Skip to main content

A comprehensive review on immune checkpoint inhibitors induced cardiotoxicity characteristics and associated factors

Abstract

Newly approved cancer drugs called ICIs have shown remarkable success in improving patient survival rates, but they also have the potential for inflammatory and immune-related side effects, including those affecting the cardiovascular system. Research has been conducted to understand the development of these toxicities and identify risk factors. This review focuses on the characteristics of ICI-induced cardiotoxicity and discusses the reported risk factors. It is important for cardio-oncologists to understand the basic concepts of these drugs to better understand how cardiotoxicities occur. It might be hard to find reports, where all patients treated with ICIs had developed cardiac toxicity, because there could be other existing and variable factors that influence the likelihood or risk of developing cardiotoxicity during treatment. Various clinical parameters have been explored as potential risk factors, and further investigation is needed through large-scale studies.

Background

Since 2011, immune checkpoint inhibitors, hereby referred to as ICIs, have become an essential part of cancer immunotherapy, particularly with the approval of anti-CTLA-4 for advanced melanoma. These ICIs have greatly impacted the field, and since 2016, other monoclonal antibodies, such as anti-PD-1 and anti-PD-L1, have also gained acceptance in oncology therapy guidelines. These ICIs are being rapidly approved by the FDA to treat various types of cancer, greatly improving patient survival rates compared to traditional chemotherapy [1, 2]. A study involving patients with advanced lung cancer found that using pembrolizumab as a first treatment resulted in better outcomes than chemotherapy. This led to the approval of pembrolizumab by the FDA [3]. However, this great improvement added significant systemic inflammatory response potential and immune-related effects affecting diverse systems, among which the cardiovascular system has been associated with their use [4, 4]. Felice Crocetto et al. highlighted through a reliable meta-analysis that despite the benefit attached to the use of ICIs in combination with anti-VEGF (vascular endothelial growth factor) versus anti-VEGF single therapy. Patients receiving anti-VEGF therapy with ICI had a higher risk of developing cardiac and blood-related clotting disorders than those who only received anti-VEGF therapy [150]. The 2022 meta-analysis by Maobai Liu et al. showed varying incidences of cardiotoxicity among different ICI therapies. For ICI monotherapy, CTLA-4 may be associated with higher grade 3–5 cardiotoxicity than PD-1 or PD-L1 for dual therapy. The cardiotoxicity of dual ICI therapy seems to be higher than that of chemotherapy or targeted therapy [151].

Because most patients did not undergo systematic and routine cardiovascular status monitoring, cardiac adverse events seemed under-reported in the literature. Initial investigations revealed that mice deficient in PD-1 developed dilated cardiomyopathy and severe myocarditis [6,7,8]. According to a 2016 pharmacovigilance analysis report, approximately 0.09% of patients treated with nivolumab, another PD1 ICI, had developed myocarditis; when combined with ipilimumab, the incidence of myocarditis was approximately 0.27%. Other cardiotoxic effects reported include pericarditis, pericardial effusion, cardiomyopathy, and new arrhythmias [9,10,11]. The incidence of ICI-induced cardiac toxicity is relatively low, ranging from less than 1% to approximately 18%, depending on the specific ICI and patient population studied. However, it can be a severe and potentially life-threatening complication.

Current investigations focus on how ICIs cause heart-related side effects and the identification of risk factors. The currently established risk factors associated with ICI-induced cardiac toxicity include pre-existing cardiovascular disease, concomitant medications, ICI type and dosage, prior exposure to cardiotoxic therapies, autoimmune diseases, age, and sex, most of which are considered traditional cardiovascular risk factors. Nevertheless, several other factors are sparse in the literature. It would still be interesting to look closely at them, as close monitoring is needed to detect and manage any potential cardiac toxicity promptly. This review focuses on the characteristics of ICI-induced cardiotoxicity and discusses the probable ICI-induced cardiotoxicity risk factors from the available literature. This concise and focused review might help design cardiotoxicity risk stratification in the setting of ICI-induced cardiotoxicity.

Overview of ICIs

Definition

ICIs block the function or effect of immune checkpoints, which are immune components expressed on the cell surface of T lymphocyte cells. There are two types of ICIs: stimulatory checkpoints that potentiate immune cell action and activation (TNF, CD27, CD40) and inhibitory checkpoints that downregulate the immune response (CTLA-4, PD1, IDO, KIR, LAG3) [12,13,14,15]. The inhibitory type is targeted by ICI therapy. In 1968, researchers discovered that lymphocyte cells from cancer patients could react against cancer cells in vitro. These inhibitory checkpoints were identified in 1995 by Ph.D. Jim Allison [16,17,18,19,20]. Jim Allison discovered that the protein CTLA-4 controls T-cell activation and recruitment [21,22,23]. PD1 is another immune checkpoint receptor that intervenes at two levels: differentiation of immature precursor T cells into effector and memory T-cell populations and activation or reactivation of circulating or resident effector and memory T-cell subsets. Blocking PD1 improves antitumour CD8 + T-cell cytotoxic capacity by reducing the tumor-suppressive impact of PDL1 and PDL2 produced by neoplastic cells [22,23,24,25,26,27,28,29,30].

Classification of ICI drugs

ICIs are classified based on the type of immune checkpoint. FDA-approved immune checkpoint blockade drugs have three main subclasses that share almost the same indications and adverse effects but have distinct pharmacodynamic and pharmacokinetic properties [27,28,29,30,31,32]. In addition, new generations of ICI drugs, including LYMPHOCYTE ACTIVATION GENE-3 INHIBITORS (LAG-3 INHIBITORS), are still being investigated for potential clinical use. Table 1 summarizes the ICI drug classification, FDA-approved year, and indications [31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47].

Table 1 Classification of ICIs

Immune checkpoint inhibitors and cardiotoxicity

Mechanism of immune checkpoint inhibitor therapy-induced cardiotoxicity (Fig. 1 )

Fig. 1
figure 1

Simplified mechanism of ICI-induced cardiotoxicity

According to the 2019 World Health Organization's global database analysis, on 12 455 401 ICI drug case safety reports, patients who received ICIs had an 11-fold higher likelihood of reporting myocarditis than those who did not receive ICIs [48, 49]. Furthermore, based on the Joe-Elie Salem et al. 2018 study, ICI treatment has related to other inflammatory cardiovascular side effects, including pericardial diseases and vasculitis, with a higher occurrence of temporal arteritis. Reports have also linked ICIs with noninflammatory cardiovascular toxicity, such as Takotsubo-like syndrome. However, it was difficult to solely attribute these effects to pembrolizumab, because patients may have received other medications known for their chronic cardiotoxic properties, such as trastuzumab [50, 51].

Other cases of noninflammatory cardiovascular toxicity have been reported, including symptom-free, noninflammatory left ventricular dysfunction, myocardial infarction, and coronary vasospasm. [52,53,54]. It is now recognized that arrhythmias can indicate cardiotoxicity in patients undergoing ICI treatment. However, arrhythmias are common among individuals with cancer and often occur alongside other immune-related adverse events. Examples of these events include acute thyrotoxicosis observed in ICI-mediated thyroiditis [48, 54]. In case reports, ICI-associated third-degree atrioventricular block and conduction disease were often attributed to conduction system disturbances secondary to myocarditis [7, 55, 56].

The mechanisms underlying cardiovascular irAEs are not well-understood. For example, ICI-induced myocarditis is characterized by infiltration of macrophages and T lymphocytes in the myocardium, leading to cell damage and death [57]. ICI-associated myocarditis involves the infiltration of CD4 + and CD8 + T cells and CD68 + macrophages into the myocardium and conduction system. B cells are in short supply. Understanding cellular tolerance and triggers for T-cell infiltration is crucial. A case series study found consistent immune aberrancy in patients with fulminant myocarditis, mainly involving striated muscle and tumours [58]. Multiple tissue types showed robust T-cell infiltration, activation, and clonal expansion with evidence of shared high-frequency T-cell receptors. The probable mechanistic hypotheses that were then proposed included the following:

  1. 1)

    T cells targeted an antigen that was simultaneously present across tumour, skeletal muscle, and heart tissues; this could be supported by the finding of high levels of muscle-specific antigens (desmin and troponin) in both patients' tumours,

  2. 2)

    The same T-cell receptor targeted a tumour antigen and a different but homologous one sharing the same spatial conformation with a specific muscle antigen

  3. 3)

    Clonal, high-frequency T-cell receptor sequences across tumour and muscle samples could be misleading, and distinct T-cell receptor specificities target dissimilar antigens

Although extensive viral profiling revealed no clear etiology, it was hypothesized that subclinical viral infection could have generated T-cell targets. However, that study failed to prove the existence of common HLA alleles among patients. This led to a nonplausible HLA/drug hypersensitivity association theory. Therefore, the underlying causes of T-cell reactivity to myocardial and other striated muscle tissue are unknown and are certainly not universal across patients.

Currently, the available theories in research mainly focus on understanding the early mechanism of immune-related reactions induced by ICIs [59, 60]. Studies have shown that immune checkpoint inhibition can lead to myocarditis. In mouse models of T-cell-mediated myocarditis, PD-1 plays a significant role in protecting against inflammation and myocyte damage [61]. However, there is a notable amount of PDL1 expression in the heart muscle of individuals with ICI-induced fulminant myocarditis, which aligns with the increased level of this marker found in preclinical studies and mouse models. This suggests that PDL1 may play a protective role in preventing heart damage [62,63,64]. PDL1 upregulation has been linked to a cytokine-induced mechanism that protects the heart and is now disrupted by ICI blockade [64]. Activated immune cells can infiltrate normal muscle cells, including those in the heart, due to similarities between tumors and body muscle antigens. This can cause cardiotoxicity via complex mechanisms, including direct cell killing and increased pro-inflammatory cytokine levels. This is considered “bystander effect” heart damage. Approval for cancer immunotherapy has been granted to cytokines, such as IL-2, which have anticancer properties; however, they are also rare causes of myocarditis, occurring in 1.5% of 652 cases [65, 66]. High levels of cytokines such as IL-2, IFN-y, TNF, IL-1, and IL-6 have been linked to myocarditis. This condition causes inflammation and damage to the heart muscles. Studies have shown that 6 out of 8 patients with high IL-2 levels had myocarditis. Similar results were observed in autopsies of subjects with elevated levels of other cytokines [67,68,69]. Activated lymphocytes can indirectly affect heart tissue through the release of interferon-alpha and interleukin-2. CD8 + T cells produce interferon-gamma and TNF-alpha when activated by Th1 cells. Blocking immune checkpoints could lead to potential tumor destruction but also harm heart tissue due to increased cytokine levels.

