart is over 20 times more common and has been reported in autopsy series in up to one in five patients dying of cancer. Cardiac metastasis of chondrosarcoma is absolutely not frequent. In the recent literature, a cardiac metastasis from chondrosarcoma has never been described. We report the case of an 18-year-old man with a diagnosis of cardiac metastasis that originated from a left scapular chondrosarcoma. Chondrosarcoma is a skeletal tumor with various grades of malignancy, rapidly evolving, and with a strong tendency to metastasize, with low responsiveness to chemotherapy. The onset of characteristic systemic symptoms in the late stage of the disease led to the diagnosis of a mass localized in the right atrium. Management and differential diagnosis of infective heart lesions were also very complex in a rapidly evolving life-threatening condition.
- Published
- 2017
- Full Text
- View/download PDF
234. ANMCO/AIOM/AICO Consensus Document on clinical and management pathways of cardio-oncology: executive summary.
- Author
-
Tarantini L, Gulizia MM, Di Lenarda A, Maurea N, Giuseppe Abrignani M, Bisceglia I, Bovelli D, De Gennaro L, Del Sindaco D, Macera F, Parrini I, Radini D, Russo G, Beatrice Scardovi A, and Inno A
- Abstract
Cardiovascular disease and cancer are leading causes of death. Both diseases share the same risk factors and, having the highest incidence and prevalence in the elderly, they often coexist in the same individual. Furthermore, the enhanced survival of cancer patients registered in the last decades and linked to early diagnosis and improvement of care, not infrequently exposes them to the appearance of ominous cardiovascular complications due to the deleterious effects of cancer treatment on the heart and circulatory system. The above considerations have led to the development of a new branch of clinical cardiology based on the principles of multidisciplinary collaboration between cardiologists and oncologists: Cardio-oncology, which aims to find solutions to the prevention, monitoring, diagnosis and treatment of heart damage induced by cancer care in order to pursue, in the individual patient, the best possible care for cancer while minimizing the risk of cardiac toxicity. In this consensus document we provide practical recommendations on how to assess, monitor, treat and supervise the candidate or patient treated with potentially cardiotoxic cancer therapy in order to treat cancer and protect the heart at all stages of the oncological disease. Cardiovascular diseases and cancer often share the same risk factors and can coexist in the same individual. Such possibility is amplified by the deleterious effects of cancer treatment on the heart. The above considerations have led to the development of a new branch of clinical cardiology, based on multidisciplinary collaboration between cardiologist and oncologist: the cardio-oncology. It aims to prevent, monitor, and treat heart damages induced by cancer therapies in order to achieve the most effective cancer treatment, while minimizing the risk of cardiac toxicity. In this paper, we provide practical recommendations on how to assess, monitor, treat and supervise patients treated with potential cardiotoxic cancer therapies.
- Published
- 2017
- Full Text
- View/download PDF
235. Does Accidental Overcorrection of Symptomatic Hyponatremia in Chronic Heart Failure Require Specific Therapeutic Adjustments for Preventing Central Pontine Myelinolysis?
- Author
-
De Vecchis R, Noutsias M, Ariano C, Cesaro A, Cioppa C, Giasi A, and Maurea N
- Abstract
This review aims at summarizing essential aspects of epidemiology and pathophysiology of hyponatremia in chronic heart failure (CHF), to set the ground for a practical as well as evidence-based approach to treatment. As a guide through the discussion of the available evidence, a clinical case of hyponatremia associated with CHF is presented. For this case, the severe neurological signs at presentation justified an emergency treatment with hypertonic saline plus furosemide, as indicated. Subsequently, as the neurological emergency began to subside, the reversion of the trend toward hyponatremia overcorrection was realized by continuous infusion of hypotonic solutions, and administration of desmopressin, so as to prevent the very feared risk of an osmotic demyelination syndrome. This very disabling complication of the hyponatremia correction is then briefly outlined. Moreover, the possible advantages related to systematic correction of the hyponatremia that occurs in the course of CHF are mentioned. Additionally, the case of tolvaptan, a vasopressin receptor antagonist, is concisely presented in order to underline the different views that have led to different norms in Europe with respect to the USA or Japan as regards the use of this drug as a therapeutic resource against the hyponatremia.
- Published
- 2017
- Full Text
- View/download PDF
236. The Impact Exerted on Clinical Outcomes of Patients With Chronic Heart Failure by Aldosterone Receptor Antagonists: A Meta-Analysis of Randomized Controlled Trials.
