Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Zhongguo Fei Ai Za Zhi ; 24(9): 668-672, 2021 Sep 20.
Artículo en Chino | MEDLINE | ID: mdl-34521189

RESUMEN

Immune checkpoint inhibitors (ICIs) is a negative regulatory factor antibody, which activates T cells to play an anti-tumor effect in immunotherapy, and can also cause immune-related adverse responses, thereby inducing a series of immune related adverse events (irAEs). Among these irAEs, although the incidence of ICIs-related myocarditis is very low, the fatality rate is significantly higher than other adverse reactions, close to 50%. Clinicians should be vigilant when applying ICIs, but the pathogenesis of ICIs-related myocarditis is still unclear. This article combines the recent research results of ICIs to summarize the mechanism and clinical manifestations of ICIs-related myocarditis, so as to improve clinicians' understanding of the adverse reactions.
.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Miocarditis , Neoplasias , Investigación Biomédica/tendencias , Cardiotoxicidad/diagnóstico , Cardiotoxicidad/etiología , Cardiotoxicidad/inmunología , Cardiotoxicidad/fisiopatología , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/efectos adversos , Miocarditis/inducido químicamente , Miocarditis/diagnóstico , Miocarditis/inmunología , Miocarditis/fisiopatología , Neoplasias/tratamiento farmacológico
2.
Eur J Cancer ; 157: 383-390, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34571335

RESUMEN

AIM: Despite unprecedented results of anti-programmed death protein (ligand) 1 (PD-(L)1) immune checkpoint inhibitor in the oncology's armamentarium, immune-related adverse events (irAEs) represent a therapeutic hurdle. Currently, there is no consensual recommendation on a routinely monitored biomarker to early detect irAE. Biological markers such as serum creatine phosphokinase (CPK) are commonly used to measure muscular tissue injury. The potential of routine serum CPK monitoring to predict cardiac or neuromuscular irAE in patients treated with immunotherapy remains unknown. METHODS: In this retrospective study between January 2016 and December 2018 at Gustave Roussy Cancer Campus, 1151 cancer patients treated with anti-PD-(L)1 immunotherapy were systematically monitored with serum CPK measurements before each immunotherapy cycle. We considered significant CPK increases according to Common Terminology Criteria for Adverse Events v5.0 (CTCAEV5) of grade ≥2 severity. Comparisons were performed in patients with immune-related CPK (ir-CPK) elevations symptomatic versus asymptomatic. RESULTS: Overall, 53 of 1151 (4.6%) patients showed a CPK increase. Elevations of CPK were deemed to be immunotherapy-related in 31 of 1151 (2.7%) patients. Among them, 12 of 31 (38.7%) patients experienced symptomatic cardiac or neuromuscular irAE, whereas the other 19 of 31 (61.3%) patients remained asymptomatic. In patients with symptomatic irAE, the mean ir-CPK level was higher compared with asymptomatic patients (1271 versus 771 UI/L, P value = 0.02). In the asymptomatic group, all patients experienced a spontaneous resolution of the ir-CPK increase, and none required medical intervention. CONCLUSION: Most patients with immune-related CPK increase remained asymptomatic. The CPK serum increase did not alter the clinical management of asymptomatic patients. The results of this study did not support a significant clinical interest for a systematic routine CPK monitoring in patients amenable to anti-PD-(L)1 immunotherapy.


Asunto(s)
Cardiotoxicidad/diagnóstico , Creatina Quinasa/sangre , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Neoplasias/tratamiento farmacológico , Enfermedades Neuromusculares/diagnóstico , Antígeno B7-H1/antagonistas & inhibidores , Biomarcadores/sangre , Cardiotoxicidad/sangre , Cardiotoxicidad/inmunología , Estudios de Factibilidad , Femenino , Humanos , Masculino , Memoria Episódica , Persona de Mediana Edad , Neoplasias/sangre , Enfermedades Neuromusculares/sangre , Enfermedades Neuromusculares/inducido químicamente , Enfermedades Neuromusculares/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Estudios Retrospectivos
3.
Int Immunopharmacol ; 99: 108024, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34333357