ICI therapy can affect immune function in immune-privileged organs, such as the heart, which has few T cells and defensive mechanisms against T-cell attacks. The myocardium secretes IFN-γ and upregulates PD-L1 to reduce T-cell damage and prevent the growth of T helper cells, causing myocarditis. [136,137,138]. The illustration below summarizes the possible patterns that could be a rationale for cardiac damage after immune checkpoint blockade in the oncological management setting.

Spectrum of ICI-induced cardiotoxicity

The heart is vulnerable to inflammation and damage due to its dense vascularity, which can cause arrhythmias. Cardiovascular toxicities from ICI therapy are becoming more common and can result in myocarditis, pericardial illnesses, vasculitis, Takotsubo syndrome, conduction problems and unstable atherosclerotic lesions [70]. A 2014–2019 pharmacovigilance study by Chenxin et al. also reported that the spectrum of ICI-induced cardiotoxicity differed between ICI drug types and regimens but shared some similarities. The top five cardiac adverse events recorded in the database were dyspnea (21%), myocarditis (5.16%), atrial fibrillation (4%), cardiac failure (4%), and pericardial effusion (3.5%). In a comprehensive analysis by Hu et al. comprising 22 clinical trials evaluating PD-1 and PD-L1 inhibitors for lung cancer, the frequency of myocarditis was 0.5%. However, the frequency of other cardiovascular adversities, such as pericardial tamponade, myocardial infarction, stroke, cardiac failure, and cardiorespiratory arrest, varied between 0.7% and 2.0%.

Myocarditis is an inflammatory disease of the myocardium caused by several factors, such as viral infections, toxins, hypersensitivity reactions, and autoimmune disease. Autoimmunity has strongly been implicated in the etiology and progression of myocarditis. Many murine and clinical studies have reported on the plausibility of myocarditis onset in association with ICI use [71, 72]. The number of myocarditis reports is gradually growing, consistent with the increasing trend of ICI-induced cardiac damage. The prevalence of myocarditis and reporting is expected to continue to rise over time. Anti-PD-1, especially nivolumab, was hypothesized to have a more robust signal value in myocarditis [73]. Myocarditis is known to occur in the acute stage of ICI treatment. A past study found that the median time to onset from initiation of therapy to occurrence of symptoms was approximately 30 days [74]. Therefore, ICI-induced myocarditis seemed to be early onset cardiac toxicity in single and combination ICI therapy settings, although most cases were observed with a single therapy [74,75,76]. The clinical tableau can differ from asymptomatic cardiac biomarkers to severe decompensation, with the propensity for end-organ damage. The American Society of Clinical Oncology (ASCO) clinical practice guidelines have proposed a classification and grading system for myocarditis as shown below [77,78,79] (see Fig. 2). When myocarditis is suspected during ICI treatment, a thorough workup is recommended regardless of severity. This includes cardiac markers, electrocardiography, thoracic radiography, echocardiography, and referral to a cardiologist for further testing, such as cardiac magnetic resonance imaging, coronary angiography, and endomyocardial biopsy [77]. Medical practitioners may not have complete knowledge about myocarditis, leading to uncertainty about when to suspect or consider the condition. Increased troponin levels without symptoms may indicate mild myocarditis, but other factors can also cause troponin elevation [80]. Consequently, another classification of myocarditis was suggested as a three-level categorization: definite myocarditis, probable myocarditis, and possible myocarditis [81]. Cardiac MRI is still the best and least invasive way to diagnose myocarditis, even though endomyocardial biopsy is the gold standard [80]. Overall, it would be ideal for patients with suspected myocarditis to go through most diagnostic studies listed above until more definitive evidence becomes available.

Fig. 2
figure 2

ICI-induced myocarditis severity grading

Immune checkpoint inhibitors (ICIs) may increase cancer patients' risk of pericardial disease, which could lead to higher mortality rates. ICI-induced pericardial disease is rare and has varying incidences and presentations, which may cause delayed diagnosis and treatment. A recent pharmacovigilance study recorded that the incidence of ICI-induced pericardial disease, including pericarditis and pericardial effusion, was estimated at 0.36% [82]. Pericardial disease can occur alongside myocarditis or on its own, leading to pericardial effusion and cardiac tamponade. Previous research has identified several cases of pericardial disease associated with immune checkpoint inhibitor (ICI) treatment, with nivolumab being the most common. Most cases showed symptoms of tamponade, while some had effusive–constrictive physiology. Pericarditis typically developed 6 weeks and 11 months after starting ICI treatment, with one exception occurring 4 days later. Pericardiocentesis was the main treatment option, performed in five cases. Analysis of the pericardial fluid showed the presence of white blood cells, mostly lymphocytes, and no signs of cancer [83,84,85,86,87]. Pericardial disease from ICI can cause chest pain, difficulty breathing, and respiratory failure. Diagnosis involves identifying pericarditis, which can lead to effusion and tamponade [88, 89]. CT scans show effusion and thickening, while MRIs show inflammation. Coexisting myocarditis may raise troponin levels [80, 88].

Cancer treatment-induced arrhythmia (CTIA) is a potential side effect that may occur during chemotherapy. It can result in diverse types of irregular heart rhythms, including fast and slow heartbeats, which may lead to a complete heart block. Patients receiving immune checkpoint inhibitors have reported several cases of CTIA [90, 91]. In a retrospective study, 268 patients who underwent immune checkpoint inhibitor therapy were examined, and it was discovered that only 1.5% of them had a clinically significant arrhythmia within 6 months. The study also found that patients who had a previous diagnosis of atrial fibrillation were more likely to experience relapse while on immune checkpoint inhibitor therapy. The conclusion of the study was that immune checkpoint inhibitors are generally well-tolerated and safe regarding arrhythmias. A different study, which used a different database, also found similar results, but it highlighted that certain factors, such as thyrotoxicosis, may contribute to the development of atrial fibrillation [91, 92]. There is a hypothesis that drug-induced arrhythmias caused by immune checkpoint inhibitors (ICIs) tend to occur shortly after starting ICI treatment. This hypothesis applies to all types of ICI regimens and suggests that arrhythmias typically develop within 1 month of starting ICI treatment [92, 93]. Diagnosing ICI-associated arrhythmia and myocarditis lacks clear criteria. Abnormalities in ECGs and echocardiograms can serve as indicators. No established biomarkers exist for predicting outcomes. Clinicians must describe and evaluate adverse drug events [94, 95]. A recent study showed that starting ICI therapy can increase the risk of adverse arrhythmic events, with a 26% mortality rate associated with CTIA. It is crucial to be aware of this risk and not overlook CTIA.

Athero-cardiovascular toxicities include large vasculitis, coronary artery disease, thromboembolic events, and even myocardial infarction. Evidence suggests that ICIs significantly contribute to atherogenic T-cell activation, atherosclerosis development and coronary function regulation. PD1-depleted mice exhibited increased development of atherosclerotic lesions compared to controls. T cells have been found to play a pivotal role in advancing atherosclerosis towards more advanced, clinically unfavorable lesions. In addition, they have been directly implicated in plaque rupture and subsequent development of acute cardiovascular events [48, 53, 96]. ICIs activate proatherogenic T-cell immunity, increasing interferon and tumor necrosis factor production and raising the risk of coronary thrombosis [97]. Recently, some cases of retrosternal chest pain that did not meet the myocarditis diagnostic criteria and were more like acute coronary syndrome angina pain have been identified among patients taking ICI medication [98, 99]. According to a pharmacovigilance study, this type of cardiotoxicity accounted for 0.53% of all case safety reports. It was reported to be more common than ventricular arrhythmia (0.07%) and cardiac death (0.43%) [49]. In the JOCARDITE registry study (n = 474), only 55.1% of ICI-myocarditis patients underwent coronary angiography, and 22.6% had concomitant CAD. In a recent pharmacovigilance study, CAD represented one-third of all cases [100]. It is possible that many patients diagnosed with ICI-induced myocarditis have coronary artery disease, leading to a lower number of reported cases of CAD in studies. Various imaging studies have shown that ICIs can contribute to inflammation of large blood vessels and the formation of atherosclerotic plaques, not limited to CAD [101,102,103]. One could argue that atherosclerosis is a slow process that takes a long time before its symptoms or complications become apparent. In patients, the presence of conventional cardiology risk factors may have contributed to the development of ICI-induced CAD and vasculitis. However, currently, traditional risk factors such as age, obesity, and smoking are still considered independent of the risk of ICI-induced cardiac toxicity [104]. CT scans can predict the risk of atherosclerosis in coronary arteries. More research is needed to determine the risk of mortality in ICI-induced CAD [105]. Coronary angiography could become a routine investigation for cancer patients on ICI therapy.

We have presented the major type of cardiac toxicity occurring with ICI medication. However, the list may not be exhaustive. Cases of Takotsubo were also reported and confirmed by negative endomyocardial biopsy [106,107,108]. Takotsubo cardiomyopathy is linked to advanced malignancies and potential triggers, such as emotional disturbances, cancer treatment, and chemotherapy. The pathophysiology is unclear, although hypotheses include coronary vasospasm, microvascular dysfunction, and excessive stress response. Facts from rodent model studies have suggested that inflammation plays a crucial role in Takotsubo cardiomyopathy [106, 108].