- Author
-
De Vecchis R, Cantatrione C, Mazzei D, Barone A, and Maurea N
- Abstract
Background: Aldosterone receptor antagonists (ARAs) have been associated with improved clinical outcomes in patients with heart failure with reduced left ventricular ejection fraction (HFREF), but not in those with heart failure with preserved left ventricular ejection fraction (HFpEF). With the aim to study this topic more deeply, we carried out a meta-analysis of selective and non-selective ARAs in HFREF and HFpEF., Methods: We searched PubMed and Scopus databases. We decided to incorporate in the meta-analysis only randomized controlled trials (RCTs) of ARAs in patients with chronic heart failure (CHF) if they met the following criteria: experimental groups included patients with CHF treated with ARAs in addition to the conventional therapy; control groups included patients with CHF receiving conventional therapy without ARAs. Outcomes of interest were all-cause death, hospitalizations from cardiovascular cause, hyperkalemia, or gynecomastia., Results: We detected 15 studies representing 15,671 patients. ARAs were associated with a reduced odds of all-cause death (odds ratio (OR): 0.79; 95% confidence interval (CI): 0.73 - 0.87) and hospitalizations from cardiovascular cause (OR: 0.73; 95% CI: 0.61 - 0.89). However, subgroup analysis showed that these advantages were limited to HFREF (all-cause death: OR: 0.77, 95% CI: 0.69 - 0.84; hospitalizations from cardiovascular cause: OR: 0.66, 95% CI: 0.51 - 0.85), but they did not affect the HFpEF group (all-cause death: OR: 0.91, 95% CI: 0.76 - 1.1; hospitalizations from cardiovascular cause: OR: 0.85, 95% CI: 0.7 - 1.09). ARAs increased the risk of hyperkalemia (OR: 2.17; 95% CI: 1.88 - 2.5). Non-selective ARAs, but not selective ARAs, increased the risk of gynecomastia (OR: 8.22, 95% CI: 4.9 - 13.81 vs. OR: 0.74, 95% CI: 0.43 - 1.27)., Conclusions: ARAs reduced the risk of adverse cardiac events in HFREF but not HFpEF. In particular, ARA use in HFpEF patients is questionable, since in this CHF type, no significant improvement in all-cause death and cardiovascular hospitalizations was demonstrated with ARA treatment, in the face of the well-known risks of hyperkalemia and/or gynecomastia that chronic ARA therapy entails. Selective ARAs were equally effective as non-selective ARAs, without the risk of gynecomastia.
- Published
- 2017
- Full Text
- View/download PDF
237. [ANMCO/AICO/AIOM Consensus document: Clinical and management pathways in cardio-oncology].
- Author
-
Tarantini L, Gulizia MM, Di Lenarda A, Maurea N, Abrignani MG, Bisceglia I, Bovelli D, De Gennaro L, Del Sindaco D, Macera F, Parrini I, Radini D, Russo G, Scardovi AB, and Inno A
- Subjects
- Consensus, Humans, Interdisciplinary Communication, Italy, Needs Assessment, Patient Care Team, Risk Assessment, Risk Factors, Antineoplastic Agents adverse effects, Cardiology, Heart Diseases diagnosis, Heart Diseases etiology, Heart Diseases prevention & control, Heart Diseases therapy, Medical Oncology, Neoplasms complications, Neoplasms diagnosis, Neoplasms prevention & control, Neoplasms therapy
- Abstract
In Italy, cardiovascular diseases and cancer are the leading causes of death. Both diseases share the same risk factors and, having the highest incidence and prevalence in the elderly, they often coexist in the same individual. Furthermore, the enhanced survival of cancer patients registered in the last decades and linked to early diagnosis and improvement of care, not infrequently exposes them to the appearance of ominous cardiovascular complications due to the deleterious effects of cancer treatment on the heart and circulatory system. The above considerations have led to the development of a new branch of clinical cardiology based on the principles of multidisciplinary collaboration between cardiologists and oncologists: Cardio-oncology, which aims to find solutions to the prevention, monitoring, diagnosis and treatment of heart damage induced by cancer care in order to pursue, in the individual patient, the best possible care for cancer while minimizing the risk of cardiac toxicity. In this consensus document we provide practical recommendations on how to assess, monitor, treat and supervise the candidate or patient treated with potentially cardiotoxic cancer therapy in order to treat cancer and protect the heart at all stages of the oncological disease.
- Published
- 2017
- Full Text
- View/download PDF
238. Trastuzumab and target-therapy side effects: Is still valid to differentiate anthracycline Type I from Type II cardiomyopathies?