RESUMEN

Baicalin (BA), a kind of flavonoids compound, comes from Scutellaria baicalensis Georgi (a kind of perennial herb) and has beneficial effects on the cardiovascular system through anti-oxidant, anti-inflammation, and anti-apoptosis actions. However, the therapeutic effects and latent mechanisms of BA on arsenic trioxide (ATO)-induced cardiac toxicity has not been reported. The present research was performed to explore the effects and mechanisms of BA on ATO-induced heart toxicity. Male Kunming mice were treated with ATO (7.5 mg/kg) to induce cardiac toxicity. After the mice received ATO, BA (50 and 100 mg/kg) was administered for estimating its cardioprotective effects. Statistical data demonstrated that BA treatment alleviated electrocardiogram abnormalities and pathological injury caused by ATO. BA could also lead to recovery of CK and LDH activities to normal range and cause a decrease in MDA levels and ROS generation, augmentation of SOD, CAT, and GSH activities. We also found that BA caused a reduction in the expression of proinflammatory cytokines, such as TNF-α and IL-6. Moreover, BA attenuated ATO-induced apoptosis by promoting the expression of Bcl-2 and suppressing the expression of Bax and caspase-3. TUNEL test result demonstrated BA caused impediment of ATO-induced apoptosis. Furthermore, BA treatment suppressed the high expression of TLR4, NF-κB and P-NF-κB caused by ATO. In conclusion, these results indicate that BA may alleviate ATO-induced cardiac toxicity by restraining oxidative stress, apoptosis, and inflammation, and its mechanism would be associated with the inhibition of the TLR4/NF-κB signaling pathway.


Asunto(s)
Antineoplásicos/toxicidad , Trióxido de Arsénico/toxicidad , Cardiotoxicidad/tratamiento farmacológico , Flavonoides/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Cardiotoxicidad/inmunología , Modelos Animales de Enfermedad , Flavonoides/uso terapéutico , Humanos , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Masculino , Ratones , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/inmunología , Scutellaria/química , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Receptor Toll-Like 4/metabolismo
4.
J Mol Cell Cardiol ; 160: 121-127, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34303670

RESUMEN

Immune checkpoint inhibitors (ICI) have changed the landscape of cancer therapy, but their use carries a high risk of cardiac immune related adverse events (iRAEs). With the expanding utilization of ICI therapy, there is a growing need to understand the underlying mechanisms behind their anti-tumor activity as well as their immune-mediated toxicities. In this review, we will focus on clinical characteristics and immune pathways of ICI cardiotoxicity, with an emphasis on single-cell technologies used to gain insights in this field. We will focus on three key areas of ICI-mediated immune pathways, including the anti-tumor immune response, the augmentation of the immune response by ICIs, and the pathologic "autoimmune" response in some individuals leading to immune-mediated toxicity, as well as local factors in the myocardial immune environment predisposing to autoimmunity. Discerning the underlying mechanisms of these immune pathways is necessary to inform the development of targeted therapies for ICI cardiotoxicities and reduce treatment related morbidity and mortality.


Asunto(s)
Antineoplásicos/efectos adversos , Arritmias Cardíacas/inducido químicamente , Aterosclerosis/inducido químicamente , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunoterapia/métodos , Miocarditis/inducido químicamente , Pericarditis/inducido químicamente , Vasculitis/inducido químicamente , Animales , Arritmias Cardíacas/inmunología , Aterosclerosis/inmunología , Autoinmunidad/efectos de los fármacos , Cardiotoxicidad/inmunología , Humanos , Ratones , Miocarditis/inmunología , Pericarditis/inmunología , Placa Aterosclerótica/inducido químicamente , Placa Aterosclerótica/inmunología , Factores de Riesgo , Resultado del Tratamiento , Vasculitis/inmunología
5.
Curr Oncol Rep ; 23(7): 79, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33937956