Hypertension is also a reported side effect of ICI use. Nevertheless, a recent meta-analysis did not find a significant increase in the short-term risk of hypertension among patients treated with these drugs [109]. ICI-induced hypertension and atherosclerosis are likely related. A case of pulmonary artery hypertension was seen in a non-smoking African American woman with lung cancer who was treated with chemotherapy and an ICI drug. She also developed insulin-dependent diabetes, hypothyroidism, and adrenal insufficiency [110]. Therefore, the greater propensity of ICI drugs to lead to autoimmune conditions puts every patient taking those drugs on the higher watch for autoimmune-related pulmonary artery hypertension (PAH type 1). Finally, heart failure is a long-term complication of both cancer and cancer treatment-induced cardiac toxicities and has been consistently reported in patients with ICI-induced myocarditis.

Parameters correlating with increased risk of ICI-induced cardiac toxicities

Identifying literature reports, where all patients who received ICI drugs developed cardiac toxicity is challenging due to other risk factors that may affect the outcome. Various parameters that vary from patient to patient have been analysed to determine if they could be considered risk factors. These parameters have been classified into three types, which are outlined in Table 2.

Table 2 Parameters associated with risk ICI-induced cardiac toxicity

General parameters

Ethnicity or race is a crucial factor with higher variability that can impact diagnosis, treatment, and prognosis. For instance, the administration of beta-blockers and ACEis in the hypertensive management approach, atherosclerotic cardiovascular disease score risk (ASCVD), and EGFR estimation depend on the patient’s ethnicity [111,112,113]. In addition, an individual's self-identified race is a reliable indicator of overall health and longevity [112]. Therefore, the rarity of studies addressing the impact of ethnicity on the risk of ICI-induced cardiac toxicity would be paradoxical. Zakary et al. 2022, a retrospective study, found that from 468 Caucasians exposed to ICI therapy, only 19 developed cardiotoxicities, while for 57 African American natives, only 7 developed the outcome. Therefore, the difference in cardiac toxicity percentage was significant (P < 0.05). This made the African American ethnicity or race more susceptible to the onset of cardiac events when exposed to ICI [111,112,]. However, although significant, this result would give clues but not answer the question. There is still a pending for some high-level perspective and meta-analysis studies to conclude the matter.

In medicine, gender is a crucial factor to consider. For example, if lung cancer is more prevalent in females, it could be because more women are exposed to ICIs than men, leading to higher rates of cardiac toxicity among women. However, when examining the range of cancer types that can be treated with ICIs, it is important to look at studies that include all types of cancers to determine if there is a consistent gender pattern of association with the risk of cardiac toxicity. Zakary et al. found that a significantly higher percentage of women experienced cardiac events compared to men. However, Maria et al.'s study did not find any significant gender association with the risk of ICI-induced cardiotoxicity [114]. However, this discrepancy is not without reason. We would like to recall that male sex is generally a risk factor for cardiovascular pathology. In contrast, females have a greater tendency to have autoimmune disease and lung cancer than men [115, 116]. Because the autoimmune reaction is also encountered with ICIs, it appears hard to predict which gender carries a greater risk for cardiac toxicity. While we wait for proper specification, patients from all genders would still need equal attention during screening, monitoring, and follow-up after initiation of ICI.

Age would be a constant parameter. The spontaneous or natural risk for cardiovascular disease increases with age; on the other hand, the severity of cancer and the effectiveness of response to therapy are all influenced by the age of the patients [117,118,119]. According to the universal theory, patients who are more advanced in age are at a higher risk of developing comorbidities, including when undergoing ICI therapy. Studies have shown that even myocarditis, which typically affects younger patients, can occur with ICI treatment, and the risk increases with age. In the Maria et al. study, the case group had a mean age of 65 years compared to 59 years in the control group, and this difference was statistically significant [114]. Consequently, when evaluating patient ICI-induced cardiotoxicity risk, cardio-oncologists could consider the elderly as a higher risk cohort and accentuate their therapeutic surveillance.

Some patients may be unable to receive adequate cancer treatment due to the excessive cost, leading to severe complications. Therefore, healthcare providers must consider a patient's financial status when reviewing their medical history, as it can influence their treatment plan. Each patient's economic situation is unique and should be given proper consideration. At present, guidelines do not prioritize financial status when figuring out care, diagnosis, and follow-ups. However, this should be re-evaluated [120]. While health is often said to be priceless, the goal of medicine should be to supply healthcare for everyone.

Clinical parameters

Studies addressing the association between cardiovascular risk factors and the risk of ICI-induced cardiac toxicity, for the majority, assessed smoking, BMI, hypertension, and diabetes mellitus. Zachary et al. reported a significant association between smoking and the risk of ICI-induced cardiotoxicity.

A study found that among the 354 smokers analysed, there was a significant incidence of cardiac toxicity (4.212 [1.289, 13.763] P < 0.05). Smoking is a well-known risk factor for lung cancer and can negatively affect various treatments for the disease. Smoking increases the presence of PD-L1, impairs the body's ability to fight inflammation, and allows cancer cells to go undetected by the immune system. It also causes inflammation in the tumor microenvironment, promoting tumor growth and exhausting T cells. [121, 122]. It was also hypothesized that smoking history in patients suffering from NSCLC significantly determines their response to ICI treatment. It seemed to depict a trend towards improvement [123]. Therefore, the action of ICI is potentiated by the presence of a smoking history; tobacco components act in synergy with ICI drugs, which could increase the risk of immune-related adverse events, such as myocarditis and pneumonitis. As a result, it could be thought that smokers may be associated with a double the risk for cardiac events, one from the tendency of smoking to induce atherosclerosis and another from the activation of T cells.

Zachary et al. also reported a nonsignificantly increased risk of developing cardiac events with BMI, hypertension, or diabetes mellitus type II; a similar observation trend could also be found in the Maria et al. report. Whether ICI can provoke hypertension has been discussed in the earlier section. However, the occurrence of cardiac events among hypertensive patients taking ICIs appears independent of the ICI drug effect. However, patients with hypertension have a greater risk for other cardiac diseases, such as ischemic heart disease and arrhythmia, whose histories were significantly associated with the onset of cardiac events in a multivariate analysis [114]. Therefore, cardiovascular disease history would be a significant predisposing factor. However, the mechanism of this association still needs to be further elucidated.

A recent study found that people with both type 2 diabetes and cancer may have worse outcomes when receiving immune checkpoint inhibitor therapy. The study involved 1395 patients with advanced solid tumours who received this therapy between 2014 and 2020. The analysis showed that patients taking diabetic medication had shorter overall survival and progression-free survival than those who were not taking the medication or likely did not have diabetes [124]. Although incomplete, because the study did not specify cardiotoxicity development among those patients. However, at least it appears clear that a history of diabetes may affect the outcome of ICI. Nevertheless, possible side effects result from ICI use, in people with diabetes present a 2–4 times greater risk for cardiac-related adverse events [125, 126]. Further studies need to be deployed to evaluate the effect of DM on the risk of ICI-induced cardiotoxicity. Nevertheless, diabetes mellitus should always be considered when estimating the risk of any cardiovascular pathologies, including cancer treatment-induced pathologies.

Obesity is a significant cardiovascular risk factor, and some studies have suggested that there is reduced efficacy of cancer treatment among obese patients for some types of treatments, particularly chemotherapy because of underdosing by providers. Nonetheless, recent research underscores the obesity paradox, which points to a correlation between increased body mass index (BMI) and favorable results in cases treated with immune checkpoint inhibitor (ICI) therapies [126, 128]. However, little data on its correlation with the risk for ICI-induced cardiotoxicity is discernible in the literature, but obesity increases exhausted T cells, with mice showing higher PD-1 expression. CD4 + and CD8 + T cells displayed reduced proliferation and cytokine production when stimulated ex vivo in mice and humans [54]. Obese melanoma patients over 60 show increased PD-1 and other exhaustion markers and lower T-cell proliferation [129]. Obese individuals with important levels of the hormone leptin may have increased expression of PD-1, a protein found in tumor-infiltrating lymphocytes, which can lead to worse cancer outcomes. Leptin triggers STAT3 activation, leading to increased transcription and translation of PD-1. This can ultimately lead to reduced T-cell function and proliferation, negatively impacting cancer treatment. However, high BMI may have a positive effect on immunotherapy effectiveness, but administering immunotherapy to obese patients may increase immune-related adverse effects [130]. Obese mice in the non-ICI group had chronic inflammation. Overweight patients have a higher chance of immune-related adverse events. However, the effects are not severe. Treatment effectiveness increases the risk of adverse events, and weight loss does not have significant benefits [131]. Zachary et al. found that obesity was significantly associated with mortality in ICI settings.

Reduced glomerular filtration, as seen in renal failure, has been significantly linked to the onset of immune-related renal adverse events [132]. Although the Maria et al. study found a nonsignificant association between a history of chronic renal failure and risk for cardiac events in patients taking ICI medications, it could still be possible to identify an indirect link between pretreatment reduced glomerular filtration rate and risk for cardiac toxicity. Pretreatment reduced GFR, leading to renal adverse events, which, in turn, affect the heart in numerous ways. Reduced ejection fraction is widely accepted as a cardiovascular pathology risk factor [133]. Therefore, renal patients undergoing ICI therapy courses should be proactively managed and followed up concerning cardiovascular toxicities.

Treatment-related anemia can raise heart rate and increase the risk of heart failure and myocardial ischemia in patients with high heart rates before treatment. Further research is needed to confirm this theory. Fever can affect the immune response and tumor microenvironment, but mild hyperthermia combined with immune checkpoint inhibitors has shown promise in preclinical studies and appears safe [134, 135]. If the action of ICIs is promoted or increased, the propensity for cardiac toxicity also increases; thus, baseline fever, which might also indicate an infection in an ICI candidate, could be considered a predisposing factor for cardiotoxicity, and this remains to be demonstrated with future studies.

Paraclinical

This section explores nonclinical parameters such as inflammatory or indices, cardiac biomarkers, genetic cancer biomarkers, imaging, and therapy-related parameters to show their relationship with cardiac toxicity in an ICI setting.