- Author
-
Riccio G, Coppola C, Piscopo G, Capasso I, Maurea C, Esposito E, De Lorenzo C, and Maurea N
- Subjects
- Anthracyclines administration & dosage, Anthracyclines adverse effects, Antineoplastic Agents administration & dosage, Breast Neoplasms drug therapy, Cardiomyopathies etiology, Cardiotoxicity etiology, Clinical Trials as Topic, Female, Heart Failure etiology, Humans, Immunotherapy adverse effects, Retrospective Studies, Trastuzumab administration & dosage, Antineoplastic Agents adverse effects, Cardiomyopathies drug therapy, Trastuzumab adverse effects
- Abstract
The improvement in cancer therapy and the increasing number of long term survivors unearth the issue of cardiovascular side effects of anticancer treatments. As a paradox in cancer survivors, delayed cardiotoxicity has emerged as a significant problem. Two categories of cardiotoxic side effects of antineoplastic drugs have been previously proposed: Type I cardiotoxicity, defined as permanent cardiotoxicity, is usually caused by anthracyclines; Type II cardiotoxicity, considered as reversible cardiotoxicity, has been mainly related to monoclonal antibodies. The cardiotoxicity of antibodies has been associated to trastuzumab, a humanized anti-ErbB2 monoclonal antibody currently in clinical use for the therapy of breast carcinomas, which induces cardiac dysfunction when used in monotherapy, or in combination with anthracyclines. Furthermore, recent retrospective studies have shown an increased incidence of heart failure and/or cardiomyopathy in patients treated with trastuzumab, that can persist many years after the conclusion of the therapy, thus suggesting that the side toxic effects are not always reversible as it was initially proposed. On the other hand, early detection and prompt therapy of anthracycline associated cardiotoxicity can lead to substantial recovery of cardiac function. On the basis of these observations, we propose to find a new different classification for cardiotoxic side effects of drugs used in cancer therapy.
- Published
- 2016
- Full Text
- View/download PDF
239. A recommended practical approach to the management of anthracycline-based chemotherapy cardiotoxicity: an opinion paper of the working group on drug cardiotoxicity and cardioprotection, Italian Society of Cardiology.
- Author
-
Spallarossa P, Maurea N, Cadeddu C, Madonna R, Mele D, Monte I, Novo G, Pagliaro P, Pepe A, Tocchetti CG, Zito C, and Mercuro G
- Subjects
- Cardiologists education, Cardiotoxicity prevention & control, Cardiovascular Diseases physiopathology, Humans, Italy, Neoplasms drug therapy, Risk Assessment, Societies, Medical, Antineoplastic Agents adverse effects, Cardiovascular Diseases chemically induced, Cardiovascular Diseases prevention & control, Disease Management
- Abstract
Anthracyclines are the mainstay of treatment of a variety of haematological malignancies and solid tumours. Unfortunately, the clinical use of these drugs is limited by cumulative, dose-related cardiotoxicity which may ultimately lead to a severe and irreversible form of cardiomyopathy. Thus, there is an increasing need for close cooperation among cardiologists, oncologists and haemato-oncologists. As anthracyclines save lives, the logical goal of this cooperation, besides preventing or mitigating cardiotoxicity, is to promote an acceptable balance between the potential cardiac side effects and the vital benefit of anticancer treatment. This manuscript, which is specifically addressed to the cardiologist who has not accumulated much experience in the field of cancer therapy, focuses on several topics, that is old and new mechanisms of cardiac toxicity, late cardiac toxicity, the importance of overall risk assessment, the key role of a cardiology consult before starting cancer therapy, and the pros and cons of primary and secondary prevention programmes.
- Published
- 2016
- Full Text
- View/download PDF
240. Strain Analysis in the Assessment of a Mouse Model of Cardiotoxicity due to Chemotherapy: Sample for Preclinical Research.
- Author
-
Rea D, Coppola C, Barbieri A, Monti MG, Misso G, Palma G, Bimonte S, Zarone MR, Luciano A, Liccardo D, Maiolino P, Cittadini A, Ciliberto G, Arra C, and Maurea N
- Subjects
- Animals, Antibiotics, Antineoplastic adverse effects, Apoptosis drug effects, Biomechanical Phenomena, Cardiomyopathies chemically induced, Cardiomyopathies diagnosis, Cardiotoxicity diagnosis, Cardiotoxicity etiology, Disease Models, Animal, Echocardiography, Female, Humans, Mice, Inbred C57BL, Myocytes, Cardiac drug effects, Myocytes, Cardiac pathology, Time Factors, Cardiomyopathies physiopathology, Cardiotoxicity physiopathology, Doxorubicin adverse effects, Myocardial Contraction drug effects
- Abstract
Background: In recent years, the development of more effective anticancer drugs has provided great benefits in patients' quality of life by improving both prognosis and disease-free survival. Nevertheless, the frequency and severity of side-effects, with particular reference to cardiac toxicity, have gained particular attention. The purpose of this study was to create a precise and sensitive preclinical model, able to identify early contractile dysfunction in mice treated with chemotherapy, through use of speckle-tracking echocardiography., Materials and Methods: We generated a mouse model of cardiotoxicity induced by doxorubicin. C57BL 6 mice were divided into two groups, treated for 7 days by intraperitoneal injections of placebo (vehicle) or doxorubicin (2.17 mg/kg), in order to characterize the cardiac phenotype in vivo., Results: We demonstrated that doxorubicin caused ealy remodeling of the left ventricle: after two days of therapy, the radial, circumferential and strain rates were reduced respectively by 35%, 34%, and 39% (p-value ≤0.001). Moreover, histological analysis revealed that doxorubicin treatment increased fibrosis, cardiomyocyte diameter and apoptosis., Conclusion: In a murine model of doxorubicin-induced cardiac injury, we detected left ventricular dysfunction followed by alterations in conventional echocardiographic indices. Our study suggests that a change in strain could be an effective early marker of myocardial dysfunction for new anticancer treatments and, in preclinical studies, it might also be a valuable indicator for the assessment of activity of cardioprotective agents., (Copyright © 2016 International Institute of Anticancer Research (Dr. John G. Delinassios), All rights reserved.)