RESUMEN

PURPOSE OF REVIEW: Immune checkpoint inhibitors (ICIs) have improved the survival of several cancers. However, they may cause a wide range of immune-related adverse events (irAEs). While most irAEs are manageable with temporary cessation of ICI and immunosuppression, cardiovascular toxicity can be associated with high rates of morbidity and mortality. As ICIs evolve to include high-risk patients with preexisting cardiovascular risk factors and disease, the risk and relevance of ICI-associated cardiotoxicity may be even higher. RECENT FINDINGS: Several cardiovascular toxicities such as myocarditis, stress cardiomyopathy, and pericardial disease have been reported in association with ICIs. Recent findings also suggest an increased risk of atherosclerosis with ICI use. ICI-associated myocarditis usually occurs early after initiation and can be fulminant. A high index of suspicion is required for timely diagnosis. Prompt treatment with high-dose corticosteroids is shown to improve outcomes. Although the overall incidence is rare, ICI cardiotoxicity, particularly myocarditis, is associated with significant morbidity and mortality, making it a major therapy-limiting adverse event. Early recognition and prompt treatment with the cessation of ICI therapy and initiation of high-dose corticosteroids are crucial to improve outcomes. Cardio-oncologists will need to play an important role not just in the management of acute cardiotoxicity but also to reduce the risk of long-term sequelae.


Asunto(s)
Aterosclerosis/diagnóstico , Cardiotoxicidad/diagnóstico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Miocarditis/diagnóstico , Neoplasias/tratamiento farmacológico , Aterosclerosis/inducido químicamente , Aterosclerosis/inmunología , COVID-19/epidemiología , COVID-19/prevención & control , COVID-19/virología , Cardiotoxicidad/etiología , Cardiotoxicidad/inmunología , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/inmunología , Miocarditis/inducido químicamente , Miocarditis/inmunología , Neoplasias/inmunología , Pandemias , Factores de Riesgo , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/fisiología
6.
Circ Res ; 128(11): 1780-1801, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33934609

RESUMEN

Immune-based therapies have revolutionized cancer treatments. Cardiovascular sequelae from these treatments, however, have emerged as critical complications, representing new challenges in cardio-oncology. Immune therapies include a broad range of novel drugs, from antibodies and other biologics, including immune checkpoint inhibitors and bispecific T-cell engagers, to cell-based therapies, such as chimeric-antigen receptor T-cell therapies. The recognition of immunotherapy-associated cardiovascular side effects has also catapulted new research questions revolving around the interactions between the immune and cardiovascular systems, and the signaling cascades affected by T cell activation, cytokine release, and immune system dysregulation. Here, we review the specific mechanisms of immune activation from immunotherapies and the resulting cardiovascular toxicities associated with immune activation and excess cytokine production.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Inmunoterapia/efectos adversos , Neoplasias/terapia , Animales , Anticuerpos Biespecíficos/efectos adversos , Anticuerpos Biespecíficos/uso terapéutico , Cardiotoxicidad/inmunología , Enfermedades Cardiovasculares/inmunología , Síndrome de Liberación de Citoquinas/complicaciones , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/terapia , Modelos Animales de Enfermedad , Perros , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia/métodos , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Activación de Linfocitos , Ratones , Ratas , Receptores Quiméricos de Antígenos , Linfocitos T/trasplante
8.
J Immunother Cancer ; 7(1): 266, 2019 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-31627742

RESUMEN

BACKGROUND: Nivolumab, a programmed death-1 (PD-1) inhibitor, is an immune checkpoint inhibitor particularly used in the treatment of malignant melanoma, non-small cell lung cancer and renal cell carcinoma. Immune-related adverse events are frequent under immunotherapies. Cardiotoxic side effects, initially thought to be rare, are more often encountered paralleling the expanding use of immune checkpoint blockade. Among them, pericardial effusion and tamponade deserve attention as they may present with unusual symptomatology. CASE PRESENTATION: We report three cases of pericardial effusion under nivolumab for lung adenocarcinoma. Two cases of early and late-onset pericardial effusion were symptomatic with tamponade and one case occurred without any symptoms. Pericardiocentesis with pericardial biopsy was performed in symptomatic pericardial effusion followed by the administration of a corticotherapy. Pericardial biopsies showed infiltration of T-lymphocytes, mostly CD4+. Nivolumab was stopped in two cases and resumed for one patient. Pericardial effusion evolved positively in all cases with or without treatment. CONCLUSIONS: We review the literature on pericardial effusion under nivolumab to further discuss the hallmarks of pericardial effusion under nivolumab and the management of nivolumab therapy in this situation. In conclusion, pericardial effusion as an immune-related adverse event under nivolumab appears less rare than initially thought and may require particular attention.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Antineoplásicos Inmunológicos/efectos adversos , Neoplasias Pulmonares/tratamiento farmacológico , Nivolumab/efectos adversos , Derrame Pericárdico/inducido químicamente , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/patología , Anciano , Cardiotoxicidad/diagnóstico , Cardiotoxicidad/inmunología , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Derrame Pericárdico/complicaciones , Derrame Pericárdico/diagnóstico , Derrame Pericárdico/inmunología , Factores de Transcripción/metabolismo
9.
Cells ; 8(10)2019 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-31600901

