Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.743
Filter
Add more filters

Publication year range
1.
Annu Rev Pharmacol Toxicol ; 61: 113-134, 2021 01 06.
Article in English | MEDLINE | ID: mdl-32776859

ABSTRACT

Immune checkpoint inhibitors (ICIs) attenuate mechanisms of self-tolerance in the immune system, enabling T cell responses to cancerous tissues and revolutionizing care for cancer patients. However, by loweringbarriers against self-reactivity, ICIs often result in varying degrees of autoimmunity. Cardiovascular complications, particularly myocarditis but also arrhythmias, pericarditis, and vasculitis, have emerged as significant complications associated with ICIs. In this review, we examine the clinical aspects and basic science principles that underlie ICI-associated myocarditis and other cardiovascular toxicities. In addition, we discuss current therapeutic approaches. We believe a better mechanistic understanding of ICI-associated toxicities can lead to improved patient outcomes by reducing treatment-related morbidity.


Subject(s)
Cardiovascular System , Myocarditis , Neoplasms , Cardiotoxicity , Humans , Immune Checkpoint Inhibitors , Myocarditis/drug therapy , Neoplasms/drug therapy
2.
Curr Opin Rheumatol ; 36(1): 76-93, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37962165

ABSTRACT

PURPOSE OF REVIEW: In systemic sclerosis (SSc) primary heart involvement (pHI) is frequent, even though often unrecognized due to its occult nature and to the lack of a specific diagnostic algorithm. The purpose of this review is to report the state of the art of the evidence in the current literature, as well as the overall diagnostic modalities and therapeutic strategies for primary heart involvement in SSc. RECENT FINDINGS: SSc-pHI is defined by the presence of cardiac abnormalities that are predominantly attributable to SSc rather than other causes and/or complications; it may be sub-clinical and must be confirmed through diagnostic investigations. Novel electrocardiographic analysis and cardiac magnetic resonance (CMR) with mapping techniques have been recently proposed, showing a great utility in the early identification of SSc-pHI and in the noninvasive characterization of myocardial tissue. Immunosuppressive therapy emerged as fundamental to curb myocardial inflammation, and recent preclinical and clinical data support the role of antifibrotic drugs to treat SSc-pHI. SUMMARY: our review will help clinicians to properly integrate the available diagnostic modalities for the assessment of SSc-pHI. The ultimate goal is to propose a feasible diagnostic algorithm for the early identification of patients with SSc-pHI, and a schematic therapeutic approach to manage SSc-pHI.


Subject(s)
Myocarditis , Scleroderma, Systemic , Humans , Scleroderma, Systemic/complications , Scleroderma, Systemic/diagnosis , Scleroderma, Systemic/therapy , Heart , Myocarditis/drug therapy , Myocarditis/etiology , Myocarditis/pathology , Magnetic Resonance Imaging , Risk Assessment
3.
J Clin Immunol ; 44(3): 81, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38485795

ABSTRACT

Myocarditis can be caused by viral infection, drug reaction or general inflammatory condition. To provide understanding on inflammatory myocarditis, we describe clinical, genetic, and immunological properties of a young male patient who suffered from recurrent myocarditis episodes since the age of four years. Electrocardiography, troponin I/T, echocardiography, myocardial magnetic resonance imaging and histological findings were consistent with recurrent myocarditis episodes. Homozygous c.245 A > G p.Tyr82Cys pathogenic variant in Hepatitis A Virus Cellular Receptor 2 (HAVCR2) gene encoding T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) receptor was found. Peripheral blood mononuclear cells were collected when the patient was asymptomatic; CD4+ and CD8+ T lymphoblasts, CD56+ natural killer cells and CD14+ monocytes were negative for surface TIM-3 expression. In vitro, TLR4 mediated interleukin-1ß (IL-1ß) response was high after LPS/ATP stimulation. Clinical symptoms responded to IL-1 receptor antagonist anakinra. TIM-3 p.Tyr82Cys CD4+ and CD8+ T cell proliferation in vitro was unrestrained. Findings on IL-2, interferon gamma, regulatory T cells, signal transducer and activator of transcription (STAT) 1, 3 and 4 phosphorylation, and PD-1 and LAG-3 checkpoint inhibitor receptor analyses were comparable to controls. We conclude that TIM-3 deficiency due to homozygous HAVCR2 c.245 A > G p.Tyr82Cys pathogenic variant in the patient described here is associated with autoinflammatory symptoms limited to early onset recurrent febrile myocarditis. Excessive IL-1ß production and defective regulation of T cell proliferation may contribute to this clinical condition responsive to anakinra treatment.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Myocarditis , Humans , Male , Child, Preschool , Hepatitis A Virus Cellular Receptor 2/genetics , Myocarditis/diagnosis , Myocarditis/drug therapy , Myocarditis/etiology , Leukocytes, Mononuclear , Interleukin 1 Receptor Antagonist Protein , Interleukin-1beta , Germ Cells
4.
Biochem Biophys Res Commun ; 733: 150702, 2024 Nov 12.
Article in English | MEDLINE | ID: mdl-39298917