- Published
- 2016
241. Pathophysiology of anthracycline cardiotoxicity.
- Author
-
Mele D, Tocchetti CG, Pagliaro P, Madonna R, Novo G, Pepe A, Zito C, Maurea N, and Spallarossa P
- Subjects
- Cardiotoxicity physiopathology, Cardiovascular Diseases genetics, Cardiovascular Diseases prevention & control, Dose-Response Relationship, Drug, Genetic Predisposition to Disease, Heart drug effects, Humans, Neoplasms drug therapy, Anthracyclines adverse effects, Antineoplastic Agents adverse effects, Cardiovascular Diseases chemically induced, Heart physiopathology
- Abstract
Anthracyclines (ANTs) are powerful drugs that have reduced the mortality of cancer patients. However, their use is limited by the development of cardiotoxicity (CTX), which is dose dependent and may lead to left ventricular dysfunction and heart failure. Although various strategies have been suggested to reduce the negative effects of ANTs, CTX is still an important unresolved clinical issue. This may be due at least partly to the incomplete characterization of the molecular and cellular mechanisms of ANT-induced CTX. In addition, although various forms of cardiac damage have been demonstrated with the use of these drugs in experimental studies, it is not yet clear how these translate to the clinical setting. Appropriate characterization of potential candidates for ANT-based therapies is essential to decide whether to administer these drugs. Hopefully, new information from genetic profiling will help to identify patients who are at high risk of developing CTX.
- Published
- 2016
- Full Text
- View/download PDF
242. A recommended practical approach to the management of target therapy and angiogenesis inhibitors cardiotoxicity: an opinion paper of the working group on drug cardiotoxicity and cardioprotection, Italian Society of Cardiology.
- Author
-
Maurea N, Spallarossa P, Cadeddu C, Madonna R, Mele D, Monte I, Novo G, Pagliaro P, Pepe A, Tocchetti CG, Zito C, and Mercuro G
- Subjects
- Cardiotoxicity prevention & control, Cardiovascular Diseases physiopathology, Humans, Italy, Monitoring, Physiologic, Neoplasms drug therapy, Practice Guidelines as Topic, Risk Assessment, Societies, Medical, Angiogenesis Inhibitors adverse effects, Cardiovascular Diseases chemically induced, Cardiovascular Diseases prevention & control, Disease Management, Molecular Targeted Therapy adverse effects
- Abstract
The US National Cancer Institute estimates that cardiotoxicity (CTX) from target therapy refers mostly to four groups of drugs: epidermal growth factor receptor 2 inhibitors, angiogenic inhibitors, directed Abelson murine leukemia viral oncogene homolog inhibitors, and proteasome inhibitors. The main cardiotoxic side-effects related to antiepidermal growth factor receptor 2 therapy are left ventricular systolic dysfunction and heart failure. Angiogenesis inhibitors are associated with hypertension, left ventricular dysfunction/heart failure, myocardial ischemia, QT prolongation, and thrombosis. Moreover, other agents may be related to CTX induced by treatment. In this study, we review the guidelines for a practical approach for the management of CTX in patients under anticancer target therapy.