Inflammation is the cornerstone mechanism by which ICI achieves its efficacy and is also a cancer-related effect [102]. Inflammation is an immune reaction. Therefore, amplifying the immune response with ICIs equals amplifying inflammation, which may lead to organ damage. No wonder most guidelines recommend using glucocorticoid or immunosuppressant therapy to manage ICI-induced toxicities, including cardiac ones [136]. As shown in previous reports, systemic inflammatory tools and indices could also be relevant for risk stratification concerning ICI-induced cardiac toxicity [105]. For instance, the platelet-to-albumin ratio (PAR), neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), lymphocyte-to-monocyte ratio (LMR), and C-reactive protein/albumin ratio (CAR), which are inflammatory indexes with prechemotherapeutic value, have been found to carry significant prognostic value in evaluating outcomes after therapy in pancreatic tumors [137,138,139,140]. Predictors that increase the risk of complications have been studied, but not in relation to ICI-induced cardiac toxicity. Immune biomarkers related to B and T cells can help assess risk. Recent studies have shown that decreased clonality and increased TCR diversity after treatment with a CTLA4-blocking antibody can lead to irAEs. CD8 + T-cell clone expansion in ICI-treated patients is also correlated with irAEs [141]. Autoantibodies can show the impact of immune checkpoint inhibitors on T and B cells, but their role in autoimmune disorders is unclear. Detecting activated T cells early on can help with patient care, and autoantibodies can predict if a patient is likely to experience immune-related adverse events. However, accurately characterizing these autoantibodies remains unresolved [142, 143].

Cardiac natriuretic peptides and troponins are useful markers in identifying patients who may be at risk of cardiotoxicity. Recent developments have made it possible to use these markers to diagnose early cardiotoxicity and predict late-onset cardiotoxicity. The current guidelines on cardio-oncology support their crucial role in detecting cardiotoxicity caused by cancer therapy [144]. The significance of monitoring troponin levels to detect cardiotoxicity has primarily been proven through investigations of individuals undergoing chemotherapy for cancer, particularly those receiving anthracyclines. A prolonged increase in troponin I levels is positively correlated with a greater degree of left ventricular (LV) dysfunction and a heightened likelihood of cardiac events compared to temporary fluctuations in troponin levels. BNP and NT-proBNP are crucial biomarkers for pressure overload, myocardial stretch, and cardiotoxicity detection. BNP can detect acute cardiotoxicity within 24 h of anthracycline chemotherapy and has been found to be useful in screening for HF in patients with dyspnea on cancer therapy. A study measured natriuretic peptide in a cohort of 600 oncologic patients, and high NT-proBNP levels indicated higher mortality risk: hazard ratio 1.54 (95% CI 1.24–1.90, p < 0.001) with a 67% 25-month survival rate compared to 49% for normal NT-proBNP levels [145, 146]. Therefore, measuring these biomarkers would help identify patients who could develop cardiotoxicity but may also help determine the degree of cardiac dysfunction [145, 146].

Some genetic markers, such as IL7 gene variants, may increase the risk of heart damage from immune checkpoint therapy. Certain biomarkers in lung cancer, such as epidermal growth factor and anaplastic lymphoma kinase, could reduce ICI treatment effectiveness. Tumors driven by the KRAS gene depicted good outcomes with ICI therapy [146, 147]. These observations provide evidence that genetic parameters can help predict therapy-induced toxicity. Further research is needed to determine which genetic parameters are relevant for immunotherapy-induced cardiac events. Cancer biomarkers such as CEA, NSE, CA125, and SCC are useful for monitoring treatment effectiveness and tumor severity. They may also have a potential link to ICI-induced heart damage. If these findings become available, they would significantly advance future cardio-oncology guidelines… [148, 149]

Past research has found several microRNA molecules that could potentially be biomarkers for heart toxicity. MicroRNA146a, miRNA 140-5p, and miRNA-377 have shown connections to cell death and mortality in animal models of doxorubicin-induced heart toxicity. However, further research is needed to understand how these molecules function, assess their variability, and confirm their usefulness in clinical applications for immune checkpoint inhibitor therapy.

Discussion and future perspectives

Cardiac toxicity from ICI treatment is a concern in cancer management. The low incidence suggests that certain factors may make individuals more susceptible. Cardio-oncologists need to understand the mechanisms and watch for any parameters that may lead to toxicity. The main mechanism is an exaggerated T-cell response in myocardial cells, with myocarditis being the most common type. Ethnicity, age, gender, financial status, history of chronic disease, inflammation biomarkers, CVD history and risk factors, heart rate, cardiac biomarkers, genetic biomarkers, fever, and microRNAs have all been associated with an increased risk for ICI-induced cardiac toxicity. However, these observations come from nonrandomized studies, and the strength of association was quite weak. Additionally, it was rare to find a parameter in isolation, as multiple parameters often interact with one another. Dual or combination of ICI therapy and non ICI therapy may also affect the risk of ICI induced cardiac toxicity [150, 151]. Creating indexes and scoring that consider multiple factors could help assess the risk of cardiotoxicity caused by ICIs. Future research needs to focus on finding predictors of severe cardiac toxicity and improving the response to high-dose corticosteroids.

A study conducted by Biagio Barone and his team in 2023 has revealed a correlation between urothelial cancer and cardiovascular disease [152]. The study suggests that there are overlapping pathways in the development of both conditions, meaning that patients with urothelial cancer already have a higher incidence of cardiovascular disease at baseline. This baseline risk is further increased in patients undergoing ICI therapy, who are even more susceptible to cardiotoxic medications. Therefore, caution must be exercised when administering cardiotoxic medication to this subset of patients. Notwithstanding, the potential for ICI-induced cardiac toxicity should not be overlooked, as it can lead to some long term bad outcomes such as the development of serious conditions such as bladder cancer.

Conclusion

This review emphasizes the importance of the early detection, prediction, and management of ICI-induced cardiac toxicity. Factors not linked to the heart or immunotherapy increase the risk of cardiac events in ICI therapy. The parameters should be thoroughly investigated in large multi-ethnic, multicentre prospective cohorts and experimental studies.

Availability of data and materials

All the data generated are available from the corresponding author upon reasonable request.

Abbreviations

ICI:

Immune checkpoint inhibitor

PD1:

Program cell death receptor 1

PDL1:

Program cell death receptor ligand 1

References

  1. Bagchi S, Yuan R, Engleman EG. Immune checkpoint inhibitors for the treatment of cancer: clinical impact and mechanisms of response and resistance. Annu Rev Pathol. 2021;16(1):223–49. https://doi.org/10.1146/annurev-pathol-042020-042741.

    Article  CAS  PubMed  Google Scholar 

  2. Ndjana Lessomo FY, Wang Z, Mukuka C. Comparative cardiotoxicity risk of pembrolizumab versus nivolumab in cancer patients undergoing immune checkpoint inhibitor therapy: A meta-analysis. Front Oncol. 2023 Mar 29;13:1080998. https://doi.org/10.3389/fonc.2023.1080998. PMID: 37064101; PMCID: PMC10090546.

  3. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi T, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small cell lung cancer. N Engl J Med. 2016;375(19):1823–33.

    Article  CAS  PubMed  Google Scholar 

  4. Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immunotherapy and new immunomodulatory targets. Nat Rev Drug Discov. 2015;14(8):561–84. https://doi.org/10.1038/nrd4591.

    Article  CAS  PubMed  Google Scholar 

  5. Weber JS, Kahler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol. 2012;30(21):2691–7. https://doi.org/10.1200/JCO.2012.41.6750.

    Article  CAS  PubMed  Google Scholar 

  6. Lucas JA, Menke J, Rabacal WA, Schoen FJ, Sharpe AH, Kelley VR. Programmed death ligand 1 regulates a critical checkpoint for autoimmune myocarditis and pneumonitis in MRL mice. J Immunol. 2008;181(4):2513–21. https://doi.org/10.4049/jimmunol.181.4.2513.

    Article  CAS  PubMed  Google Scholar 

  7. Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 2001;291(5502):319–22. https://doi.org/10.1126/science.291.5502.319.

    Article  CAS  PubMed  Google Scholar 

  8. Wang J, Okazaki IM, Yoshida T, Chikuma S, Kato Y, Nakaki F, et al. PD-1 deficiency results in the development of fatal myocarditis in MRL mice. Int Immunol. 2010;22(6):443–52. https://doi.org/10.1093/intimm/dxq026.

    Article  CAS  PubMed  Google Scholar 

  9. Okazaki T, Tanaka Y, Nishio R, Mitsuiye T, Mizoguchi A, Wang J, et al. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med. 2003;9(12):1477–83. https://doi.org/10.1038/nm955.

    Article  CAS  PubMed  Google Scholar 

  10. Tajiri K, Aonuma K, Sekine I. Immune checkpoint inhibitor-related myocarditis. Jpn J Clin Oncol. 2018;48(1):7–12. https://doi.org/10.1093/jjco/hyx154.

    Article  PubMed  Google Scholar 

  11. Yun S, Vincelette ND, Mansour I, Hariri D, Motamed S. Late onset ipilimumab-induced pericarditis and pericardial effusion: a rare but life threatening complication. Case Rep Oncol Med. 2015;2015: 794842. https://doi.org/10.1155/2015/794842.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Nirschl CJ, Drake CG. Molecular pathways: coexpression of immune checkpoint molecules: signaling pathways and implications for cancer immunotherapy. Clin Cancer Res. 2013;19(18):4917–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lauer B, Schannwell M, Kühl U, Strauer B-E, Schultheiss H-P. Antimyosin autoantibodies are associated with deterioration of systolic and diastolic left ventricular function in patients with chronic myocarditis. J Am Coll Cardiol. 2000;35(1):11–8.

    Article  CAS  PubMed  Google Scholar 

  15. De Felice F, Musio D, Tombolini V. Immune check-point inhibitors and standard chemoradiotherapy in definitive head and neck cancer treatment. J Pers Med. 2021;11(5):393. https://doi.org/10.3390/jpm11050393.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Hendriks J, Gravestein LA, Tesselaar K, van Lier RA, Schumacher TN, Borst J. CD27 is needed for generation and long-term maintenance of T-cell immunity. Nat Immunol. 2000;1(5):433–40. https://doi.org/10.1038/80877.