ABSTRACT

Myocarditis is an inflammatory lesion of the myocardium that is caused by a variety of factors. At present, treatment of symptoms remains the main clinical intervention, but it cannot reduce the myocarditis damage caused by inflammation. M1 macrophages are thought to contribute significantly to the occurrence and development of inflammation by secreting a large number of proinflammatory factors. Puerarin is an isoflavone derivative isolated from pueraria that can be used as a dietary supplement and exerts wide range of anti-inflammatory and antioxidant effects. However, the mechanism underlying its anti-inflammatory effects needs to be further studied. The objective of this study was to investigate whether puerarin inhibited M1 polarization by affecting the JAK-STAT signaling pathway in a mouse model of autoimmune myocarditis, thus inhibiting the occurrence of inflammation in experimental autoimmune myocarditis (EAM) model mice. The results showed that EAM model mice treated with puerarin showed milder clinical symptoms and inflammatory infiltration than EAM model mice. Puerarin suppressed the in vivo and in vitro JAK1/2-STAT1 signal transduction in macrophages, thus inhibiting M1 polarization, reducing the secretion of proinflammatory factors, and ultimately decreasing IFN-γ and TNF-α levels in vivo, which led to myocardial apoptosis. Thus, puerarin could alleviate myocardial damage caused by inflammation. The conclusion of this study was that puerarin reduced myocardial damage in EAM model mice by regulating the polarization of macrophages toward M1, and this inhibitory effect may be achieved by inhibiting JAK1/2-STAT1 signaling.


Subject(s)
Disease Models, Animal , Isoflavones , Macrophages , Myocarditis , STAT1 Transcription Factor , Animals , STAT1 Transcription Factor/metabolism , Isoflavones/pharmacology , Isoflavones/therapeutic use , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Myocarditis/drug therapy , Myocarditis/pathology , Myocarditis/metabolism , Myocarditis/prevention & control , Mice , Male , Signal Transduction/drug effects , Myocardium/pathology , Myocardium/metabolism , Mice, Inbred BALB C , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Autoimmune Diseases/metabolism , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , Anti-Inflammatory Agents/pharmacology
5.
Cytokine ; 179: 156620, 2024 07.
Article in English | MEDLINE | ID: mdl-38701735

ABSTRACT

PURPOSE: The emergence of immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment, but these drugs can also cause severe immune-related adverse effects (irAEs), including myocarditis. Researchers have become interested in exploring ways to mitigate this side effect, and one promising avenue is the use of baricitinib, a Janus kinase inhibitor known to have anti-inflammatory properties. This study aimed to examine the potential mechanism by which baricitinib in ICIs-related myocarditis. METHODS: To establish an ICIs-related myocarditis model, BALB/c mice were administered murine cardiac troponin I (cTnI) peptide and anti-mouse programmed death 1 (PD-1) antibodies. Subsequently, baricitinib was administered to the mice via intragastric administration. Echocardiography, HE staining, and Masson staining were performed to evaluate myocardial functions, inflammation, and fibrosis. Immunofluorescence was used to detect macrophages in the cardiac tissue of the mice.In vitro experiments utilized raw264.7 cells to induce macrophage polarization using anti-PD-1 antibodies. Different concentrations of baricitinib were applied to assess cell viability, and the release of pro-inflammatory cytokines was measured. The activation of the JAK1/STAT3 signaling pathway was evaluated through western blot analysis. RESULTS: Baricitinib demonstrated its ability to improve cardiac function and reduce cardiac inflammation, as well as fibrosis induced by ICIs. Mechanistically, baricitinib treatment promoted the polarization of macrophages towards the M2 phenotype. In vitro and in vivo experiments showed that anti-PD-1 promoted the release of inflammatory factors. However, treatment with baricitinib significantly inhibited the phosphorylation of JAK1 and STAT3. Additionally, the use of RO8191 reversed the effects of baricitinib, further confirming our findings. CONCLUSION: Baricitinib demonstrated its potential as a protective agent against ICIs-related myocarditis by modulating macrophage polarization. These findings provide a solid theoretical foundation for the development of future treatments for ICIs-related myocarditis.