RESUMEN

Doxorubicin (Dox) is an effective antineoplastic agent used to treat cancers, but its use is limited as Dox induces adverse cardiotoxic effects. Dox-induced cardiotoxicity (DIC) can lead to heart failure and death. There is no study that investigates whether embryonic stem cell-derived exosomes (ES-Exos) in DIC can attenuate inflammation-induced pyroptosis, pro-inflammatory M1 macrophages, inflammatory cell signaling, and adverse cardiac remodeling. For this purpose, we transplanted ES-Exos and compared with ES-cells (ESCs) to examine pyroptosis, inflammation, cell signaling, adverse cardiac remodeling, and their influence on DIC induced cardiac dysfunction. Therefore, we used C57BL/6J mice ages 10 ± 2 weeks and divided them into four groups (n = 6-8/group): Control, Dox, Dox + ESCs, and Dox + ES-Exos. Our data shows that the Dox treatment significantly increased expression of inflammasome markers (TLR4 and NLRP3), pyroptotic markers (caspase-1, IL1-ß, and IL-18), cell signaling proteins (MyD88, p-P38, and p-JNK), pro-inflammatory M1 macrophages, and TNF-α cytokine. This increased pyroptosis, inflammation, and cell signaling proteins were inhibited with ES-Exos or ESCs. Moreover, ES-Exos or ESCs increased M2 macrophages and anti-inflammatory cytokine, IL-10. Additionally, ES-Exos or ESCs treatment inhibited significantly cytoplasmic vacuolization, myofibril loss, hypertrophy, and improved heart function. In conclusion, for the first time we demonstrated that Dox-induced pyroptosis and cardiac remodeling are ameliorated by ES-Exos or ESCs.


Asunto(s)
Cardiotoxicidad/terapia , Doxorrubicina/efectos adversos , Exosomas/trasplante , Macrófagos/inmunología , Células Madre Embrionarias de Ratones/citología , Animales , Cardiotoxicidad/etiología , Cardiotoxicidad/inmunología , Cardiotoxicidad/fisiopatología , Línea Celular , Femenino , Regulación de la Expresión Génica , Pruebas de Función Cardíaca , Masculino , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Piroptosis , Transducción de Señal , Receptor Toll-Like 4/metabolismo
10.
Clin Exp Allergy ; 49(12): 1615-1623, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31519068