- Published
- 2016
- Full Text
- View/download PDF
243. Pathophysiology of cardiotoxicity from target therapy and angiogenesis inhibitors.
- Author
-
Maurea N, Coppola C, Piscopo G, Galletta F, Riccio G, Esposito E, De Lorenzo C, De Laurentiis M, Spallarossa P, and Mercuro G
- Subjects
- Antibodies, Monoclonal adverse effects, Cardiotoxicity physiopathology, Cardiovascular Diseases prevention & control, Heart drug effects, Humans, Neoplasms drug therapy, Proteasome Inhibitors adverse effects, Receptor, ErbB-2 antagonists & inhibitors, Vascular Endothelial Growth Factors adverse effects, Angiogenesis Inhibitors adverse effects, Cardiovascular Diseases chemically induced, Heart physiopathology, Molecular Targeted Therapy adverse effects
- Abstract
The progress in cancer therapy and the increase in number of long-term survivors reveal the issue of cardiovascular side-effects of anticancer drugs. Cardiotoxicity has become a significant problem, and the risks of adverse cardiac events induced by systemic drugs need to be seriously considered. Potential cardiovascular toxicities linked to anticancer agents include arrhythmias, myocardial ischemia and infarction, hypertension, thromboembolism, left ventricular dysfunction, and heart failure. It has been shown that several anticancer drugs seriously affect the cardiovascular system, such as ErbB2 inhibitors, vascular endothelial growth factor (VEGF) inhibitors, multitargeted kinase inhibitors, Abelson murine leukemia viral oncogene homolog inhibitors, and others. Each of these agents has a different mechanism through which it affects the cardiovascular system. ErbB2 inhibitors block the ErbB4/ErbB2 heterodimerization pathway triggered by Neuregulin-1, which is essential for cardiomyocyte survival. VEGF signaling is crucial for vascular growth, but it also has a major impact on myocardial function, and the VEGF pathway is also essential for maintenance of cardiovascular homeostasis. Drugs that inhibit the VEGF signaling pathway lead to a net reduction in capillary density and loss of contractile function. Here, we review the mechanisms and pathophysiology of the most significant cardiotoxic effects of ErbB2 inhibitors and antiangiogenic drugs. Moreover, we highlight the role of cardioncology in recognizing these toxicities, developing strategies to prevent or minimize cardiovascular toxicity, and reducing long-term cardiotoxic effects.
- Published
- 2016
- Full Text
- View/download PDF
244. WITHDRAWN:A Practical Approach for Management of QT Prolongation Induced by Anticancer Drugs.
- Author
-
Maurea N, Coppola C, Piscopo G, Galletta F, Riccio G, and De Lorenzo C
- Abstract
Ahead of Print article withdrawn by publisher., (©AlphaMed Press.)
- Published
- 2016
- Full Text
- View/download PDF
245. Phase 1/2 study of valproic acid and short-course radiotherapy plus capecitabine as preoperative treatment in low-moderate risk rectal cancer-V-shoRT-R3 (Valproic acid--short Radiotherapy--rectum 3rd trial).
- Author
-
Avallone A, Piccirillo MC, Delrio P, Pecori B, Di Gennaro E, Aloj L, Tatangelo F, D'Angelo V, Granata C, Cavalcanti E, Maurea N, Maiolino P, Bianco F, Montano M, Silvestro L, Terranova Barberio M, Roca MS, Di Maio M, Marone P, Botti G, Petrillo A, Daniele G, Lastoria S, Iaffaioli VR, Romano G, Caracò C, Muto P, Gallo C, Perrone F, and Budillon A
- Subjects
- Adolescent, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols adverse effects, Capecitabine, Combined Modality Therapy, Deoxycytidine administration & dosage, Deoxycytidine adverse effects, Deoxycytidine analogs & derivatives, Female, Fluorouracil administration & dosage, Fluorouracil adverse effects, Fluorouracil analogs & derivatives, Humans, Male, Middle Aged, Preoperative Care, Radiotherapy methods, Rectal Neoplasms drug therapy, Rectal Neoplasms radiotherapy, Rectal Neoplasms surgery, Research Design, Valproic Acid administration & dosage, Valproic Acid adverse effects, Young Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Rectal Neoplasms therapy
- Abstract
Background: Locally advanced rectal cancer (LARC) is a heterogeneous group of tumors where a risk-adapted therapeutic strategy is needed. Short-course radiotherapy (SCRT) is a more convenient option for LARC patients than preoperative long-course RT plus capecitabine. Histone-deacetylase inhibitors (HDACi) have shown activity in combination with RT and chemotherapy in the treatment of solid tumors. Valproic acid (VPA) is an anti-epileptic drug with HDACi and anticancer activity. In preclinical studies, our group showed that the addition of HDACi, including VPA, to capecitabine produces synergistic antitumour effects by up-regulating thymidine phosphorylase (TP), the key enzyme converting capecitabine to 5-FU, and by downregulating thymidylate synthase (TS), the 5-FU target., Methods/design: Two parallel phase-1 studies will assess the safety of preoperative SCRT (5 fractions each of 5 Gy, on days 1 to 5) combined with (a) capecitabine alone (increasing dose levels: 500-825 mg/m2/bid), on days 1-21, or (b) capecitabine as above plus VPA (oral daily day -14 to 21, with an intra-patient titration for a target serum level of 50-100 microg/ml) followed by surgery 8 weeks after the end of SCRT, in low-moderate risk RC patients. Also, a randomized phase-2 study will be performed to explore whether the addition of VPA and/or capecitabine to preoperative SCRT might increase pathologic complete tumor regression (TRG1) rate. A sample size of 86 patients (21-22/arm) was calculated under the hypothesis that the addition of capecitabine or VPA to SCRT can improve the TRG1 rate from 5% to 20%, with one-sided alpha = 0.10 and 80% power.Several biomarkers will be evaluated comparing normal mucosa with tumor (TP, TS, VEGF, RAD51, XRCC1, Histones/proteins acetylation, HDAC isoforms) and on blood samples (polymorphisms of DPD, TS, XRCC1, GSTP1, RAD51 and XRCC3, circulating endothelial and progenitors cells; PBMCs-Histones/proteins acetylation). Tumor metabolism will be measured by 18FDG-PET at baseline and 15 days after the beginning of SCRT., Discussion: This project aims to improve the efficacy of preoperative treatment of LARC and to decrease the inconvenience and the cost of standard long-course RT. Correlative studies could identify both prognostic and predictive biomarkers and could add new insight in the mechanism of interaction between VPA, capecitabine and RT.EudraCT Number: 2012-002831-28., Trial Registration: ClinicalTrials.gov number, NCT01898104.