Subject(s)
Azetidines , Janus Kinase 1 , Macrophages , Mice, Inbred BALB C , Myocarditis , Purines , Pyrazoles , STAT3 Transcription Factor , Sulfonamides , Animals , Male , Mice , Azetidines/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Janus Kinase 1/metabolism , Macrophage Activation/drug effects , Macrophages/metabolism , Macrophages/drug effects , Myocarditis/chemically induced , Myocarditis/drug therapy , Myocarditis/pathology , Myocarditis/metabolism , Purines/pharmacology , Pyrazoles/pharmacology , RAW 264.7 Cells , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism , Sulfonamides/pharmacology , Troponin I/metabolism
6.
Mol Pharm ; 21(6): 2865-2877, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38666508

ABSTRACT

Imaging strategies for the specific detection and therapeutic monitoring of myocarditis are still lacking. Stimulator of interferon genes (STING) is a signal transduction molecule involved in an innate immune response. Here, we evaluated the feasibility of the recently developed STING-targeted radiotracer [18F]FBTA for positron emission tomography (PET) imaging to detect myocardial inflammation and monitor treatment in myocarditis mice. [18F]FBTA-PET imaging was performed in myocarditis mice and normal mice to verify the specificity of [18F]FBTA for the diagnosis of myocarditis. We also performed PET imaging in mice with myocarditis treated to verify the ability of [18F]FBTA in therapeutic monitoring. The expression of STING and inflammatory cell types was confirmed by flow cytometry and immunohistochemistry. [18F]FDG-PET imaging of myocarditis was used as a contrast. [18F]FBTA-PET imaging showed that the average radioactive uptake was significantly higher in the hearts of the myocarditis group than in the control group. STING was highly overexpressed in cardiac inflammatory cells, including macrophages, dendritic cells (DCs), and T cells. However, there was no significant difference in cardiac radiotracer uptake of [18F]FDG between the myocarditis group and the control group. Moreover, cardiac uptake of [18F]FBTA was significantly reduced in cyclosporin A-treated myocarditis mice and myocardial STING expression was also significantly reduced after the treatment. Overall, we showed that a STING-targeted PET tracer [18F]FBTA can be used to monitor changes in the inflammatory microenvironment in myocarditis. Besides, [18F]FBTA-PET is also suitable for real-time monitoring of myocarditis treatment, representing a promising diagnostic and therapeutic monitoring approach for myocarditis.


Subject(s)
Membrane Proteins , Myocarditis , Positron-Emission Tomography , Animals , Male , Mice , Cyclosporine , Dendritic Cells/metabolism , Disease Models, Animal , Fluorodeoxyglucose F18 , Membrane Proteins/metabolism , Mice, Inbred BALB C , Myocarditis/diagnostic imaging , Myocarditis/drug therapy , Myocardium/metabolism , Myocardium/pathology , Radiopharmaceuticals
7.
J Cardiovasc Pharmacol ; 83(5): 364-376, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38335530

ABSTRACT

ABSTRACT: Myocarditis is an inflammatory disease of the myocardium characterized by a great heterogeneity of presentation and evolution. Treatment of myocarditis is often supportive, and the evidence for immunosuppression is scarce and debated. Conventional treatment is based on clinical presentation, ranging from conservative to advanced mechanical assist devices. In this setting, immunosuppression and immunomodulation therapies are mostly reserved for patients presenting with major clinical syndromes. In this review, we will summarize the current evidence and strategies for conventional and immunosuppressive treatments for patients presenting with acute myocarditis.


Subject(s)
Immunosuppressive Agents , Myocarditis , Myocarditis/immunology , Myocarditis/therapy , Myocarditis/drug therapy , Humans , Acute Disease , Treatment Outcome , Immunosuppressive Agents/therapeutic use , Animals , Immunomodulating Agents/therapeutic use , Immunomodulation
8.
Pediatr Cardiol ; 45(5): 1048-1054, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38509207