RESUMEN

The symptoms of chronic urticaria, be it chronic spontaneous urticaria (CSU) or chronic inducible urticaria (CindU), are mediated primarily by the actions of histamine on H1 receptors located on endothelial cells (the weal) and on sensory nerves (neurogenic flare and pruritus). Thus, second-generation H1 antihistamines (sgAHs) are the primary treatment of these conditions. However, many patients are poorly responsive to licensed doses of antihistamines. In these patients, the current EAACI/GA2 LEN/EDF/WAO guideline for urticaria suggests updosing of sgAHs up to fourfold. However, such updosing is off-label and the responsibility resides with the prescribing physician. Therefore, the safety of the drug when used above its licensed dose is of paramount importance. An important aspect of safety is potential cardiotoxicity. This problem was initially identified some 20 years ago with cardiotoxic deaths occurring with astemizole and terfenadine, two early sgAHs. In this review, we discuss the mechanisms and assessments of potential cardiotoxicity of H1 antihistamines when updosed to four times their licensed dose. In particular, we have focused on the potential of H1 antihistamines to block hERG (human Ether-a-go-go-Related Gene) voltage-gated K+ channels, also known as Kv11.1 channels according to the IUPHAR classification. Blockade of these channels causes QT prolongation leading to torsade de pointes that may possibly degenerate into ventricular fibrillation and sudden death. We considered in detail bilastine, cetirizine, levocetirizine, ebastine, fexofenadine, loratadine, desloratadine, mizolastine and rupatadine and concluded that all these drugs have an excellent safety profile with no evidence of cardiotoxicity even when updosed up to four times their standard licensed dose, provided that the prescribers carefully consider and rule out potential risk factors for cardiotoxicity, such as the presence of inherited long QT syndrome, older age, cardiovascular disorders, hypokalemia and hypomagnesemia, or the use of drugs that either have direct QT prolonging effects or inhibit sgAH metabolism.


Asunto(s)
Cardiotoxicidad , Urticaria Crónica , Canal de Potasio ERG1 , Antagonistas de los Receptores Histamínicos H1 , Torsades de Pointes , Factores de Edad , Cardiotoxicidad/inmunología , Cardiotoxicidad/patología , Cardiotoxicidad/prevención & control , Urticaria Crónica/tratamiento farmacológico , Urticaria Crónica/inmunología , Urticaria Crónica/patología , Canal de Potasio ERG1/inmunología , Femenino , Antagonistas de los Receptores Histamínicos H1/efectos adversos , Antagonistas de los Receptores Histamínicos H1/uso terapéutico , Humanos , Síndrome de QT Prolongado/inmunología , Síndrome de QT Prolongado/patología , Masculino , Factores de Riesgo , Torsades de Pointes/inducido químicamente , Torsades de Pointes/inmunología , Torsades de Pointes/patología
12.
Immunotherapy ; 11(8): 725-735, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31088241

RESUMEN

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


Asunto(s)
Antígeno B7-H1 , Carcinoma de Pulmón de Células no Pequeñas , Inmunoterapia , Neoplasias Pulmonares , Melanoma , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/terapia , Cardiotoxicidad/inmunología , Cardiotoxicidad/mortalidad , Cardiotoxicidad/patología , Cardiotoxicidad/prevención & control , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Masculino , Melanoma/inmunología , Melanoma/mortalidad , Melanoma/patología , Melanoma/terapia , Ensayos Clínicos Controlados Aleatorios como Asunto
13.
J Pathol ; 247(3): 320-332, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30426505

RESUMEN

Immune responses contribute to a large extent to heart diseases. However, it is still not clear how the key inflammatory mediator interferon-γ (IFNγ) plays a role in doxorubicin (DOX)-induced cardiomyopathy. We report here that DOX-induced heart dysfunction involves IFNγ signaling in mice. The IFNγ receptor was found to be highly expressed on cardiomyocytes, and its downstream signaling was activated in heart tissues upon DOX treatment. In vitro, IFNγ strongly aggravated the injury of cardiomyocytes exposed to DOX. Although not affecting DOX-induced cell death, IFNγ disrupted mitochondrial respiration and fatty acid oxidation in DOX-exposed cardiomyocytes. IFNγ extended the suppression of the AMP-activated protein kinase (AMPK)/acetyl-CoA carboxylase (ACC) axis by DOX to a p38-dependent branch. Activation of AMPK or inhibition of p38 inhibited the enhancing effect of IFNγ on the DOX-induced cardiotoxicity and prolonged the survival time in DOX-treated mice. Taken together, our results indicate that reprogramming of cardiac metabolism by IFNγ represents a previously unidentified key step for DOX-induced cardiomyopathy. This unavoidable impact of IFNγ on cardiomyocyte metabolism during chemotherapy redirects our attention to the balance between beneficial immunosurveillance of cancer cells and unwanted toxic side-effects. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Antibióticos Antineoplásicos/toxicidad , Cardiotoxicidad/inmunología , Doxorrubicina/toxicidad , Interferón gamma/inmunología , Miocitos Cardíacos/efectos de los fármacos , Animales , Antibióticos Antineoplásicos/farmacología , Cardiotoxicidad/etiología , Cardiotoxicidad/patología , Respiración de la Célula/fisiología , Células Cultivadas , Reprogramación Celular/efectos de los fármacos , Reprogramación Celular/inmunología , Doxorrubicina/farmacología , Ácidos Grasos/metabolismo , Femenino , Ratones Endogámicos C57BL , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/inmunología , Oxidación-Reducción , Receptor de Interferón alfa y beta/metabolismo , Transducción de Señal/inmunología
14.
Acta Pharmacol Sin ; 39(11): 1693-1698, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29991709