- Published
- 2014
- Full Text
- View/download PDF
246. Metabolic syndrome-breast cancer link varies by intrinsic molecular subtype.
- Author
-
Capasso I, Esposito E, de Laurentiis M, Maurea N, Cavalcanti E, Botti G, Petrillo A, Montella M, D'Aiuto M, Coppola C, Crispo A, Grimaldi M, Frasci G, Fucito A, Ciliberto G, and D'Aiuto G
- Abstract
Background: Metabolic syndrome (MS) has been shown to increase the risk of breast cancer. Existing data suggest that the strength of metabolic syndrome-breast cancer link varies by intrinsic molecular subtype, but results from worldwide literature are controversial. Primary endpoint of the study was to assess whether MS is a predictor of specific breast cancer (BC) subtype. Secondary endpoint was to determine whether components of MS can individually increase the risk of specific breast cancer subtype., Methods: Anthropometric and metabolic variables were correlated to breast cancer specific subgroups, retrospectively. Statistical significance was considered when p ≤ 0.05 and 95% CI., Results: Data analysis suggests that MS per se represents a modifiable risk factor for BC in postmenopausal [OR 6.28 (95% CI 2.79-14.11) p < 0.00001]. MS per se prevalence is higher among Luminal breast cancers in postmenopausal [OR 1.37 (95% CI 1.07-2.80) p = 0.03]. Body Mass Index (BMI) alone is associated to Luminal A subtype breast cancer risk [OR 1.12 (95% CI 0.96-2.196 p = 0.2]. Waist Circumference > 88 cm has been shown to be specifically and statistically significant associated to HER-2+ breast cancer subtypes in postmenopausal [OR 2.72 (95% CI 1.69- 10.72) p = 0.01], whilst in Luminal B it was only marginally statistical associated [OR 2.21 (95% CI 0.77-2.60) p = 0.1]. Insulin resistance showed statistical significant association to HER-2+ and Luminal B tumors [OR 2.11 (95% CI 1.66-6.69) p = 0.05] and [OR 2.33 (95% CI 1.2-4.2) p = 0.006], respectively. Hence, it has emerged that BMI is weakly associated to Luminal A breast cancers in this case series, whereas visceral obesity and insulin resistance are likely to be linked to more aggressive breast cancer subtypes., Conclusions: New molecular biomarkers unveiling metabolic syndrome related breast carcinogenesis need to be detected to further stratify breast cancer risk by subtypes.
- Published
- 2014
- Full Text
- View/download PDF
247. Ranolazine protects from doxorubicin-induced oxidative stress and cardiac dysfunction.
- Author
-
Tocchetti CG, Carpi A, Coppola C, Quintavalle C, Rea D, Campesan M, Arcari A, Piscopo G, Cipresso C, Monti MG, De Lorenzo C, Arra C, Condorelli G, Di Lisa F, and Maurea N
- Subjects
- Animals, Atrial Natriuretic Factor genetics, Blotting, Western methods, Cardiotoxicity diagnostic imaging, Cardiotoxicity prevention & control, Connective Tissue Growth Factor genetics, Matrix Metalloproteinase 2 genetics, Mice, Mice, Inbred C57BL, Myocytes, Cardiac cytology, Natriuretic Peptide, Brain genetics, Oxidative Stress genetics, Poly(ADP-ribose) Polymerases metabolism, RNA, Messenger genetics, Ranolazine, Reactive Oxygen Species metabolism, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction methods, Sodium blood, Ultrasonography, Ventricular Dysfunction, Left chemically induced, Ventricular Dysfunction, Left metabolism, Acetanilides therapeutic use, Antibiotics, Antineoplastic toxicity, Doxorubicin toxicity, Enzyme Inhibitors therapeutic use, Oxidative Stress drug effects, Piperazines therapeutic use, Ventricular Dysfunction, Left prevention & control
- Abstract
Aims: Doxorubicin is widely used against cancer; however, it can produce heart failure (HF). Among other hallmarks, oxidative stress is a major contributor to HF pathophysiology. The late INa inhibitor ranolazine has proven effective in treating experimental HF. Since elevated [Na+]i is present in failing myocytes, and has been recently linked with reactive oxygen species (ROS) production, our aim was to assess whether ranolazine prevents doxorubicin-induced cardiotoxicity, and whether blunted oxidative stress is a mechanism accounting for such protection., Methods and Result: In C57BL6 mice, doxorubicin treatment for 7 days produced LV dilation and decreased echo-measured fractional shortening (FS). Ranolazine (305 mg/kg/day) prevented LV dilation and dysfunction when co-administered with doxorubicin. Doxorubicin-induced cardiotoxicity was accompanied instead by elevations in atrial natriuretic peptide (ANP), BNP, connective tissue growth factor (CTGF), and matrix metalloproteinase 2 (MMP2) mRNAs, which were not elevated on co-treatment with ranolazine. Alterations in extracellular matrix remodelling were confirmed by an increase in interstitial collagen, which did not rise in ranolazine-co-treated hearts. Levels of poly(ADP-ribose) polymerase (PARP) and pro-caspase-3 measured by western blotting were lowered with doxorubicin, with increased cleavage of caspase-3, indicating activation of the proapoptotic machinery. Again, ranolazine prevented this activation. Furthermore, in HL-1 cardiomyocytes transfected with HyPer to monitor H2O2 emission, besides reducing the extent of cell death, ranolazine prevented the occurrence of oxidative stress caused by doxorubicin. Interestingly, similar protective results were obtained with the Na+/Ca2+ exchanger (NCX) inhibitor KB-R7943., Conclusions: Ranolazine protects against experimental doxorubicin cardiotoxicity. Such protection is accompanied by a reduction in oxidative stress, suggesting that INa modulates cardiac redox balance, resulting in functional and morphological derangements., (© 2014 The Authors. European Journal of Heart Failure © 2013 European Society of Cardiology.)
- Published
- 2014
- Full Text
- View/download PDF
248. Role of hypertension on new onset congestive heart failure in patients receiving trastuzumab therapy for breast cancer.
- Author
-
Russo G, Cioffi G, Gori S, Tuccia F, Boccardi L, Khoury G, Lestuzzi C, Maurea N, Oliva S, Faggiano P, and Tarantini L
- Subjects
- Adult, Aged, Chi-Square Distribution, Female, Heart Failure diagnosis, Heart Failure epidemiology, Heart Failure physiopathology, Humans, Hypertension diagnosis, Hypertension epidemiology, Hypertension physiopathology, Incidence, Italy epidemiology, Logistic Models, Middle Aged, Multivariate Analysis, Odds Ratio, Retrospective Studies, Risk Assessment, Risk Factors, Stroke Volume drug effects, Time Factors, Trastuzumab, Ventricular Function, Left drug effects, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents adverse effects, Breast Neoplasms drug therapy, Heart Failure chemically induced, Hypertension chemically induced
- Abstract
Background: Adjuvant trastuzumab therapy improves survival of Human Epidermal growth factor receptor 2 (HER2)-positive women with early breast cancer (EBC). Trastuzumab-induced cardiotoxicity is not uncommon. In the setting of community patients, the incidence, timing and phenotype of new onset congestive heart failure (CHF) is unknown., Methods: Forty hundred and ninety nine consecutive HER2-positive women (mean age 55 ± 11) with EBC treated with trastuzumab between January 2008 and June 2009 at 10 Italian institutions were followed-up for 1 year. We evaluated incidence, time of occurrence, clinical features associated with CHF. Left ventricular ejection fraction (LVEF) was evaluated by echocardiography at baseline and 3, 6, 9and 12 months during trastuzumab therapy., Results: CHF occurred in 16 patients (3.2%), who were older, more hypertensive and with a higher degree of hypertension in comparison with patients who did not have CHF. All CHF patients had a significant reduction in LVEF with a mean peak of -12 points % detected at 3-month follow-up. CHF occurred in seven patients (44%) within 3-month follow-up, in four patients (25%) between 3-6 months, in three patients (19%) between 6-9 months and in two patients (12%) between 9 and 12 months. Trastuzumab was discontinued in 10 of 16 patients and re-started in five after LVEF recovery and clinical improvement. New onset CHF was predicted by the presence of hypertension [OR 2.9 (CI 1.1-7.9])., Conclusion: New onset CHF occurs seldom in HER2-positive women with EBC, prevalently in the first 6 months of therapy. CHF is associated with a significant reduction in LVEF and is predicted by a history of hypertension.
- Published
- 2014
- Full Text
- View/download PDF
249. Combination of inositol and alpha lipoic acid in metabolic syndrome-affected women: a randomized placebo-controlled trial.
- Author
-
Capasso I, Esposito E, Maurea N, Montella M, Crispo A, De Laurentiis M, D'Aiuto M, Frasci G, Botti G, Grimaldi M, Cavalcanti E, Esposito G, Fucito A, Brillante G, D'Aiuto G, and Ciliberto G
- Subjects
- Aged, Biomarkers blood, Blood Glucose drug effects, Blood Glucose metabolism, Body Mass Index, Breast Neoplasms etiology, Breast Neoplasms prevention & control, Caloric Restriction, Cholesterol, HDL blood, Drug Therapy, Combination, Female, Humans, Insulin blood, Insulin Resistance, Italy, Metabolic Syndrome blood, Metabolic Syndrome diagnosis, Middle Aged, Postmenopause, Prospective Studies, Time Factors, Treatment Outcome, Triglycerides blood, Waist-Hip Ratio, Dietary Supplements, Inositol therapeutic use, Metabolic Syndrome drug therapy, Thioctic Acid therapeutic use
- Abstract
Background: Inositol has been reported to improve insulin sensitivity since it works as a second messenger achieving insulin-like effects on metabolic enzymes. The aim of this study was to evaluate the inositol and alpha lipoic acid combination effectiveness on metabolic syndrome features in postmenopausal women at risk of breast cancer., Methods: A six-month prospective, randomized placebo-controlled trial was carried out on a total of 155 postmenopausal women affected by metabolic syndrome at risk of breast cancer, the INOSIDEX trial. All women were asked to follow a low-calorie diet and were assigned randomly to daily consumption of a combination of inositol and alpha lipoic acid (77 pts) or placebo (78 pts) for six months. Primary outcomes we wanted to achieve were both reduction of more than 20% of the HOMA-IR index and of triglycerides serum levels. Secondary outcomes expected were both the improvement of high-density lipoprotein cholesterol levels and the reduction of anthropometric features such as body mass index and waist-hip ratio., Results: A significant HOMA-IR reduction of more than 20% was evidenced in 66.7% (P <0.0001) of patients, associated with a serum insulin level decrease in 89.3% (P <0.0000). A decrease in triglycerides was evidenced in 43.2% of patients consuming the supplement (P <0.0001). An increase in HDL cholesterol (48.6%) was found in the group consuming inositol with respect to the placebo group. A reduction in waist circumference and waist-hip ratio was found in the treated group with respect to the placebo group., Conclusions: Inositol combined with alpha lipoic acid can be used as a dietary supplement in insulin-resistant patients in order to increase their insulin sensitiveness. Daily consumption of inositol combined with alpha lipoic acid has a significant bearing on metabolic syndrome. As metabolic syndrome is considered a modifiable risk factor of breast tumorigenesis, further studies are required to assess whether inositol combined with alpha lipoic acid can be administered as a dietary supplement in breast cancer primary prevention., Trial Registration: Current Controlled Trial ISRCTN74096908.
- Published
- 2013
- Full Text
- View/download PDF
250. The emerging issue of cardiac dysfunction induced by antineoplastic angiogenesis inhibitors.
- Author
-
Tocchetti CG, Gallucci G, Coppola C, Piscopo G, Cipresso C, Maurea C, Giudice A, Iaffaioli RV, Arra C, and Maurea N
- Subjects
- Cardiovascular System drug effects, Humans, Risk Factors, Vascular Endothelial Growth Factor A antagonists & inhibitors, Ventricular Dysfunction, Left physiopathology, Angiogenesis Inhibitors adverse effects, Antineoplastic Agents adverse effects, Cardiovascular System physiopathology, Ventricular Dysfunction, Left chemically induced
- Abstract
Left ventricular dysfunction from anticancer drugs has emerged as a relevant problem in the clinical and scientific communities. Anthracycline toxicity has always been the most relevant, but with the increasing use of biological targeted therapies in treatment protocols, with an increasing number of cancer survivors, new toxicities have been increasing in more recent years. Cardiomyopathy after ErbB2 inhibitors has been intensively studied. Another important class of biological anticancer drugs are vascular endothelial growth factor (VEGF) inhibitors. VEGF signalling is crucial for vascular growth, but it also has a major impact on myocardial function. Also, it is important to note that such angiogenesis inhibitors are multitargeted in most cases, and can produce a broad spectrum of cardiovascular side effects. Here we review the mechanisms and pathophysiology of the most significant cardiotoxic effects of antiangiogenic drugs, and particular attention is drawn to LV dysfunction, discussing the assessment and management on the basis of the most recent cardio-oncological findings and heart failure guidelines.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.