ABSTRACT

The purpose of this paper was to unravel the clinical effect analysis of different doses of creatine phosphate sodium (CPS) combined with immunoglobulin in the treatment of pediatric viral myocarditis (VMC). One hundred and twenty children with VMC were recruited and randomized into three groups (40 patients each). Group I received 1.0 g of CPS dissolved in 100 mL of 5% glucose injection intravenously 1 time/day; group II received 1.25 g of CPS dissolved in 125 mL of 5% glucose injection intravenously 1 time/day; group III received 1.5 g of CPS dissolved in 150 mL of 5% glucose injection intravenously 1 time/day; then all three groups were treated with combined use of immunoglobulin (300-400 mg/day) intravenously once a day; and all three groups were treated for 14 days. The clinical efficacy, cardiac function, serum inflammatory factor levels, immune function, and the occurrence of drug toxicity and adverse effects of the children in the three groups were compared after 14 days of treatment. All three groups achieved better therapeutic effects after treatment, in which the effective rate of the Group II and Group III was notably higher versus the Group I. Lower levels of cTnI, CK-MB, LDH, AST, IL-18, IL-6, IFN-γ, and LVEDD and higher CD3+, CD4+, and CD4+/CD8+, FS, and LVEF values were noted in the Group II and Group III versus the Group I, and the results were more pronounced in the high-dose group. The liver and kidney functions of the children in the three groups before and after treatment did not show any significant changes and the incidence of adverse reactions during the treatment period was low in all three groups. Children with VMC can be treated with high-dose CPS in combination with immunoglobulin, which can improve their cardiac function and immune function and reduce the inflammatory response with good overall therapeutic efficacy and fewer adverse effects.


Subject(s)
Myocarditis , Phosphocreatine , Humans , Myocarditis/drug therapy , Male , Female , Child , Child, Preschool , Treatment Outcome , Drug Therapy, Combination , Dose-Response Relationship, Drug , Virus Diseases/drug therapy , Immunoglobulins/administration & dosage , Immunoglobulins/therapeutic use
9.
Int J Mol Sci ; 25(18)2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39337458

ABSTRACT

Myocarditis is a major cause of heart failure and death, particularly in young individuals. Current treatments are mainly symptomatic, but emerging therapies focus on targeting inflammation and fibrosis pathways. Natural bioactive compounds like flavonoids and phenolic acids show promising anti-inflammatory and antioxidant properties. Corticosteroids are frequently employed in the treatment of autoimmune myocarditis and appear to lower mortality rates compared to conventional therapies for heart failure. This study aims to explore the effects of Mangiferin on pro-inflammatory cytokine levels, nitro-oxidative stress markers, histopathological alterations, and cardiac function in experimental myosin-induced autoimmune myocarditis. The effects were compared to Prednisone, used as a reference anti-inflammatory compound, and Trolox, used as a reference antioxidant. The study involved 30 male Wistar-Bratislava rats, which were randomly divided into five groups: a negative control group (C-), a positive control group with induced myocarditis using a porcine myosin solution (C+), three groups with induced myocarditis receiving Mangiferin (M), Prednisone (P), or Trolox (T) as treatment. Cardiac function was evaluated using echocardiography. Biochemical measurements of nitro-oxidative stress and inflammatory markers were conducted. Finally, histopathological changes were assessed. At echocardiography, the evaluation of the untreated myocarditis group showed a trend toward decreased left ventricular ejection fraction (LVEF) but was not statistically significant, while all treated groups showed some improvement in LVEF and left ventricular fraction shortening (LVFS). Significant changes were seen in the Mangiferin group, with lower end-diastolic left ventricular posterior wall (LVPWd) by day 21 compared to the Trolox group (p < 0.001). In the first week of the experiment, levels of interleukins (IL)-1ß, IL-6, and tumour necrosis factor (TNF)-α were significantly higher in the myosin group compared to the negative control group (p < 0.001, p < 0.001, p < 0.01), indicating the progression of inflammation in this group. Treatment with Mangiferin, Prednisone, and Trolox caused a significant reduction in IL-1ß compared to the positive control group (p < 0.001). Notably, Mangiferin resulted in a superior reduction in IL-1ß compared to Prednisone (p < 0.05) and Trolox (p < 0.05). Furthermore, Mangiferin treatment led to a statistically significant increase in total oxidative capacity (TAC) (p < 0.001) and a significant reduction in nitric oxide (NOx) levels (p < 0.001) compared to the negative control group. Furthermore, when compared to the Prednisone-treated group, Mangiferin significantly reduced NOx levels (p < 0.001) and increased TAC levels (p < 0.001). Mangiferin treatment significantly lowered creatine kinase (CK) and aspartate aminotransferase (AST) levels on day 7 (p < 0.001 and p < 0.01, respectively) and reduced CK levels on day 21 (p < 0.01) compared to the untreated group. In the nontreated group, the histological findings at the end of the experiment were consistent with myocarditis. In the group treated with Mangiferin, only one case exhibited mild inflammatory infiltrates, represented by mononucleated leukocytes admixed with few neutrophils, with the severity graded as mild. Statistically significant correlations between the grades (0 vs. 1-2) and the study groups have been highlighted (p < 0.005). This study demonstrated Mangiferin's cardioprotective effects in autoimmune myocarditis, showing reduced oxidative stress and inflammation. Mangiferin appears promising as a treatment for acute myocarditis, but further research is needed to compare its efficacy with other treatments like Trolox and Prednisone.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Disease Models, Animal , Myocarditis , Oxidative Stress , Rats, Wistar , Xanthones , Animals , Myocarditis/drug therapy , Myocarditis/metabolism , Myocarditis/pathology , Antioxidants/pharmacology , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Male , Xanthones/pharmacology , Xanthones/therapeutic use , Rats , Oxidative Stress/drug effects , Cytokines/metabolism , Myocardium/metabolism , Myocardium/pathology , Chromans
10.
Invest New Drugs ; 41(6): 816-824, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37902905