    Article  CAS  PubMed  Google Scholar 

  17. Leone RD, Lo YC, Powell JD. A2aR antagonists: Next generation checkpoint blockade for cancer immunotherapy. Comput Struct Biotechnol J. 2015. https://doi.org/10.1016/j.csbj.2015.03.008.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Immune checkpoint inhibitors. National Cancer Institute. 2019. https://www.cancer.gov/about-cancer/treatment/types/immunotherapy/checkpoint-inhibitors Accessed 1 Jun 2023.

  19. A brief history of checkpoint inhibitors. Cancer Today. Cancer Today, Magazine. 2020. https://www.cancertodaymag.org/fall2020/a-brief-history-of-checkpoint-inhibitors/.Accessed 1 Jun 2023.

  20. Hellstrom I, Hellstrom KE, Pierce GE, Yang JP. Cellular and humoral immunity to different types of human neoplasms. Nature. 1968;220:1352–4.

    Article  CAS  PubMed  Google Scholar 

  21. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 2018;8:1069–86.

    Article  PubMed  Google Scholar 

  22. Walker LS, Sansom DM. Confusing signals: recent progress in CTLA-4 biology. Trends Immunol. 2015;36:63–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J. 1992;11:3887–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Jubel JM, Barbati ZR, Burger C, Wirtz DC, Schildberg FA. The role of PD-1 in acute and chronic infection. Front Immunol. 2020;11:487.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sauce D, et al. PD-1 expression on human CD8 T cells depends on both state of differentiation and activation status. AIDS. 2007;21:2005–13.

    Article  CAS  PubMed  Google Scholar 

  26. Kamphorst AO, et al. Proliferation of PD-1+ CD8 T cells in peripheral blood after PD-1-targeted therapy in lung cancer patients. Proc Natl Acad Sci USA. 2017;114:4993–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Borst J, Busselaar J, Bosma DMT, Ossendorp F. Mechanism of action of PD-1 receptor/ligand targeted cancer immunotherapy. Eur J Immunol. 2021;51(8):1911–20. https://doi.org/10.1002/eji.202048994.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hurwitz AA, Foster BA, Kwon ED, Truong T, Choi EM, Greenberg NM, et al. Combination immunotherapy of primary prostate cancer in a transgenic mouse model using CTLA-4 blockade. Cancer Res. 2000;60:2444–8.

    CAS  PubMed  Google Scholar 

  29. Leach DR, Krummel MF, Allison JP. Enhancement of antitumour immunity by CTLA-4 blockade. Science. 1996;271:1734–6. https://doi.org/10.1126/science.271.5256.1734.

    Article  CAS  PubMed  Google Scholar 

  30. van Elsas A, Hurwitz AA, Allison JP. Combination immunotherapy of B16 melanoma using anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and granulocyte/macrophage colony-stimulating factor (GM-CSF)-producing vaccines induces rejection of subcutaneous and metastatic tumors accompanied. J Exp Med. 1999;190:355–66. https://doi.org/10.1084/jem.190.3.355.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 2010;236:219–42. https://doi.org/10.1111/j.1600-065X.2010.00923.x.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Robert C, Ghiringhelli F. What is the role of cytotoxic T lymphocyte-associated antigen 4 blockade in patients with metastatic melanoma? Oncologist. 2009;14(8):848–61. https://doi.org/10.1634/theoncologist.2009-0028.PMID19648604.

    Article  CAS  PubMed  Google Scholar 

  33. Wilkes GM. Margaret Barton-Burke. 2010 oncology nursing drug handbook. Jones & Bartlett Learning. pp. 1. 2009. ISBN 978-0-7637-8124-8. Accessed 30 Mar 2011.

  34. Harivardhan Reddy L. Patrick Couvreur (1 June 2009). Macromolecular Anticancer Therapeutics. Springer. pp. 522. ISBN 978-1-4419-0506-2. Accessed 30 Mar 2011.

  35. Bellaguarda E, Hanauer S. Checkpoint inhibitor-induced colitis. Am J Gastroenterol. 2020;115(2):202–10.

    Article  PubMed  Google Scholar 

  36. Mulcahy N. Two cases of myasthenia gravis seen with ipilimumab, Medscape. 2014. http://www.medscape.com/viewarticle/824347. Accessed: 1 Mar 2023.

  37. ^ Jump up to:a b "Yervoy Annex I: Summary of Product Characteristics" (PDF). Accessed 2 Nov 2014.

  38. Side effects. Yervoy.com. no date. https://www.yervoy.com/side-Effects.

  39. Ipilimumab. National Cancer Institute. 2011. https://www.cancer.gov/about-cancer/treatment/drugs/ipilimumab. Accessed 1 Jun 2023.

  40. Saad P, Kasi A. Ipilimumab. StatPearls Publishing. 2022

  41. Albiges L, Tannir NM, Burotto M, McDermott D, Plimack ER, Barthélémy P, Porta C, Powles T, Donskov F, George S, Kollmannsberger CK, Gurney H, Grimm MO, Tomita Y, Castellano D, Rini BI, Choueiri TK, Saggi SS, McHenry MB, Motzer RJ. Nivolumab plus ipilimumab versus sunitinib for first-line treatment of advanced renal cell carcinoma: extended 4-year follow-up of the phase III CheckMate 214 trial. ESMO Open. 2020;5(6): e001079.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Green SE, McCusker MG, Mehra R. Emerging immune checkpoint inhibitors for treating head and neck cancers. Expert Opin Emerg Drugs. 2020;25(4):501–14.

    Article  CAS  PubMed  Google Scholar 

  43. Zhang S, Bi M. The efficiency and safety of immune checkpoint inhibitors in treating small cell lung cancer: a meta-analysis. Ann Palliat Med. 2020;9(6):4081–8.

    Article  PubMed  Google Scholar 

  44. Wang C, Thudium KB, Han M, Wang XT, Huang H, Feingersh D, Garcia C, Wu Y, Kuhne M, Srinivasan M, Singh S, Wong S, Garner N, Leblanc H, Bunch RT, Blanset D, Selby MJ, Korman AJ. In vitro characterization of the anti-PD-1 antibody nivolumab, BMS-936558, and in vivo toxicology in nonhuman primates. Cancer Immunol Res. 2014;2(9):846–56.

    Article  CAS  PubMed  Google Scholar 

  45. Melisa Puckey B. Pembrolizumab, Drugs.com. no date. https://www.drugs.com/pembrolizumab.html .

  46. Inman BA, et al. Atezolizumab: a PD-L1-blocking antibody for bladder cancer. Clin Cancer Res. 2017;23(8):1886–90.

    Article  CAS  PubMed  Google Scholar 

  47. Deng R, et al. Preclinical pharmacokinetics, pharmacodynamics, tissue distribution, and tumor penetration of anti-PD-L1 monoclonal antibody, an immune checkpoint inhibitor. MAbs. 2016;8(3):593–603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Akinleye A, Rasool Z. Immune checkpoint inhibitors of PD-L1 as cancer therapeutics. J Hematol Oncol. 2019;12(1):92. https://doi.org/10.1186/s13045-019-0779-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hu JR, Florido R, Lipson EJ, Naidoo J, Ardehali R, Tocchetti CG, Lyon AR, Padera RF, Johnson DB, Moslehi J. Cardiovascular toxicities associated with immune checkpoint inhibitors. Cardiovasc Res. 2019;115(5):854–68. https://doi.org/10.1093/cvr/cvz026.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Salem J-E, Manouchehri A, Moey M, Lebrun-Vignes B, Bastarache L, Pariente A, Gobert A, Spano J-P, Balko JM, Bonaca MP, Roden DM, Johnson DB, Moslehi JJ. Cardiovascular toxicities associated with immune checkpoint inhibitors: an observational, retrospective, pharmacovigilance study. Lancet Oncol. 2018;19:1579–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. De Marzo V, Zimarino M. Apical takotsubo syndrome in a patient with metastatic breast carcinoma on novel immunotherapy. Int J Cardiol. 2016;222:760–1. https://doi.org/10.1016/j.ijcard.2016.07.291.

    Article  Google Scholar 

  52. Geisler BP, Raad RA, Esaian D, Sharon E, Schwartz DR. Apical ballooning and cardiomyopathy in a melanoma patient treated with ipilimumab: a case of takotsubo-like syndrome. J ImmunoTher Cancer. 2015;3:4.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Weinstock C, Khozin S, Suzman D, Zhang L, Tang S, Wahby S, Goldberg KB, Kim G, Pazdur RUS. Food and drug administration approval summary: atezolizumab for metastatic non–small cell lung cancer. Clin Cancer Res. 2017;23:4534–9.

    Article  CAS  PubMed  Google Scholar 

  54. Nykl R, Fischer O, Vykoupil K, Taborsky M. A unique reason for coronary spasm causing temporary ST elevation myocardial infarction (inferior STEMI) – systemic inflammatory response syndrome after use of pembrolizumab. Arch Med Sci Atheroscler Dis. 2017;2:100-e102.

    Article  Google Scholar 

  55. Roth ME, Muluneh B, Jensen BC, Madamanchi C, Lee CB. Left ventricular dysfunction after treatment with ipilimumab for metastatic melanoma. Am J Ther. 2016;23:e1925–8.

    Article  PubMed  Google Scholar 

  56. Gibson R, Delaune J, Szady A, Markham M. Suspected autoimmune myocarditis and cardiac conduction abnormalities with nivolumab therapy for non-small cell lung cancer. BMJ Case Rep. 2016. https://doi.org/10.1136/bcr-2016-216228.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Behling J, Kaes J, Münzel T, Grabbe S, Loquai C. New-onset third-degree atrioventricular block because of autoimmune-induced myositis under treatment with anti-programmed cell death-1 (nivolumab) for metastatic melanoma. Melanoma Res. 2017;27:155.