RESUMEN

The immune checkpoint molecules are emerged in the evolution to protect the host from self-attacks by activated T cells. However, cancer cells, as a strategy to survive and expand, can hijack these molecules and mechanisms to suppress T cell-mediated immune responses. Therefore, an idea of blocking the checkpoint molecules to enhance the anti-tumor activities of the host immune system has been developed and applied to the cancer therapy after discovery of the inhibitory T cell co-receptor, cytotoxic T-lymphocyte associated protein 4 (CTLA-4), and further enhanced on the identification of PD-1 and its ligands. Since 2010, several checkpoint inhibitors have been approved by FDA and many more are in clinical trials. In the treatment of advanced cancers, these inhibitors significantly increased response rates and survival benefits. However, accompanied with the striking results, immune-related adverse events (irAEs) that broadly occurred in many organs were observed and reported, some of which were fatal. Herein, we first review the recent progressions in the research of the immune checkpoint molecules and the application of their blocking antibodies in cancer treatment, and then discuss the cardiac toxicity induced by the therapy and the strategy to monitor, manage this adverse event when it occurs.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Enfermedades Autoinmunes/etiología , Cardiotoxicidad/etiología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Receptores Coestimuladores e Inhibidores de Linfocitos T/antagonistas & inhibidores , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/inmunología , Antineoplásicos/efectos adversos , Antineoplásicos/inmunología , Enfermedades Autoinmunes/inmunología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Antígeno CTLA-4/antagonistas & inhibidores , Antígeno CTLA-4/inmunología , Cardiotoxicidad/inmunología , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Humanos , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología
15.
Curr Oncol Rep ; 20(6): 44, 2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29644505

RESUMEN

PURPOSE OF REVIEW: This review describes cardiotoxicity associated with adoptive T cell therapy and immune checkpoint blockade. RECENT FINDINGS: Cardiotoxicity is a rare but potentially fatal complication associated with novel immunotherapies. Both affinity-enhanced and chimeric antigen receptor T cells have been reported to cause hypotension, arrhythmia, and left ventricular dysfunction, typically in the setting of cytokine release syndrome. Immune checkpoint inhibitors are generally well-tolerated but have the potential to cause myocarditis, with clinical presentations ranging from asymptomatic cardiac biomarker elevation to heart failure, arrhythmia, and cardiogenic shock. Electrocardiography, cardiac biomarker measurement, and cardiac imaging are key components of the diagnostic evaluation. For suspected myocarditis, endomyocardial biopsy is recommended if the diagnosis remains unclear after initial testing. The incidence of immunotherapy-associated cardiotoxicity is likely underestimated and may increase as adoptive T cell therapy and immune checkpoint inhibitors are used in larger populations and for longer durations of therapy. Baseline and serial cardiac evaluation is recommended to facilitate early identification and treatment of cardiotoxicity.