ABSTRACT

Myocarditis is a rare immune-related adverse events (irAEs) with high mortality rates, with few reports on its clinical characteristics and prognostic impact. This study designed to explore the associations between cardiac parameters and outcomes of myocarditis in advanced non-small cell lung cancer (NSCLC) who treated with immune checkpoint inhibitor (ICI). Fourteen patients diagnosed with ICI-associated myocarditis by clinicians were admitted to the study analysis. By Cox univariate and multivariate survival analyses, potential risk factors for the development of severe myocarditis were identified. Survival analysis was also performed to explore the prognosis of patients with myocarditis. Among patients with myocarditis, higher B-type natriuretic peptide (BNP) levels (P = 0.04) and conduction block (P = 0.03) were associated with progression to severe myocarditis. In addition, high lactate dehydrogenase (LHD) levels (P = .04) and myocarditis onset within 2 months (P = 0.02) were prognostic factors of severe myocarditis. The median progression-free survival (PFS) time and median overall survival (OS) time for all patients were 5.9 months and 18.5 months, respectively. However, there were no statistical differences between mild and severe cohorts in terms of PFS and OS (PFS: 4.5 vs. 8.5 months, P = 0.17; OS: 21.3 vs. 18.5months, P = 0.36). And we found that the earlier occurrence of myocarditis, worse PFS prognosis (4.5 months vs. 10.5 months, P = 0.008), while no difference in OS (18.5 months vs. 21.3 months, P = 0.35). Compared to mild myocarditis, severe myocarditis presented with higher BNP levels and cardiac conduction abnormalities. In addition, patients with mild and early myocarditis tended to have better survival rates.


Subject(s)
Antineoplastic Agents, Immunological , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Myocarditis , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Prognosis , Lung Neoplasms/drug therapy , Immune Checkpoint Inhibitors/adverse effects , Myocarditis/chemically induced , Myocarditis/drug therapy , Antineoplastic Agents, Immunological/therapeutic use , Retrospective Studies
11.
Invest New Drugs ; 41(2): 333-339, 2023 04.
Article in English | MEDLINE | ID: mdl-36988830

ABSTRACT

BACKGROUND: To investigate the clinical features of nivolumab-induced myasthenia gravis (MG) and provide evidence for the rational use of nivolumab in the clinic. METHODS: We collected case reports and case series of nivolumab-induced MG for retrospective analysis by searching Chinese and English databases from 2014 to October 31, 2022. RESULTS: Of the 67 patients included, the median age was 72.5 years (range 34-86), including 44 males (65.7%). MG occurred in the median 2nd treatment cycle (range, 1st-6th) after nivolumab treatment, being mild in 12 patients (17.9%) and moderate to severe in 44 patients (65.7%). Ptosis (n = 48,71.6%), diplopia (n = 34,50.7%), dyspnea (n = 30, 44.8%), limb muscle weakness (n = 30, 44.8%) and dysphagia (n = 27, 40.3%) were the most common symptoms. Fifty-six patients (83.6%) were classified as having generalized myasthenia gravis (GMG), the remaining 11 patients (16.4%) isolated ocular myasthenia gravis (OMG). Twenty-one patients (31.3%) had MG combined with myositis, 10 patients (14.9%) had myocarditis, and 9 patients (13.4%) had both myositis and myocarditis. Forty patients (59.7%) were positive for anti-acetylcholine receptor antibodies. The serum creatine kinase level was significantly increased in 37 patients (55.2%), with a median value of 4000 IU/L (219,14229). After discontinuation of nivolumab and immunosuppressive therapy, 46 patients (68.7%) finally recovered or improved their MG symptoms, while 15 patients (22.4%) did not recover. Eleven patients (16.4%) died of MG complications. CONCLUSION: MG is a serious and rare adverse reaction to nivolumab. Nivolumab-induced MG should be timely and correctly identified, and immunotherapy should be given.


Subject(s)
Myasthenia Gravis , Myocarditis , Myositis , Male , Humans , Adult , Middle Aged , Aged , Aged, 80 and over , Nivolumab/adverse effects , Retrospective Studies , Myocarditis/chemically induced , Myocarditis/complications , Myocarditis/drug therapy , Myasthenia Gravis/chemically induced , Myasthenia Gravis/drug therapy , Myasthenia Gravis/complications , Myositis/chemically induced , Myositis/diagnosis , Myositis/drug therapy , Treatment Outcome
12.
Eur J Nucl Med Mol Imaging ; 50(3): 792-812, 2023 02.
Article in English | MEDLINE | ID: mdl-36334105

ABSTRACT

Cardiotoxicity may present as (pulmonary) hypertension, acute and chronic coronary syndromes, venous thromboembolism, cardiomyopathies/heart failure, arrhythmia, valvular heart disease, peripheral arterial disease, and myocarditis. Many of these disease entities can be diagnosed by established cardiovascular diagnostic pathways. Nuclear medicine, however, has proven promising in the diagnosis of cardiomyopathies/heart failure, and peri- and myocarditis as well as arterial inflammation. This article first outlines the spectrum of cardiotoxic cancer therapies and the potential side effects. This will be complemented by the definition of cardiotoxicity using non-nuclear cardiovascular imaging (echocardiography, CMR) and biomarkers. Available nuclear imaging techniques are then presented and specific suggestions are made for their application and potential role in the diagnosis of cardiotoxicity.


Subject(s)
Antineoplastic Agents , Cardiomyopathies , Heart Failure , Myocarditis , Neoplasms , Nuclear Medicine , Humans , Antineoplastic Agents/therapeutic use , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Cardiotoxicity/diagnostic imaging , Cardiotoxicity/etiology , Cardiotoxicity/drug therapy , Myocarditis/chemically induced , Myocarditis/drug therapy
14.
J Natl Compr Canc Netw ; 21(9): 894-899, 2023 09.
Article in English | MEDLINE | ID: mdl-37673116

ABSTRACT

Immune checkpoint inhibitors (ICIs) induce profound benefits in cancer patients with mismatch repair gene mutations or high levels of microsatellite instability. Herein, we present a case of a patient with history of Muir-Torre/Lynch syndrome and metastatic gastric adenocarcinoma in the presence of an MSH2 gene mutation. The patient was initially treated with a PD-1 inhibitor, pembrolizumab, but developed grade 4 myocarditis requiring treatment with infliximab and a prolonged steroid taper. Following discontinuation of pembrolizumab, surveillance testing showed no radiographic or endoscopic evidence of progression for 7 months, until biopsy results from a repeat upper endoscopy indicated local disease recurrence. The patient was subsequently rechallenged with another PD-1 inhibitor, nivolumab, at a 50% dose reduction without recurrent adverse events and eventually achieved a complete response after 13 cycles. This case highlights the relative importance of considering careful rechallenge with ICI therapy in patients with microsatellite instability-high malignancies and a high risk of severe adverse events.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Myocarditis , Humans , Immune Checkpoint Inhibitors/adverse effects , Colorectal Neoplasms, Hereditary Nonpolyposis/drug therapy , Microsatellite Instability , Myocarditis/drug therapy , Myocarditis/etiology , Nivolumab/adverse effects
15.
BMC Cardiovasc Disord ; 23(1): 328, 2023 06 28.
Article in English | MEDLINE | ID: mdl-37380955

ABSTRACT

BACKGROUND: Monkeypox is a zoonotic viral infection first reported in May 2022. Monkeypox cases present with prodromal symptoms, rash, and/or systemic complications. This study systematically reviews the monkeypox cases presented with any cardiac complications. METHODS: A systematic literature search was done to locate papers that discuss any cardiac complications associated with monkeypox; then, data were analyzed qualitatively. RESULTS: Nine articles, including the 13 cases that reported cardiac complications of the disease, were included in the review. Five cases previously had sex with men, and two cases had unprotected intercourse, which reveals the importance of the sexual route in disease transmission. All cases have a wide spectrum of cardiac complications, such as acute myocarditis, pericarditis, pericardial effusion, and myopericarditis. CONCLUSION: This study clarifies the potential for cardiac complications in monkeypox cases and provides avenues for future research to determine the underlying mechanism. Also, we found that the cases with pericarditis were treated with colchicine, and those with myocarditis were treated with supportive care or cardioprotective treatment (Bisoprolol and Ramipril). Furthermore, Tecovirimat is used as an antiviral drug for 14 days.


Subject(s)
Mpox (monkeypox) , Myocarditis , Pericardial Effusion , Pericarditis , Male , Humans , Myocarditis/diagnosis , Myocarditis/drug therapy , Heart , Pericarditis/diagnosis , Pericarditis/drug therapy , Pericardial Effusion/etiology
16.
Support Care Cancer ; 31(9): 518, 2023 Aug 12.
Article in English | MEDLINE | ID: mdl-37572133

ABSTRACT

PURPOSE: Myasthenia gravis (MG) is a rare but life-threatening complication of immune-checkpoint inhibitor (ICI) therapy and often co-presents with myositis and myocarditis. Previous case series of ICI-related MG have reported high mortality rates. We present a series of ten patients from a tertiary oncology centre outlining outcomes of an early multi-modal immunosuppression strategy. METHODS: We reviewed The Christie Hospital database of immunotherapy-related toxicity from 2017 to 2020. Symptom severity was assessed using the Myasthenia Gravis Foundation of America (MGFA) classification. RESULTS: Ten patients with ICI-related MG were identified. All patients presented following 1 (n = 4) or 2 (n = 6) cycles of ICI. Symptom progression was rapid with a median of 3 days from onset of symptoms to admission. Concomitant myositis and myocarditis were observed in nine patients. AChR or MuSK autoantibodies were positive in six patients. All patients received urgent treatment with intravenous methylprednisolone (IVMP) and eight received intravenous immunoglobulin (IVIG). A single patient died from myasthenia-related symptoms; the remaining 9 patients were successfully discharged. CONCLUSION: In our cohort, we demonstrate good outcomes associated with early intensive immunosuppressive treatment with IVIG and IVMP. An agreed national treatment protocol or clinical discussion forum would be beneficial.


Subject(s)
Myasthenia Gravis , Myocarditis , Myositis , Humans , Immune Checkpoint Inhibitors/adverse effects , Immunoglobulins, Intravenous/therapeutic use , Immunosuppression Therapy , Myasthenia Gravis/chemically induced , Myasthenia Gravis/drug therapy , Myasthenia Gravis/complications , Myocarditis/chemically induced , Myocarditis/drug therapy , Myocarditis/complications , Myositis/chemically induced , Myositis/drug therapy , Myositis/complications
17.
BMC Pulm Med ; 23(1): 119, 2023 Apr 14.
Article in English | MEDLINE | ID: mdl-37060029

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) have been widely used in the treatment of cancer. Moreover, immune-related adverse events (irAEs) have become a new clinical challenge. ICI-associated myocarditis is a rare but fatal condition among diverse organ injuries, and early recognition and effective interventions are critical for patients. CASE PRESENTATION: In this report, we present the case of a healthy 60-year-old male who was diagnosed with lung squamous cell carcinomas following chemotherapy and received ICIs. The patient presented with asymptomatic cardiac biomarker elevation followed by immune-related myocarditis. Fortunately, the patient achieved a good clinical result after receiving high-dose steroids. The treatment with ICIs was discontinued because of recurrent increases in troponin T. CONCLUSION: ICI-mediated associated myocarditis is an uncommon but potentially life-threatening adverse event. The current data suggest that clinicians need to be cautious about reinitiation in low-grade patients; however, further study of the diagnosis and treatment is necessary.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Myocarditis , Male , Humans , Middle Aged , Carcinoma, Non-Small-Cell Lung/pathology , Immune Checkpoint Inhibitors/adverse effects , Lung Neoplasms/pathology , Myocarditis/chemically induced , Myocarditis/diagnosis , Myocarditis/drug therapy , Immunotherapy/adverse effects
18.
J Electrocardiol ; 80: 45-50, 2023.
Article in English | MEDLINE | ID: mdl-37187131

ABSTRACT

We present a case of a previously healthy 23-year-old male who presented with chest pain, palpitations and spontaneous type 1 Brugada electrocardiographic (ECG) pattern. Positive family history for sudden cardiac death (SCD) was remarkable. Initially, clinical symptoms in combination with myocardial enzymes elevation, regional myocardial oedema with late gadolinium enhancement (LGE) on cardiac magnetic resonance (CMR) and inflammatory lymphocytoid-cell infiltrates in the endomyocardial biopsy (EMB) suggested the diagnosis of a myocarditis-induced Brugada phenocopy (BrP). Under immunosuppressive therapy with methylprednisolone and azathioprine, a complete remission of both symptoms and biomarkers was accomplished. However, the Brugada pattern did not resolve. The eventually spontaneous Brugada pattern type 1 established the diagnosis of Brugada syndrome (BrS). Due to his previous history of syncope, the patient was offered an ICD that he declined. After his discharge he experienced a new episode of arrhythmic syncope. He was readmitted and received an ICD.


Subject(s)
Brugada Syndrome , Myocarditis , Male , Humans , Young Adult , Adult , Brugada Syndrome/complications , Brugada Syndrome/diagnosis , Myocarditis/complications , Myocarditis/diagnosis , Myocarditis/drug therapy , Contrast Media , Electrocardiography , Gadolinium , Syncope/diagnosis , Syncope/etiology
19.
Eur Heart J ; 43(36): 3463-3473, 2022 09 21.
Article in English | MEDLINE | ID: mdl-35831932

ABSTRACT

AIMS: Long-term results of the Tailored IMmunosuppression in virus-negative Inflammatory Cardiomyopathy (TIMIC) trial protocol have been evaluated. METHODS AND RESULTS: Eighty-five patients with endomyocardial biopsy-proven virus-negative chronic inflammatory cardiomyopathy were enrolled in the randomized, double-blind, placebo-controlled TIMIC trial and received prednisone and azathioprine (n = 43) vs. placebo (n = 42) for 6 months. Immunosuppressive treatment promoted an improvement in cardiac function in 88% of the cases compared with none of the patients in the placebo group, which were switched to a 6-month immunosuppressive therapy at the end of the 6-month study period. Long-term (up to 20 years) clinical outcomes of the whole cohort of 85 patients originally enrolled in the TIMIC trial (Group A) were compared with those of a 1:2 propensity score-matched control cohort of patients untreated with the TIMIC protocol (Group B) and followed for a comparable period of time. The primary outcome was a composite of cardiovascular death and heart transplantation. At long-term follow-up, the risk of cardiovascular death [hazard ratio (HR) 6.77; 95% confidence interval (CI) 2.36-19.45] and heart transplantation (HR 7.92; 95% CI 1.80-34.88) was significantly higher in Group B patients. Group A showed a persistent improvement in the left ventricular ejection fraction compared with Group B (HR 7.24; 95% CI 3.05-17.18). A higher number of Group B patients underwent implantable cardioverter defibrillator implantation. The incidence of recurrent myocarditis was similar between groups, and patients with evidence of a recurrent cardiac inflammatory process promptly responded to a TIMIC protocol application. CONCLUSION: Virus-negative inflammatory cardiomyopathy benefits from immunosuppressive therapy even after long-term follow-up. Recurrence appears to respond to a new TIMIC protocol application.


Subject(s)
Myocarditis , Azathioprine/therapeutic use , Follow-Up Studies , Humans , Immunosuppression Therapy , Myocarditis/drug therapy , Prednisone/therapeutic use , Stroke Volume , Treatment Outcome , Ventricular Function, Left
20.
Immunopharmacol Immunotoxicol ; 45(6): 780-787, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37339370

ABSTRACT

Neuromuscular associated respiratory failure is a rare toxicity of immunotherapy for malignant tumors. In most cases, it may overlap with the symptoms of the primary disease or myocarditis, myositis and myasthenia gravis, resulting in difficult etiological diagnosis. Early detection and optimal treatment are still topics that need attention. Here, a case of 51-year-old male lung cancer patient with sintilimab-associated myasthenia gravis, myositis, and myocarditis overlap syndrome involving the diaphragm who developed severe type II respiratory failure was reported. After high-dose methylprednisolone, immunoglobulin and pyridostigmine intravenous injection with non-invasive positive pressure ventilation, the patient's symptoms improved significantly and was discharged. One year later, the patient received immunotherapy again due to tumor progression. After 53 days, he developed dyspnea again. Chest X-ray demonstrated marked elevation of the diaphragm, and the electromyogram demonstrated dysfunction of diaphragm. With rapid diagnosis and timely treatment, the patient was finally discharged safely. A comprehensive search of PubMed, EMBASE was performed to identify all previously reported cases of immune checkpoint inhibitors-associated respiratory failure. The potential mechanisms of respiratory failure caused by ICI-associated diaphragmatic dysfunction may be related to T cell-mediated immune disturbances and we proposed possible diagnostic processes. For patients with unexplained respiratory failure who are receiving immunotherapy, standardized diagnostic strategies should be implemented immediately on admission before deciding whether to conduct a more invasive diagnostic procedure or empirical treatment.


Subject(s)
Antineoplastic Agents, Immunological , Lung Neoplasms , Myasthenia Gravis , Myocarditis , Myositis , Respiratory Insufficiency , Male , Humans , Middle Aged , Immune Checkpoint Inhibitors/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Myocarditis/chemically induced , Myocarditis/drug therapy , Lung Neoplasms/drug therapy , Myasthenia Gravis/diagnosis , Myasthenia Gravis/drug therapy , Myositis/chemically induced , Myositis/drug therapy , Myositis/pathology , Respiratory Insufficiency/chemically induced , Respiratory Insufficiency/therapy
SELECTION OF CITATIONS
SEARCH DETAIL