    Article  CAS  PubMed  Google Scholar 

  58. Wei SC, et al. A genetic mouse model recapitulates immune checkpoint inhibitor-associated myocarditis and supports a mechanism-based therapeutic intervention. Cancer Discov. 2020. https://doi.org/10.1158/2159-8290.CD-20-0856.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Johnson DB, Balko JM, Compton ML, Chalkias S, Gorham J, Xu Y, Hicks M, Puzanov I, Alexander MR, Bloomer TL, Becker JR, Slosky DA, Phillips EJ, Pilkinton MA, Craig-Owens L, Kola N, Plautz G, Reshef DS, Deutsch JS, Deering RP, Olenchock BA, Lichtman AH, Roden DM, Seidman CE, Koralnik IJ, Seidman JG, Hoffman RD, Taube JM, Diaz LA Jr, Anders RA, Sosman JA, Moslehi JJ. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. 2016;375(18):1749–55. https://doi.org/10.1056/NEJMoa1609214.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Dubin K, Callahan MK, Ren B, et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun. 2016;7:10391.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Iwama S, De Remigis A, Callahan MK, Slovin SF, Wolchok JD, Caturegli P. Pituitary expression of CTLA-4 mediates hypophysitis secondary to administration of CTLA-4 blocking antibody. Sci Transl Med. 2014;6:23045.

    Article  Google Scholar 

  62. Tarrio ML, Grabie N, Bu DX, Sharpe AH, Lichtman AH. PD-1 protects against inflammation and myocyte damage in T-cell-mediated myocarditis. J Immunol. 2012;188:4876–84.

    Article  CAS  PubMed  Google Scholar 

  63. Nishimura H, Okazaki T, Tanaka Y, et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science. 2001;291:319–22.

    Article  CAS  Google Scholar 

  64. Okazaki T, Tanaka Y, Nishio R, et al. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med. 2003;9:1477–83.

    Article  CAS  PubMed  Google Scholar 

  65. Grabie N, Gotsman I, DaCosta R, et al. Endothelial programmed death-1 ligand 1 (PD-L1) regulates CD8+ T-cell mediated injury in the heart. Circulation. 2007;116:2062–71.

    Article  CAS  PubMed  Google Scholar 

  66. Grabie N, Lichtman AH, Padera R. T-cell checkpoint regulators in the heart. Cardiovasc Res. 2019;115(5):869–77. https://doi.org/10.1093/cvr/cvz025.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. White RL, et al. Cardiopulmonary toxicity of treatment with high dose interleukin-2 in 199 consecutive patients with metastatic melanoma or renal cell carcinoma. Cancer. 1994;74(12):3212–22.

    Article  PubMed  Google Scholar 

  68. Kragel AH, Travis WD, Steis RG, Rosenberg SA, Roberts WC. Myocarditis or acute myocardial infarction associated with interleukin-2 therapy for cancer. Cancer. 1990;66(7):1513–6.

    Article  CAS  PubMed  Google Scholar 

  69. Conlon KC, et al. Redistribution, hyperproliferation, activation of natural killer cells and CD8 T cells, and cytokine production during first-in-human clinical trial of recombinant human interleukin-15 in patients with cancer. JCO. 2015;33(1):74–82. https://doi.org/10.1200/JCO.2014.57.3329.[.

    Article  CAS  Google Scholar 

  70. Crown J, et al. A phase I trial of recombinant human interleukin-1 beta alone and in combination with myelosuppressive doses of 5-fluorouracil in patients with gastrointestinal cancer. Blood. 1991;78(6):1420–7.

    Article  CAS  PubMed  Google Scholar 

  71. Khunger A, Battel L, Wadhawan A, et al. New insights into mechanisms of immune checkpoint inhibitor-induced cardiovascular toxicity. Curr Oncol Rep. 2020;22:65. https://doi.org/10.1007/s11912-020-00925-8.

    Article  PubMed  Google Scholar 

  72. Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, Sharpe AH. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity. 1995;3(5):541–7.

    Article  CAS  PubMed  Google Scholar 

  73. Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, et al. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science. 1995;270(5238):985–8.

    Article  CAS  PubMed  Google Scholar 

  74. Chen C et al. Cardiotoxicity induced by immune checkpoint inhibitors: a pharmacovigilance study from 2014 to 2019 based on FAERS. Front Pharmacol 2021. https://doi.org/10.3389/fphar.2021.616505

  75. Sznol M, Ferrucci PF, Hogg D, et al. Pooled analysis safety profile of nivolumab and ipilimumab combination therapy in patients with advanced melanoma. J Clin Oncol. 2017;35(34):3815–22.

    Article  CAS  PubMed  Google Scholar 

  76. Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377:1345–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Mahmood SS, et al. Myocarditis in patients treated with immune checkpoint inhibitors. J Am Coll Cardiol. 2018;71(16):1755–64. https://doi.org/10.1016/j.jacc.2018.02.037.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, Chau I, Ernstoff MS, Gardner JM, Ginex P, Hallmeyer S, Holter Chakrabarty J, Leighl NB, Mammen JS, McDermott DF, Naing A, Nastoupil LJ, Phillips T, Porter LD, Puzanov I, Reichner CA, Santomasso BD, Seigel C, Spira A, Suarez-Almazor ME, Wang Y, Weber JS, Wolchok JD, Thompson JA. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2018;36:1714–68.

    Article  CAS  PubMed  Google Scholar 

  79. Norwood TG, Westbrook BC, Johnson DB, Litovsky SH, Terry NL, McKee SB, Gertler AS, Moslehi JJ, Conry RM. Smoldering myocarditis following immune checkpoint blockade. J Immunother Cancer. 2017;5:91.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Koelzer VH, Rothschild SI, Zihler D, Wicki A, Willi B, Willi N, Voegeli M, Cathomas G, Zippelius A, Mertz KD. Systemic inflammation in a melanoma patient treated with immune checkpoint inhibitors-an autopsy study. J Immunother Cancer. 2016;4:13.

    Article  PubMed  PubMed Central  Google Scholar 

  81. Spallarossa P, Tini G, Sarocchi M, Arboscello E, Grossi F, Queirolo P, Zoppoli G, Ameri P. Identification and management of immune checkpoint inhibitor-related myocarditis: use troponin wisely. J Clin Oncol. 2019;37:2201–5. https://doi.org/10.1200/JCO.18.02464.

    Article  CAS  PubMed  Google Scholar 

  82. Bonaca Marc P, Olenchock Benjamin A, Salem J-E, Wiviott Stephen D, Ederhy S, Cohen A, Stewart Garrick C, Choueiri Toni K, Di Carli M, Allenbach Y, Kumbhani Dharam J, Heinzerling L, Amiri-Kordestani L, Lyon Alexander R, Thavendiranathan P, Padera R, Lichtman A, Liu Peter P, Johnson Douglas B, Moslehi J. Myocarditis in the setting of cancer therapeutics. Circulation. 2019;140:80–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zarifa A, Kim JW, Lopez-Mattei J, Palaskas N, Iliescu C, Kim PY. Cardiac toxicities associated with immune checkpoints inhibitors: mechanisms manifestations and management. Korean Circ J. 2021;51(7):579–97. https://doi.org/10.4070/kcj.2021.0089.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Chen DY, Huang WK, Chien-Chia WuV, Chang WC, Chen JS, Chuang CK, Chu PH. Cardiovascular toxicity of immune checkpoint inhibitors in cancer patients: a review when cardiology meets immuno-oncology. J Formos Med Assoc. 2020;119(10):1461–75. https://doi.org/10.1016/j.jfma.2019.07.025.

    Article  CAS  PubMed  Google Scholar 

  85. Shaheen S, Mirshahidi H, Nagaraj G, Hsueh CT. Conservative management of nivolumab-induced pericardial effusion: a case report and review of literature. Exp Hematol Oncol. 2018;7:11.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Chu YC, Fang KC, Chen HC, Yeh YC, Tseng CE, Chou TY, et al. Pericardial tamponade caused by a hypersensitivity response to tuberculosis reactivation after anti-PD-1 treatment in a patient with advanced pulmonary adenocarcinoma. J Thorac Oncol. 2017;12:e111–4.

    Article  PubMed  Google Scholar 

  87. Dasanu CA, Jen T, Skulski R. Late-onset pericardial tamponade, bilateral pleural effusions and recurrent immune monoarthritis induced by ipilimumab use for metastatic melanoma. J Oncol Pharm Pract. 2017;23:231–4.

    Article  PubMed  Google Scholar 

  88. Kushnir I, Wolf I. Nivolumab-induced pericardial tamponade: a case report and discussion. Cardiology. 2017;136:49–51.

    Article  CAS  PubMed  Google Scholar 

  89. Gong J, Drobni ZD, Zafar A, et al. Pericardial disease in patients treated with immune checkpoint inhibitors. J Immuno Ther Cancer. 2021;9:e002771. https://doi.org/10.1136/jitc-2021-002771.

    Article  Google Scholar 

  90. Buza V, Rajagopalan B, Curtis AB. Cancer treatment-induced arrhythmias: focus on chemotherapy and targeted therapies. Circ Arrhythm Electrophysiol. 2017;10(8):e005443.

    Article  PubMed  Google Scholar 

  91. Ball S, Ghosh RK, Wongsaengsak S, Bandyopadhyay D, Ghosh GC, Aronow WS, Fonarow GC, Lenihan DJ, Bhatt DL. Cardiovascular toxicities of immune checkpoint inhibitors: JACC review topic of the week. J Am Coll Cardiol. 2019;74(13):1714–27.

    Article  CAS  PubMed  Google Scholar 

  92. Joseph L, De Luna G, Bernit E, Cougoul P, Santin A, Faucher B, Habibi A, Garou A, Loko G, Mattioni S, Manceau S, Arlet JB, Lionnet F. Incidence of cancer treatment induced arrhythmia associated with immune checkpoint inhibitors. J Atr Fibrillation. 2021;13(5):2461. https://doi.org/10.4022/jafib.2461.

    Article  PubMed  PubMed Central  Google Scholar 

  93. Wang F, Wei Q, Wu X. Cardiac arrhythmias associated with immune checkpoint inhibitors: a comprehensive disproportionality analysis of the FDA adverse event reporting system. Front Pharmacol. 2022;13: 986357. https://doi.org/10.3389/fphar.2022.986357.

    Article  CAS  PubMed  Google Scholar 

  94. Zhou YW, Zhu YJ, Wang MN, Xie Y, Chen CY, Zhang T, et al. Immune checkpoint inhibitor-associated cardiotoxicity: current understanding on its mechanism, diagnosis and management. Front Pharmacol. 2019;10:1350. https://doi.org/10.3389/fphar.2019.01350.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Xu Y, Song Y, Liu X, et al. Prediction of major adverse cardiac events is the first critical task in the management of immune checkpoint inhibitor-associated myocarditis. Cancer Commun (Lond). 2022;42(9):902–5.

    Article  PubMed  Google Scholar 

  96. Liu Y, Chen Y, Zeng Z, Liu A. Arrhythmic events associated with immune checkpoint inhibitors therapy: a real-world study based on the food and drug administration adverse event reporting system database. Cancer Med. 2023;12:6637–48. https://doi.org/10.1002/cam4.5438.

    Article  CAS  PubMed  Google Scholar 

  97. Foks AC, Kuiper J. Immune checkpoint proteins: exploring their therapeutic potential to regulate atherosclerosis. Br J Pharmacol. 2017;174:3940–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Bu DX, Tarrio M, Maganto-Garcia E, et al. Impairment of the programmed cell death-1 pathway increases atherosclerotic lesion development and inflammation. Arterioscler Thromb Vasc Biol. 2011;31:1100–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Tomita Y, Sueta D, Kakiuchi Y, et al. Acute coronary syndrome as a possible immune-related adverse event in a lung cancer patient achieving a complete response to anti-PD-1 immune checkpoint antibody. Ann Oncol. 2017;28:2893–5. https://doi.org/10.1093/annonc/mdx326.

    Article  CAS  PubMed  Google Scholar 

  100. Bar J, Markel G, Gottfried T, et al. Acute vascular events as a possibly related adverse event of immunotherapy: a single-institute retrospective study. Eur J Cancer. 2019;120:122–31. https://doi.org/10.1016/j.ejca.2019.06.021.

    Article  CAS  PubMed  Google Scholar 

  101. Maria ATJ, Delmas C, Coustal C, Palassin P, Roubille F. Immune checkpoint inhibitor-associated myocarditis and coronary artery disease: There may be more than meets the eye! Eur J Cancer. 2022;177:194–6. https://doi.org/10.1016/j.ejca.2022.09.028.

    Article  CAS  PubMed  Google Scholar 

  102. Poels K, van Leent MMT, Boutros C, et al. Immune checkpoint inhibitor therapy aggravates T-Cell–Driven plaque inflammation in atherosclerosis. JACC CardioOncol. 2020;2:599–610. https://doi.org/10.1016/j.jaccao.2020.08.007.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Calabretta R, Hoeller C, Pichler V, et al. Immune checkpoint inhibitor therapy induces inflammatory activity in large arteries. Circulation. 2020;142:2396–8. https://doi.org/10.1161/CIRCULATIONAHA.120.048708.

    Article  CAS  PubMed  Google Scholar 

  104. Lamberti G, Gelsomino F, Brocchi S, et al. New disappearance of complicated atheromatous plaques on rechallenge with PD-1/PD-L1 axis blockade in non-small cell lung cancer patient: follow up of an unexpected event. Ther Adv Med Oncol. 2020;12:1758835920913801. https://doi.org/10.1177/1758835920913801.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Schiffer WB, Deych E, Lenihan DJ, et al. Coronary and aortic calcification are associated with cardiovascular events on immune checkpoint inhibitor therapy. Int J Cardiol. 2021;322:177–82. https://doi.org/10.1016/j.ijcard.2020.08.024.

    Article  PubMed  Google Scholar 

  106. Lorenz MW, Markus HS, Bots ML, et al. Prediction of clinical cardiovascular events with carotid intima-media thickness: a systematic review and meta-analysis. Circulation. 2007;115:459–67. https://doi.org/10.1161/CIRCULATIONAHA.106.628875.

    Article  PubMed  Google Scholar 

  107. Oldfield K, Jayasinghe R, Niranjan S, Chadha S. Immune checkpoint inhibitor-induced takotsubo syndrome and diabetic ketoacidosis: rare reactions. BMJ Case Rep. 2021;14(2): e237217. https://doi.org/10.1136/bcr-2020-237217.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Serzan M, Rapisuwon S, Krishnan J, Chang IC, Barac A. Takotsubo cardiomyopathy associated with checkpoint inhibitor therapy: endomyocardial biopsy provides pathological insights to dual diseases. JACC CardioOncol. 2021;3(2):330–4. https://doi.org/10.1016/j.jaccao.2021.02.005.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Wilson HM, Cheyne L, Brown PA. Characterization of the myocardial inflammatory response in acute stress-induced (Takotsubo) cardiomyopathy. J Am Coll Cardiol Basic Trans Science. 2018;3:766–78.

    Google Scholar 

  110. Minegishi S, Kinguchi S, Horita N, Namkoong H, Briasoulis A, Ishigami T, Tamura K, Nishiyama A, Yano Y. Japanese Society of Hypertension working group “Onco-Hypertension”. Immune checkpoint inhibitors do not increase short-term risk of hypertension in cancer patients: a systematic literature review and meta-analysis. Hypertension. 2022;79(11):2611–2621.

  111. Glick M, Baxter C, Lopez D, Mufti K, Sawada S, Lahm T. Releasing the brakes: a case report of pulmonary arterial hypertension induced by immune checkpoint inhibitor therapy. Pulm Circ. 2020;10(4):2045894020960967. https://doi.org/10.1177/2045894020960967.

    Article  PubMed  PubMed Central  Google Scholar 

  112. Brumberger ZL, Branch ME, Klein MW, Seals A, Shapiro MD, Vasu S. Cardiotoxicity risk factors with immune checkpoint inhibitors. Cardiooncology. 2022;8(1):3. https://doi.org/10.1186/s40959-022-00130-5.

    Article  PubMed  PubMed Central  Google Scholar 

  113. Vyas DA, Eisenstein LG, Jones DS. Hidden in Plain Sight—Reconsidering the Use of Race Correction in Clinical Algorithms. N Engl J Med. 2020;383(9):874–882 and Cerdeña JP, Plaisime MV, Tsai J. From race-based to race-conscious medicine: how anti-racist uprisings call us to act. Lancet. 2020; 396(10257): 125–1128.

  114. Tong M, Artiga S. Use of race in clinical diagnosis and decision making: overview and implications, KFF. 2021. https://www.kff.org/racial-equity-and-health-policy/issue-brief/use-of-race-in-clinical-diagnosis-and-decision-making-overview-and-implications/. Accessed 1 Jun 2023.

  115. Torrente M, Blanco M, Franco F, Garitaonaindia Y, Calvo V, Collazo-Lorduy A, Gutiérrez L, Sánchez JC, González-del-Alba A, Hernández R, Méndez M, Cantos B, Núñez B, Sousa PAC, Provencio M. Assessing the risk of cardiovascular events in patients receiving immune checkpoint inhibitors. Front Cardiovasc Med. 2022;9:1062858. https://doi.org/10.3389/fcvm.2022.1062858.

    Article  PubMed  Google Scholar 

  116. Tan YY, Gast GC, van der Schouw YT. Gender differences in risk factors for coronary heart disease. Maturitas. 2010;65(2):149–60. https://doi.org/10.1016/j.maturitas.2009.09.023.

    Article  PubMed  Google Scholar 

  117. de Groot PM, Wu CC, Carter BW, Munden RF. The epidemiology of lung cancer. Transl Lung Cancer Res. 2018;7(3):220–33. https://doi.org/10.21037/tlcr.2018.05.06.

    Article  CAS  PubMed Central  Google Scholar 

  118. Rodgers JL, Jones J, Bolleddu SI, Vanthenapalli S, Rodgers LE, Shah K, Karia K, Panguluri SK. Cardiovascular Risks Associated with Gender and Aging. J Cardiovasc Dev Dis. 2019;6(2):19. https://doi.org/10.3390/jcdd6020019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Tranvåg EJ, Norheim OF, Ottersen T. Clinical decision making in cancer care: a review of current and future roles of patient age. BMC Cancer. 2018;18:546. https://doi.org/10.1186/s12885-018-4456-9.

    Article  PubMed  PubMed Central  Google Scholar 

  120. Wang J, Wang FW. Impact of age on clinical presentation, treatment, and cancer-specific survival of patients with small-cell carcinoma of the prostate. Clin Interv Aging. 2013;8:871–7. https://doi.org/10.2147/CIA.S44772.

    Article  PubMed  Google Scholar 

  121. Weida EB, Phojanakong P, Patel F, Chilton M. Financial health as a measurable social determinant of health. PLoS ONE. 2020;15(5): e0233359. https://doi.org/10.1371/journal.pone.0233359.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Qiu F, Liang CL, Liu H, Zeng YQ, Hou S, Huang S, Lai X, Dai Z. Impacts of cigarette smoking on immune responsiveness: Up and down or upside down? Oncotarget. 2017;8:268–84. https://doi.org/10.18632/oncotarget.13613.

    Article  PubMed  Google Scholar 

  123. Arimilli S, Schmidt E, Damratoski BE, Prasad GL. Role of oxidative stress in the suppression of immune responses in peripheral blood mononuclear cells exposed to combustible tobacco product preparation. Inflammation. 2017;40:1622–30. https://doi.org/10.1007/s10753-017-0602-9.

    Article  CAS  PubMed  Google Scholar 

  124. Corke LK, Li JJN, Leighl NB, Eng L. Tobacco use and response to immune checkpoint inhibitor therapy in non-small cell lung cancer. Curr Oncol. 2022;29(9):6260–76. https://doi.org/10.3390/curroncol29090492.

    Article  PubMed Central  Google Scholar 

  125. Cortellini A, D’Alessio A, Cleary S, et al. Type 2 diabetes mellitus and efficacy outcomes from immune checkpoint blockade in patients with cancer. Clin Cancer Res. 2023. https://doi.org/10.1158/1078-0432.CCR-22-3116.

    Article  PubMed  Google Scholar 

  126. Youssef N, Noureldein M, Daoud G, Eid AA. Immune checkpoint inhibitors and diabetes: Mechanisms and predictors. Diabetes Metab. 2021;47(3): 101193. https://doi.org/10.1016/j.diabet.2020.09.003.

    Article  CAS  PubMed  Google Scholar 

  127. Venetsanaki V, Boutis A, Chrisoulidou A, Papakotoulas P. Diabetes mellitus secondary to treatment with immune checkpoint inhibitors. Curr Oncol. 2019;26(1):e111–4. https://doi.org/10.3747/co.26.4151.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Ross KH, Gogineni K, Subhedar PD, Lin JY, McCullough LE. Obesity and cancer treatment efficacy: Existing challenges and opportunities. Cancer. 2019;125:1588–92. https://doi.org/10.1002/cncr.31976.

    Article  PubMed  Google Scholar 

  129. Mojibi Y, Seif F, Mojibi N, Aghamajidi A, Mohsenzadegan M, Torang H-A. Efficacy of immunotherapy in obese patients with cancer. Immunopharmacol Immunotoxicol. 2022;44(4):471–83. https://doi.org/10.1080/08923973.2022.2061989.

    Article  CAS  PubMed  Google Scholar 

  130. Wang Z, Aguilar EG, Luna JI, Dunai C, Khuat LT, Le CT, Mirsoian A, Minnar CM, Stoffel KM, Sturgill IR, et al. Paradoxical effects of obesity on T-cell function during tumor progression and PD-1 checkpoint blockade. Nat Med. 2019;25:141–51. https://doi.org/10.1038/s41591-018-0221-5.[.

    Article  CAS  PubMed  Google Scholar 

  131. Mirsoian A, Bouchlaka MN, Sckisel GD, Chen M, Pai CC, Maverakis E, Spencer RG, Fishbein KW, Siddiqui S, Monjazeb AM, et al. Adiposity induces lethal cytokine storm after systemic administration of stimulatory immunotherapy regimens in aged mice. J Exp Med. 2014;211:2373–83. https://doi.org/10.1084/jem.20140116.

    Article  PubMed  PubMed Central  Google Scholar 

  132. Woodall MJ, Neumann S, Campbell K, Pattison ST, Young SL. The effects of obesity on anti-cancer immunity and cancer immunotherapy. Cancers (Basel). 2020;12(5):1230. https://doi.org/10.3390/cancers12051230.

    Article  CAS  PubMed  Google Scholar 

  133. Bao Z, Luo L, Xu T, Yang J, Lv M, Ni L, Sun X, Chen W, Zhou L, Wang X, Xiang Y, Gao B. Risk factors and prognostic role of renal adverse event in patients receiving immune checkpoint inhibitor therapy: analysis of data from a retrospective cohort study. Ann Transl Med. 2022;10(18):967. https://doi.org/10.21037/atm-22-3684.

    Article  CAS  PubMed  Google Scholar 

  134. https://www.nejm.org/doi/full/10.1056/NEJMoa041365

  135. Yang X, Gao M, Xu R, Tao Y, Luo W, Wang B, Zhong W, He L, He Y. Hyperthermia combined with immune checkpoint inhibitor therapy in the treatment of primary and metastatic tumors. Front Immunol. 2022;13: 969447. https://doi.org/10.3389/fimmu.2022.969447.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Li Z, Deng J, Sun J, Ma Y. Hyperthermia targeting the tumor microenvironment facilitates immune checkpoint inhibitors. Front Immunol. 2020;9(11): 595207. https://doi.org/10.3389/fimmu.2020.595207.

    Article  CAS  Google Scholar 

  137. Irabor OC, Nelson N, Shah Y, Niazi MK, Poiset S, Storozynsky E, Singla DK, Hooper DC, Lu B. Overcoming the cardiac toxicities of cancer therapy immune checkpoint inhibitors. Front Oncol. 2022;12: 940127. https://doi.org/10.3389/fonc.2022.940127.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Kucuk A, Topkan E, Selek U, Haksoyler V, Mertsoylu H, Besen AA, Pehlivan B. High measures of pre-chemoradiotherapy platelet-to-albumin ratio indicates poor prognosis in locally advanced pancreatic cancer patients. Ther Clin Risk Manag. 2022;18:421–8. https://doi.org/10.2147/TCRM.S359553.

    Article  PubMed  PubMed Central  Google Scholar 

  139. Stotz M, Gerger A, Eisner A, et al. Increased neutrophil-lymphocyte ratio is a poor prognostic factor in patients with primary operable and inoperable pancreatic cancer. Br J Cancer. 2013;109:416–21. https://doi.org/10.1038/bjc.2013.332.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Shirai Y, Shiba H, Sakamoto T, et al. Preoperative platelet to lymphocyte ratio predicts outcome of patients with pancreatic ductal adenocarcinoma after pancreatic resection. Surgery. 2015;158(2):360–5. https://doi.org/10.1016/j.surg.2015.03.043.

    Article  PubMed  Google Scholar 

  141. Ahmad J, Grimes N, Farid S, Morris-Stiff G. Inflammatory response related scoring systems in assessing the prognosis of patients with pancreatic ductal adenocarcinoma: a systematic review. Hepatobiliary Pancreat Dis Int. 2014;13:474–81. https://doi.org/10.1016/s1499-3872(14)60284-8.

    Article  PubMed  Google Scholar 

  142. Subudhi SK, et al. Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc Natl Acad Sci USA. 2016;113:11919–24.

    Article  CAS  PubMed  Google Scholar 

  143. Toi Y, et al. Profiling preexisting antibodies in patients treated with anti-PD-1 therapy for advanced non-small cell lung cancer. JAMA Oncol. 2019;5:376–83.

    Article  PubMed  Google Scholar 

  144. de Moel EC, et al. Autoantibody development under treatment with immune-checkpoint inhibitors. Cancer Immunol Res. 2019;7:6–11.

    Article  PubMed  Google Scholar 

  145. Hinrichs L, Mrotzek SM, Mincu RI, Pohl J, Röll A, Michel L, Mahabadi AA, Al-Rashid F, Totzeck M, Rassaf T. Troponins and natriuretic peptides in cardio-oncology patients-data from the ECoR registry. Front Pharmacol. 2020;19(11):740. https://doi.org/10.3389/fphar.2020.00740.

    Article  CAS  Google Scholar 

  146. Pavo N, Raderer M, Hülsmann M, et al. Cardiovascular biomarkers in patients with cancer and their association with all-cause mortality. Heart. 2015;101(23):1874–80. https://doi.org/10.1136/heartjnl-2015-307848.

    Article  CAS  PubMed  Google Scholar 

  147. Cardinale D, Sandri MT, Colombo A, et al. Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing high-dose chemotherapy. Circulation. 2004;109(22):2749–54. https://doi.org/10.1161/01.CIR.0000130926.51766.CC.

    Article  CAS  PubMed  Google Scholar 

  148. Vokes NI, Pan K, Le X. Efficacy of immunotherapy in oncogene-driven non-small cell lung cancer. Ther Adv Med Oncol. 2023. https://doi.org/10.1177/17588359231161409.

    Article  PubMed Central  Google Scholar 

  149. Joolharzadeh P, Rodriguez M, Zaghlol R, et al. Recent advances in serum biomarkers for risk stratification and patient management in cardio-oncology. Curr Cardiol Rep. 2023;25:133–46. https://doi.org/10.1007/s11886-022-01834-x.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Crocetto F, Ferro M, Buonerba C, Bardi L, Dolce P, Scafuri L, Mirto BF, Verde A, Sciarra A, Barone B, Calogero A, Sagnelli C, Busetto GM, Del Giudice F, Cilio S, Sonpavde G, Di Trolio R, Della Ratta GL, Barbato G, Di Lorenzo G. Comparing cardiovascular adverse events in cancer patients: a meta-analysis of combination therapy with angiogenesis inhibitors and immune checkpoint inhibitors versus angiogenesis inhibitors alone. Crit Rev Oncol Hematol. 2023;188: 104059. https://doi.org/10.1016/j.critrevonc.2023.104059.

    Article  PubMed  Google Scholar 

  151. Liu M, Cheng X, Ni R, Zheng B, Huang S, Yang J. Cardiotoxicity of immune checkpoint inhibitors: a frequency network meta-analysis. Front Immunol. 2022;13:1006860. https://doi.org/10.3389/fimmu.2022.1006860.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Barone B, Finati M, Cinelli F, Fanelli A, Del Giudice F, De Berardinis E, Sciarra A, Russo G, Mancini V, D’Altilia N, Ferro M, Porreca A, Chung BI, Basran S, Bettocchi C, Cormio L, Imbimbo C, Carrieri G, Crocetto F, Busetto GM. Bladder Cancer and risk factors: data from a multi-institutional long-term analysis on cardiovascular disease and cancer incidence. J Pers Med. 2023;13(3):512. https://doi.org/10.3390/jpm13030512.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

ZW supervised the work, FYNL drafted the manuscript, and OMO and CM proofread the paper. All the authors agreed to the submission of this final draft.

Corresponding authors

Correspondence to Fabrice Yves Ndjana lessomo or Zhi-Quan Wang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The Author confirms: that this review has not been published before, nor is it under consideration for publication elsewhere; that all co-authors have approved its publication.

Competing interests

None declared.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

lessomo, F.Y.N., Mandizadza, O.O., Mukuka, C. et al. A comprehensive review on immune checkpoint inhibitors induced cardiotoxicity characteristics and associated factors. Eur J Med Res 28, 495 (2023). https://doi.org/10.1186/s40001-023-01464-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40001-023-01464-1

Keywords