Asunto(s)
Cardiotoxicidad/inmunología , Inmunoterapia/efectos adversos , Neoplasias/terapia , Arritmias Cardíacas/epidemiología , Arritmias Cardíacas/etiología , Arritmias Cardíacas/inmunología , Cardiotoxicidad/etiología , Cardiotoxicidad/patología , Insuficiencia Cardíaca/epidemiología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/inmunología , Humanos , Hipotensión/epidemiología , Hipotensión/etiología , Hipotensión/inmunología , Neoplasias/complicaciones , Neoplasias/inmunología , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/inmunología , Disfunción Ventricular Izquierda/epidemiología , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/inmunología
16.
J Biochem Mol Toxicol ; 32(2)2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29315967

RESUMEN

Doxorubicin (DXR) is a highly effective drug for chemotherapy. However, cardiotoxicity reduces its clinical utility in humans. The present study aimed to assess the ameliorative effect of curcumin against DXR-induced cardiotoxicity in rats. Rats were subjected to oral treatment of curcumin (100 and 200 mg/kg body weight) for 7 days. Cardiotoxicity was induced by single intraperitoneal injection of DXR (40 mg/kg body weight) on the 5th day and the rats sacrificed on 8th day. Curcumin ameliorated DXR-induced lipid peroxidation, glutathione depletion, decrease in antioxidant (superoxide dismutase, catalase, and glutathione peroxidase) enzyme activities, and cardiac toxicity markers (CK-MB, LDH, and cTn-I). Curcumin also attenuated activities of Caspase-3, cyclooxygenase-2, inducible nitric oxide synthase, and levels of nuclear factor kappa-B, tumor necrosis factor-α, and interleukin-1ß, and cardiac tissue damages that were induced by DXR. Moreover, curcumin decreased the expression of 8-OHdG and 3,3'-dityrosine. This study demonstrated that curcumin has a multi-cardioprotective effect due to its antioxidant, anti-inflammatory, and antiapoptotic properties.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Apoptosis/efectos de los fármacos , Cardiotónicos/uso terapéutico , Cardiotoxicidad/prevención & control , Curcumina/uso terapéutico , Ventrículos Cardíacos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/efectos adversos , Antibióticos Antineoplásicos/efectos adversos , Antioxidantes/administración & dosificación , Antioxidantes/efectos adversos , Antioxidantes/uso terapéutico , Biomarcadores/metabolismo , Cardiotónicos/administración & dosificación , Cardiotónicos/efectos adversos , Cardiotoxicidad/inmunología , Cardiotoxicidad/metabolismo , Cardiotoxicidad/patología , Curcumina/administración & dosificación , Curcumina/efectos adversos , Daño del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Doxorrubicina/efectos adversos , Glutatión/metabolismo , Ventrículos Cardíacos/inmunología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Oxidación-Reducción , Distribución Aleatoria , Ratas Wistar
18.
Toxins (Basel) ; 7(7): 2371-84, 2015 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-26120785

RESUMEN

Protective immunity against anthrax is inferred from measurement of vaccine antigen-specific neutralizing antibody titers in serum samples. In animal models, in vivo challenges with toxin and/or spores can also be performed. However, neither of these approaches considers toxin-induced damage to specific organ systems. It is therefore important to determine to what extent anthrax vaccines and existing or candidate adjuvants can provide organ-specific protection against intoxication. We therefore compared the ability of Alum, CpG DNA and the CD1d ligand α-galactosylceramide (αGC) to enhance protective antigen-specific antibody titers, to protect mice against challenge with lethal toxin, and to block cardiotoxicity and hepatotoxicity. By measurement of serum cardiac Troponin I (cTnI), and hepatic alanine aminotransferase (ALT), and aspartate aminotransferase (AST), it was apparent that neither vaccine modality prevented hepatic intoxication, despite high Ab titers and ultimate survival of the subject. In contrast, cardiotoxicity was greatly diminished by prior immunization. This shows that a vaccine that confers survival following toxin exposure may still have an associated morbidity. We propose that organ-specific intoxication should be monitored routinely during research into new vaccine modalities.


Asunto(s)
Vacunas contra el Carbunco/farmacología , Carbunco/complicaciones , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , Cardiotoxicidad/prevención & control , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Alanina Transaminasa/sangre , Animales , Carbunco/inmunología , Vacunas contra el Carbunco/inmunología , Aspartato Aminotransferasas/sangre , Cardiotoxicidad/sangre , Cardiotoxicidad/inmunología , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/inmunología , Ratones Endogámicos C57BL , Análisis de Supervivencia , Troponina I/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA