Next Article in Journal
Efficacy of an Anticoagulation Clinic in Low-Income Brazilian Patients with Heart Disease: A Randomized Clinical Trial
Next Article in Special Issue
The Role of Allogeneic Transplantation in Chronic Myeloid Leukemia in 2023: A Case-Based Concise Review
Previous Article in Journal
Mediastinal Gray-Zone Lymphoma: Still an Open Issue
Previous Article in Special Issue
Cardiotoxicity of Tyrosine Kinase Inhibitors in Philadelphia-Positive Leukemia Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Direct and Indirect Effects of Tyrosine Kinase Inhibitors on the Cardiovascular System in Chronic Myeloid Leukemia

1
Hematology Unit, Businco Hospital, Department of Medical Sciences and Public Health, University of Cagliari, 09121 Cagliari, Italy
2
Department of Cardiac, Thoracic, Vascular and Public Health, University of Padua, 35100 Padua, Italy
*
Author to whom correspondence should be addressed.
Hemato 2023, 4(3), 207-226; https://doi.org/10.3390/hemato4030017
Submission received: 15 May 2023 / Revised: 3 July 2023 / Accepted: 13 July 2023 / Published: 14 July 2023

Abstract

:
Since their introduction, tyrosine kinase inhibitors (TKIs) have radically changed the treatment paradigm of Chronic Myeloid Leukemia (CML), leading to deep and lasting molecular responses and profoundly influencing survival. However, cancer-therapy-related Cardiovascular Toxicities (CTR-CVTs) associated with BCR::ABL1 TKIs are one of the main sources of concern: hypertension, arterial occlusive events, arrhythmias, dysmetabolic alteration, and glomerular filtration impairment are frequently reported in clinical trials and real-life experiences. Therefore, a close interaction between hematologists and cardiologists becomes crucial to implementing prevention protocols based on a comprehensive assessment of baseline cardiovascular risk, the management of any detectable and modifiable risk factors, and the elaboration of a monitoring plan for CTR-CVTs during treatment. Here, we provide the most comprehensive and recent evidence in the literature on the pathophysiological patterns underlying CTR-CVTs, providing useful evidence-based guidance on the prevention and management of CVD risk factors at baseline and during treatment with BCR::ABL1 TKIs.

1. Introduction

The advent of target therapy with tyrosine kinase inhibitors (TKIs) has completely revolutionized the Chronic Myeloid Leukemia (CML) treatment paradigm, leading to a survival equal to that of the general population and causing patients to die mainly from causes unrelated to CML [1,2]. Despite their well-recognized efficacy and excellent long-term survival outcomes in responsive patients, cancer-therapy-related Cardiovascular Toxicities (CTR-CVTs) associated with BCR::ABL1 TKIs are still a source of concern. Endothelial damage, the promotion of atherogenesis, metabolic dysfunctions, and glomerular function impairment are the most frequently reported rationales in the literature.
Particularly, the second- and third-generation TKIs (2G/3G-TKIs), nilotinib and ponatinib, have significantly increased the reporting for myocardial infarction (MI), stroke, peripheral arterial occlusive disease (PAOD), and venous thromboembolic events (VTEs) [3,4,5,6,7]. Similar to other Vascular Endothelial Growth Factor Receptor (VEGFR) inhibitors, ponatinib can cause new-onset arterial hypertension [8], while dasatinib can determine pulmonary arterial hypertension (PAH) [9]. Among new agents, asciminib [10] and vodobatinib [11,12] have shown a good safety profile in clinical trials, while olverembatinib, a third-generation TKI drawn on the ponatinib scaffold, induced arterial hypertension, pericardial effusion, and ventricular extrasystoles in 32.1% of the patients included in phase I/II trials [13]. In this scenario, we identify two major issues: (1) CTR-CVTs may require the premature discontinuation of TKIs, resulting in worsening outcomes; and (2) the burden of cardiovascular comorbidity may be challenging to manage, requiring great experience, active surveillance, and patient adherence. In order to mitigate the cardiovasc1ular risk at baseline and once therapy is started, the interaction between hematologists and cardiologists becomes crucial. For this purpose, the first Guidelines on Cardio-Oncology were published in 2022 by the European Society of Cardiology (ESC), providing an individualized approach to care based on basal cardiovascular risk assessment and new surveillance protocols during cancer treatment [14]. Moreover, the Cardio-Oncology Study Group of the Heart Failure Association (HFA) of the ESC, in collaboration with the International Cardio-Oncology Society (ICOS), proposed a new scoring tool that can be used specifically to stratify the cardiovascular risk in cancer patients before starting potential cardiotoxic cancer therapies [15].
The aim of this review is to present the most important evidence regarding the underlying mechanism of CTR-CVTs and the cardiovascular safety profiles of BCR::ABL1 TKIs. Accordingly, we try to highlight the preventive management strategies for cardiovascular assessment and risk factor modification before, during, and after TKI treatment.

2. Pathogenetic Overview of TKIs-Induced Cardiovascular Events

Through reversible phosphorylation, kinases regulate many signaling pathways that control metabolism, cell cycle progression, cell differentiation, proliferation, and death [16]. All TKIs approved for CML share the ability to inhibit BCR::ABL1, but have also shown heterogeneous off-target toxicities on kinases involved in several physiological processes, such as VEGFR 1 to 3, TIE-3, and platelet-derived growth factor receptors (FGFR) 1 to 4 [17,18,19,20].
The off-target toxicities related to TKI treatment and their rationale are summarized in Figure 1.

2.1. Atherogenesis and Occlusive Events

Common hypotheses for adverse occlusive events (AOEs) include pro-atherogenic, anti-angiogenic, and metabolic effects. Initial studies focused on platelet function, driven mainly by the prevalence of arterial vascular events. Different mechanisms seem to be involved at different times of the onset of AOEs: vasospasm [21] or acute thrombotic events can underlie early adverse events, while later events may reflect a chronic inflammatory or pro-thrombotic state [22]. While it is accepted that imatinib, dasatinib, and bosutinib do not influence platelet function towards a prothrombotic state, less clear is the role of nilotinib. In some evidence, nilotinib has not been shown to significantly alter platelet activity [23,24], while others have reported it having an influence on platelet secretion, activation, and adhesion [25]. Indeed, nilotinib has enhanced thrombus growth and stability in human and murine models via ex vivo adhesion on type I collagen under arterial flow and increased in vivo thrombus growth in mouse models [25]. This capacity has been reported recently also for ponatinib [22,25]. The latter has also increased serum P-selectin secretion, known as a marker of platelet and endothelial activation, and other inflammatory markers, including TNF-a, IFN-g, and IL-6 [22].
Other studies have focused on TKIs’ influence on the endothelium and atherogenesis. It has been reported that nilotinib induces the expression of pro-atherogenic molecules such as ICAM-1, VCAM1, and E-selectin on endothelial cells (ECs), chemokines such as CCL2, CXCL8, and CXCL2, and pro-apoptotic markers such as caspase-3 and -7 [6,26]. In addition, IL-1b, typically associated with inflammation, atherosclerosis, and type 2 diabetes (T2D), seems to be increased in patients treated with nilotinib [27,28]. Interestingly, a higher frequency of mutation in TET2, ASXL1, and DNMT3A has been noted in CML patients who developed AOEs. Indeed, studies on clonal hematopoiesis have shown an increased risk of cardiovascular disease (CVD) and a greater frequency with age, thus supporting the influence of aging in increasing basal cardiovascular risk and suggesting possible clinical implications in the future [26,29]. Conversely, less is known about the third-generation TKI, ponatinib. Still, it is likely that, like nilotinib, the drug causes the switch from an antithrombotic phenotype to a pro-thrombotic one in ECs. Ponatinib increases the expression of VCAM1 [30], activates the TLR4/NLRP3/IL1-b pathway in cardiac and systemic myeloid cells both in vitro and in vivo [31], and inhibits VEGFR. These abilities may interfere with angiogenesis and increase systemic arterial pressure (BP) [32]. Furthermore, a metabolomic analysis has shown a peculiar metabolic profile in patients treated with nilotinib and ponatinib, such as a higher serum tyrosine level, which is well known to be a marker of systemic inflammation and the impaired functionality of vessel walls [33]. Altogether, these data seem to suggest that nilotinib- and ponatinib-induced atherogenesis and occlusive events depend on their effect on the viability of ECs and on interference with the defensive process from recanalizing and vessel repair once stenosis is established [26,30,31].

2.2. Hypertension

The off-target effect of ponatinib and nilotinib includes the inhibition of VEGFR, a tyrosine kinase receptor predominantly expressed on the surface of ECs, which normally regulates endothelial proliferation and survival, improves vascular permeability, and drives angiogenesis. The inhibition of VEGFR is associated with a reduction in nitric oxide (NO) production, a potent vasodilator, and an increase in oxidative stress and endothelin 1, which are associated with inflammation and vasoconstriction [26,32]. Therefore, VEGFR inhibition seems to justify the increased incidence of hypertension in these patients, probably by altering the NO pathway, increasing the production of endothelin-1 and/or inducing microvascular rarefaction [34]. Indeed, NO also participates in tubule-glomerular feedback and sodium metabolism, so its inhibition may contribute to hypertension through sodium retention and another direct renal mechanism. Finally, it is likely that the inflammatory state associated with ponatinib may exacerbate a pre-existing hypertensive condition; according to recent evidence, this could be the result of the polarization of M1 macrophages induced by an alteration in circulating substrates, such as glucose or lipid, lipotoxicity, and tissue hypoxia [35].

2.3. Arrhythmias

Nilotinib and dasatinib have also been found to be potent inhibitors of the human ether-à-go-go-related gene (hERG), also known as KCNH2, which encodes for the pore-forming subunit of the channel-conduction IKr, which is critical for the repolarization of human ventricles [36]. By blocking the efflux of potassium ions, these drugs lengthen the amount of time necessary to regenerate the cardiac action potential, causing changes in the post-contraction refractory period and leading to life-threatening ventricular arrhythmias [37]. Nilotinib has been found to exert inhibitory effects on hERG with an IC50 value of 0.13 µM, indicating its ability to prolong the QT interval and potentially induce atrial fibrillation. Furthermore, chronic exposure to nilotinib seems to downregulate the amount of hERG on the cell membrane of human-induced pluripotent stem-cell-derived cardiomyocytes. In the same study, dasatinib demonstrated activity on the same target with an IC50 of 14.3 µM, but appeared to rarely cause QT interval prolongation events [37].

2.4. Pulmonary Hypertension

Pulmonary hypertension (PH), defined by a mean pulmonary arterial pressure of >20 mmHg at rest [38], is a rare and life-threatening side effect of dasatinib [9]. The exact mechanism is still not entirely clear, but increased vascular resistance in the pulmonary circulation appears to have a multifactorial genesis, including vasoconstriction, the remodeling of the lung vessel wall, and thrombosis [39]. More specifically, some evidence has shown that the impact of dasatinib on ECs is mainly due to an increased production of mitochondrial ROS, and through attenuated hypoxic pulmonary vasoconstriction responses [40].

2.5. Metabolic Effects

Several studies have focused on the metabolic effects induced by TKIs, reporting contrasting results between TKIs. In fact, imatinib has been shown to improve in vitro and in vivo glucose metabolism [41,42,43] through the internal translocation of glucose transporters (GLUT1, GLUT3, and GLUT6), by reducing glucose intake, switching from glycolysis to the tricarboxylic acid cycle, and increasing insulin secretion by limiting pancreatic b-cell apoptosis [44]. In addition, adiponectin, a hormone secreted by adipocytes with an important effect on insulin sensitivity regulation, has been found to increase in patients treated with imatinib. In this context, imatinib seems to promote the adipogenic differentiation of mesenchymal stromal cells, probably by the inhibition of PDGFRa and PDGFRb, thus interfering in the normal signaling regulation of macrophages on pre-adipocytes and stimulating adipogenesis [45]. With regard to dasatinib, some studies have reported improved plasma glucose levels, probably through its direct effect on c-Abl [46,47,48,49] or a senolytic action on pancreatic islets [50,51,52], while other authors have reported increased glucose levels in patients treated with dasatinib, possibly related to the inhibition of c-KIT, which has proved to be essential for the survival of b-cells in mice [53].
Numerous pieces of evidence support the ability of nilotinib to increase the plasma glucose levels through an insulin resistance (IR) status [54,55,56]. This condition, characterized by a reduced glucose intake in the skeletal and cardiac muscle, adipose tissue, and liver, has been associated with a low-grade chronic inflammatory state and the production of pro-inflammatory cytokines, including IL-6, IL-8, and TNF-a, thus contributing to EC dysfunction and the acceleration of atherosclerosis [57]. The downregulation of Low-density Lipoprotein Receptor (LDLR), Very-Low-Density Lipoprotein (VLDLR), and Insulin Receptor Substrate 1 (IRS1) and a progressive increase in circulating oxidized LDL levels have also been associated with nilotinib [6,58]. Inhibited lipid internalization by adipose tissue contributes to the IR by increasing free fatty acids (FFA), which are known to contribute to atherogenesis via macrophage activation, foam cell formation, smooth muscle cell proliferation, and angiogenesis [6].
Ponatinib, on the other hand, has been shown to increase in vitro levels of desmosterol, as well as other substrates of 24-dehydro-cholesterol reductase (DHCR24), which is responsible for converting desmosterol into cholesterol [59]. Desmosterol appears to possess a physiological role in vivo by modulating cholesterol metabolism and the inflammatory responses in foam cells, and its depletion in myeloid cells via the overexpression of DHCR24 seems to favor the progression of atherosclerosis by attenuating macrophage anti-inflammatory mediators [60,61,62]. In addition, a study conducted on APOE*3Leiden.CEPT transgenic mice reported reduced plasma levels of VLDL-LDL and cholesterol in the liver [63].

2.6. Renal Impairment

Renal dysfunction associated with TKIs, especially imatinib and bosutinib [64,65], can further increase cardiovascular risk [66]. Pieces of evidence in the literature have reported this association and several hypotheses have been advanced.
Renal injury may result from a tumor lysis syndrome [67], while vomiting and diarrhea, commonly known side effects of bosutinib and imatinib, may all contribute to the hypovolemic state that could induce renal dysfunction [68]. Direct toxic tubular damage can depend on imatinib’s ability to inhibit the PDGFR, which usually regulates the proliferation and regeneration of tubular cells [66]. Increased serum creatinine may also depend on the inhibitory action of imatinib and bosutinib toward the Organic Cation Transporter 2 (OCT2), encoded by the SCL22A2 gene [69,70]. In this regard, a 20% increase in serum creatinine was observed with a median serum bosutinib concentration of 60–70 ng/mL, with a significant increase, especially in patients harboring the 808G/G polymorphism of SCL22A2 [70].
Cases of acute kidney injury (AKI) have been reported in patients treated with dasatinib. Ozkurt et al. reported a case of gastroenteritis and acute kidney injury in a patient with imatinib-resistant CML, which was resolved after the discontinuation of dasatinib [71]. Additionally, case reports have described the development of nephrotic syndrome in adult and pediatric CML patients. Dasatinib, being a multi-kinase inhibitor, may potentially damage podocyte and endothelial cells through the direct inhibition of SRC kinases, with indirect effects on VEGF [72].
Nevertheless, these mechanisms remain unclear, and other factors, alone or in combination, may contribute to kidney damage. Further investigations are needed to clarify the mechanisms underlying renal impairment during imatinib treatment and to identify potential predictive factors.

2.7. Platelet Dysfunction

Ischemic heart disease and stroke are one of the leading causes of mortality globally. The underlying cause of these conditions is often thrombotic complications resulting from the rupture or erosion of atherosclerotic plaques. Antiplatelet therapy (e.g., aspirin and clopidogrel) is the mainstay of the current treatment for arterial thrombosis, but it has limitations, such as irreversible action and increased bleeding risk [14].
Platelet dysfunction has been reported in 40% of patients treated with dasatinib, with 10% of them experiencing grade ≥3 dysfunction. Specifically, the drug appears to impair platelet aggregation, without being associated with bleeding diathesis in treated patients. Quintas-Cardama et al. demonstrated that dasatinib did not interfere with secondary hemostasis, but rather affected platelet aggregation, similar to aspirin. The authors examined this platelet aggregation in 87 patients with CP-CML, including 27 patients treated with dasatinib, 32 with bosutinib, 19 with imatinib, and 9 with nilotinib. Among the patients treated with dasatinib, impaired platelet aggregation upon stimulation with arachidonic acid, epinephrine, or both was observed in 70%, 85%, and 59% of them, respectively [73]. A second study evaluating the correlation between TKI-induced platelet dysfunction and bleeding diathesis in CML patients found no correlation between bleeding presence, bleeding scores, and platelet dysfunction [74].
Dasatinib interferes with platelet aggregation, which is important because antiplatelet agents such as aspirin or clopidogrel are commonly used to prevent thrombotic cardiovascular events. While caution is advised when combining anticoagulant and antiplatelet therapy with dasatinib, especially in elderly patients or those prone to bleeding, the overall risk of bleeding without these factors is likely low [75].

3. Cardiovascular Toxicity Risk Assessment and Stratification

In the “TKIs era”, the need to evaluate the cardiovascular risk, both in the initial treatment decision and in the long-term management of CML, has become established. The identification and management of risk factors are invaluable for recognizing the patients most likely to develop CTR-CVTs and implementing strategies to mitigate or reverse the risk at baseline. For this purpose, the first Guidelines on Cardio-Oncology were published in 2022 by the European Society of Cardiology (ESC), providing a personalized approach to care based on basal cardiovascular toxicity risk assessment and new surveillance protocols during cancer treatment [14].
The absolute risk for CTR-CVTs is correlated with cardiovascular risk at baseline and changes over time during treatment. Therefore, diagnosis and before starting TKIs are the optimal timepoints for the elaboration of preventive strategies. History assessment, physical examination, blood pressure (BP) measurement, and glucose and lipids dosing are essential to stratifying CML patients within a specific risk category and achieving a considered therapeutic decision. Other cardiac tests (i.e., ECG, biomarkers, and imaging) should be considered individually based on the cardiovascular risk and planned treatment strategy (Figure 2). Especially with nilotinib or ponatinib, cardiovascular risk assessment is recommended every 3 months during the first year and every 6–12 months thereafter [14].
In recent years, the most widely used CV risk assessment system in the general population is the Systemic Coronary Risk Estimation (SCORE) algorithm, which evaluates the 10-year risk of death for CVD based on sex, age, systolic pressure, smoking, and total cholesterol levels [76]. In CML, the SCORE algorithm was applied retrospectively in 85 patients treated with ponatinib, where patients with a high to very high SCORE risk showed significantly higher rates of AOEs than those with lower risk classes (74.3% vs. 15.2%, p < 0.001) [77]. The same algorithm, when applied to a cohort of 369 patients treated with nilotinib, showed a significantly higher incidence of AOEs (34.4 ± 6% vs. 10 ± 2.1%, p < 0.001) in patients in a high and very high SCORE risk class (SCORE > 5%) [78]. Overall, these data appear to be in line with those reported in a recent meta-analysis, where high and very high SCORE risks were correlated with a higher risk for AOEs in CML patients treated with nilotinib (HR 3.5 IV 95% 1.4–8.7 and HR 4.4 IC 95% 2–9.8, respectively) [6].
In 2021, the SCORE algorithm was updated in the SCORE2 for apparently healthy people of 40–69, with the inclusion of fatal and non-fatal events (e.g., myocardial infarction and stroke), and SCORE2-Older Patients (SCORE2-OP) for those aged >70 years [14]; compared to the previous one, the updated algorithm (1) has been validated on much larger derivation and validation cohorts, (2) provides risk estimates for the combined outcome of 10 years of fatal and non-fatal CVD events, and (3) improves the overall risk discrimination, especially in younger patients [79]. However, this updated version has considerable advantages over the previous one, but still needs to be evaluated on CML patients.
The recently proposed HFA/ICOS risk assessment tool provides a new stratification tool at baseline in cancer patients undergoing cardiotoxic therapies, according to the category of exposure drugs, including BCR::ABL1 TKIs, and based on cardiovascular risk factors [15]. This score, included in the ESC Guidelines on Cardio-oncology, was retrospectively validated on a monocentric cohort of 58 CML patients, 32 of whom were men, with median age of 59 ± 15 y.o., where the HFA/ICOS stratification model allowed for a better risk evaluation of CML patients with a higher sensitivity when compared to the SCORE algorithm [80].
Regardless of the risk category, some general measures should be pursued, such as encouraging a healthy lifestyle, stopping smoking, and following a healthy and balanced diet. Moreover, risk modifiers (i.e., psychosocial factors, ethnicity, and imaging), lifetime CVD risk, treatment benefits, comorbidities, frailty, and patient preferences should also be taken into account.
Once stratified, if significant cardiovascular risks are found, it is mandatory to report the patients to a cardiologist in order to carry out further investigations and correct them before starting CML-specific therapy [14].

3.1. Modifiable Risk Factors

3.1.1. Hypertension

Arterial hypertension is one of the most important causes of preventable morbidity and mortality worldwide, affecting about 30–45% of the general population and predisposing them for severe pathologies such as coronary artery disease (CAD), cerebrovascular disease, atrial fibrillation (AF), and chronic kidney disease (CKD) [81]. In addition, it is also the most commonly reported cardiovascular comorbidity in cancer registries, reaching up to a third of cancer treatment [82].
In the CML population, the rate of hypertension was 10% in patients treated with 2G/3G-TKIs, especially in association with nilotinib (RR 2, p = 0.0002) and ponatinib (RR 9.21, p = 0.0002) [83]. In the last 10-year update of the pivotal ENESTnd trial, 16.1% of patients in the nilotinib 300 mg BID arm vs. 20.2% in the nilotinib 400 mg BID arm vs. 6.1% in the imatinib 400 mg QD arm developed hypertension [84]. In a real-life cohort, 94 patients were treated with frontline nilotinib, with a median age of 58 years at diagnosis, and arterial hypertension was the most commonly reported comorbidity (23.4%); after 28.6 months of treatment, 6 patients developed CTR-CVTs, 3 of which had baseline hypertension [85].
A high BP has also been found in several trials with ponatinib, both in pivotal trials and in real-world experiences. Notably, in 37% of the patients included in the PACE trial and 31% in the phase III EPIC trial, any grade of hypertension was found [3,86]. More recently, a dose optimization strategy was investigated in the OPTIC trial, where a dose reduction in ponatinib was performed according to molecular response. More than 20% of the 283 enrolled patients had controlled hypertension at baseline. The most common non-hematological side effect was, as expected, hypertension (28%), 9% of which was grade ≥ 3 in the last 3-year follow-up [87,88]. Finally, the OITI trial, a prospective–retrospective non-interventional trial based on the real-world data of patients treated with ponatinib, enrolled 119 patients, including 110 with CML in the Chronic Phase (CML-CP), with a median age of 60 years. Of these, 47.1% of the patients had a history of cardiovascular disease or hypertension; in a first and interim analysis, 59.7% of the patients experienced adverse events, among which grade 1–2 hypertension was one of the most commonly reported ones [89].
Among new agents, the Specifically Targeting the ABL Myristoyl Pocket (STAMP) inhibitor asciminib has been found to be particularly effective, even in presently pretreated patients, with an excellent toxicity profile. The safety and efficacy of asciminib in monotherapy or in combination with nilotinib, imatinib, or dasatinib were investigated in a phase I trial on CML patients resistant or intolerant to ≥2 different TKIs. Few AEs were reported, mainly represented by an increased lipase level in 26.7%, fatigue in 29.3%, headache in 28%, hypertension in 19%, and arthralgia and nausea in 24% [90]. In the phase 3 trial ASCEMBL, CML-CP patients treated with ≥2 previous TKIs were randomized to asciminib 40 mg BID vs. bosutinib 500 mg QD. At a median follow-up of 2.3 years, asciminib showed and maintained a higher efficacy than bosutinib, with a better toxicity profile. Overall, any-grade AEs were found in 91% of the patients treated with asciminib, 56.4% of which were grade 3. Among these, 18 (11.54%) experienced hypertension, of which 13 cases were within the first 12 months of treatment [91].
The pharmacological agents currently approved for hypertension treatment are divided into five classes of drugs, including Angiotensin-converting enzyme inhibitors (ACEi), Angiotensin II Receptor blockers (ARB), Beta-blockers, Calcium antagonists, and diuretics [92]. An over-activation of the RAS components in CML patients has been noted [93]. In addition, a study conducted on a large cohort of CML patients with arterial hypertension at diagnosis and treated with 2G/3G-TKIs reported a lower incidence of AOEs in the patients treated with RAS inhibitors compared to other treatments available (14.8% ± 4.2% vs. 44 ± 1%, p < 0.001) [94]. Overall, these data align with what is already known about the pro-atherogenic role of over-activated RAS in arterial hypertension and suggest the potential role of RAS inhibitors in CVD prevention for CML patients.
Recommendations: based on the aforementioned evidence, nilotinib and ponatinib have been shown to increase BP levels, so attention should be paid to patients with baseline hypertension or other cardiovascular risks factors, such as a history of CVD, T2D, or kidney failure. Standardized BP measurements, target organ damage research, and the exclusion of secondary causes of hypertension are necessary before TKI treatment. According to the evidence available in the literature, RAS inhibitors (ACEi or ARBs) could be a good option compared to other anti-hypertensive classes in patients with CML and treated with 2G/3G-TKIs TKIs [93].

3.1.2. Dyslipidemia

Dyslipidemia has a causal role in atherogenesis and is therefore considered to be one of the major cardiovascular risk factors: increased plasma levels of LDL can cause endothelial accumulation, thus inducing dysfunction and the subsequent inflammation of the vascular wall [95].
Treatment with nilotinib has been associated with dyslipidemia. Rea et al. first reported increased triglycerides and cholesterol plasma levels in 27 CML patients treated with nilotinib, identifying age, the duration of treatment, and pre-existing metabolic risk as risk factors for hypercholesterolemia [96]. Data from a large retrospective cohort of 369 CML patients treated with nilotinib showed a significantly increased incidence of AOEs at three months in patients with plasma cholesterol levels of >200 mg/dL and LDL levels of >70 mg/dL (21.9% vs. 6.2%, p < 0.01) [78].
Ponatinib has been proven to be particularly effective at inducing deep, early, and long-lasting molecular response, but this recourse has been limited by the increased incidence of AOEs in treated patients. Clinical trials and real-life cohorts have reported dyslipidemia as a frequent baseline comorbidity in patients who experience AOEs during treatment with ponatinib [3,86,97,98,99]. In particular, the association between dyslipidemia and AOEs during treatment with ponatinib was investigated in a cohort of 116 CML patients. Overall, 22% of the patients had dyslipidemia at diagnosis with CML, and 35% before starting ponatinib; a cardiovascular risk assessment using the SCORE algorithm defined 72% of patients as low risk (SCORE ≤5%) for CVD, while 28% were defined as high to very high risk (SCORE >5%). In a multivariate analysis, a high to very high SCORE risk confirmed this significant association with AOEs (HR 2.9, IC 95% 1–9.1; p = 0.04). In addition, a higher incidence of vascular events was reported in patients with pretreatment triglyceride levels of >200 mg/dL and after three months of treatment in patients with plasma cholesterol levels of >200 mg/dL and LDL levels of >70 mg/dL [100].
Recommendations: Nilotinib or ponatinib may be associated with an increased risk of CTR-CVTs, especially in older patients or those with pre-existing CV risk factors.
According to the 2022 ESC guidelines, for a comprehensive cardiovascular risk assessment, baseline laboratory tests, including those on lipid profile (total cholesterol, LDL, HDL, and triglycerides) and glycated hemoglobin (HbA1c), are recommended prior to initiating therapy [14]. Based on specific evidence derived from the CML population and in accordance with the 2021 ESC guidelines on cardiovascular risk prevention, special attention should be given to patients with a high SCORE2/OP risk who are candidates for nilotinib. In these patients, the LDL target should be <70 mg/dL. Caution is recommended in patients with a history of atherosclerotic cardiovascular disease (ASCVD), chronic kidney disease (CKD), and type 2 diabetes (T2D) with organ damage, particularly in very-high-risk patients. In these cases, plasma cholesterol levels should be maintained below 55 mg/dL [78,101].
Based on specific evidence derived from the CML population, patients undergoing ponatinib treatment aged >60 with established ASCVD, dyslipidemia, diabetes, or another cardiovascular risk factor, should be considered to be at a high risk for CTR-CVTs [100]. Lifestyle modification and target levels of LDL of <70 mg/dL are recommended for these patients. Moreover, a lipid-lowering statin-based therapy should be suggested to keep LDL levels <70 mg/dL. A combination with ezetimibe can be considered when the maximum dosage of statin therapy alone is insufficient to reach the target value. Treatment with a statin has been associated with adverse events that may limit its efficacy in lipid lowering; in these cases, a different schedule, such as every other day or twice a week, may be used [101]. Finally, prophylaxis with aspirin at 75–100 mg/day, or with clopidogrel at 75 mg/day in case of aspirin intolerance, should be considered in patients with CV risk factors, especially if they are aged >60 or have a preexisting history of CVD [77,102].

3.1.3. Diabetes and Pre-Diabetes

Diabetes mellitus and pre-diabetes are known independent risk factors for CAD and cerebrovascular disease, doubling the risk of ASCVD, and are frequently concomitant to dyslipidemia and hypertension. If not properly treated, diabetes is also responsible for life-threatening complications such as CKD, neuropathy, and retinopathy. Its treatment options include lifestyle changes, diet, oral hypoglycemics, and insulin therapy [103]. The increasing prevalence of T2D makes clear the importance of adequate glucose management, even in the CML population. Although imatinib and, less clearly, dasatinib, appear to have a positive effect on glucose metabolism, nilotinib has been associated with increased glucose levels and IR status during treatment [6]. In contrast, ponatinib has not been directly associated with hyperglycemia, but AOEs occur more frequently in patients with at least one CVD risk factor at baseline, including diabetes [102].
Recommendations: Based on the aforementioned evidence, nilotinib and ponatinib should be carefully evaluated in patients with uncontrolled diabetes, and a fasting plasma glucose (FPG) assessment is recommended before starting treatment. Furthermore, referring patients to an evaluation for diabetes is recommended in the case of FPG > 126 mg/dL (≥7.0 mmol/L) or Hb1Ac > 6.5% (48 mmol/mol), and in the case of FPG 100–125 mg/dL or Hb1Ac 5.6–6.4%, which are indicative of a pre-diabetes status. According to the 2019 ESC guidelines on diabetes, smoking cessation, a healthy lifestyle, and physical activity are the cornerstones of non-drug treatment. At the same time, oral hypoglycemic medications and insulin are recommended in patients where lifestyle changes are not enough. The therapeutic targets in these patients are represented by an Hb1Ac of <7.0% [103].

3.1.4. Smoking

Cigarette smoking, one of the most important risk factors for cardiovascular disease in the general population, is responsible for more than 50% of all avoidable deaths in smokers and increases the risk of CVD in smokers aged <50 by about five times compared to nonsmokers [101,104,105].
Smoking has proven to be a risk factor for developing CML [106,107], but has also been associated with an extremely unfavorable prognostic significance. Lauseker et al. assessed the impact of smoking on survival and progression towards the advanced stages of the disease: the 8-year survival probability for a nonsmoking patient was 87% vs. 83% for a patient who smoked, with a risk of death that was 2.08 times higher for smokers vs. nonsmokers, and a 2.11-times higher cause-specific hazard of disease progression [108]. Indeed, cigarette smoking is notoriously associated with an increased risk of atherogenesis, to which it contributes through several mechanisms, including the reduced bioavailability of NO, an increased expression of adhesion molecules, endothelial dysfunction, and increased levels of oxidized LDL, all factors that are potentially aggravated by TKIs such nilotinib or ponatinib [109,110]. Although there are no specific studies assessing the impact of early smoking cessation after diagnosis on the prognosis of CML patients, it would be of particular interest to assess whether the prognosis of early quitters is similar to that of nonsmokers.
Recommendations: According to the 2021 ESC guidelines on cardiovascular risk prevention, permanent smoking cessation should be encouraged in all smokers at diagnosis, since it is probably the most effective of all the preventive measures. Drug support includes nicotine replacement therapy (chewing gum, transdermal nicotine patches, nasal spray, inhaler, and sublingual tablets), bupropion, varenicline, and cytisine, although specific studies on the CML population are lacking [101].

3.1.5. Obesity

Obesity is a common disorder associated with an increased risk of comorbidity, including CVD, CKD, T2D, and hypertension [105], and many studies have supported the association between a high Body Mass Index (BMI) and an increased risk of cancer, both solid and hematologic [107]. Moreover, obesity has been recognized as a risk factor for CML [111,112].
The outcomes of obese CML patients were investigated by Breccia et al., who reported a correlation between an increased BMI and response to imatinib vs. nilotinib and dasatinib, with a delay in achieving a Complete Cytogenetic Response (CCyR, 6.8 months vs. 3.3 months, p = 0.01), a reduced incidence of major molecular response (MMR, 77% vs. 58% p = 0.03), and an increased rate of resistance [113]. In contrast, subsequent studies have found no significant difference in the molecular responses between patients with a low (<25 kg/m2) and high (>25 kg/m2) BMI treated with nilotinib or dasatinib in the front line [114].
Among the therapies for obese patients with a BMI ≥ 35 mg/m2 is bariatric surgery [115]. These surgeries alter bioavailability and drug metabolism, so these patients are often excluded from clinical trials. In a retrospective analysis of 22 patients with a history of bariatric surgery, a lower rate of early molecular responses (68% vs. 91%; p = 0.05) and a longer time to CCyR (6 vs. 3 months; p = 0.001) or MMR (12 vs. 6 months; p = 0.001) were reported when compared to 44 matched-control patients with no history of bariatric surgery. Moreover, bariatric surgery was associated with inferior event-free survival (5-year, 60% vs. 77%; p = 0.004) and failure-free survival (5-year, 32% vs. 63%; p < 0.0001) [116].
Recommendations: Based on specific evidence derived from the CML population and in accordance with the 2021 ESC guidelines on cardiovascular risk prevention, BMI should be included in the preliminary evaluation before starting TKI treatment. Compared to imatinib, 2G-TKIs appear to be the most suitable choice in obese subjects who need to achieve early and deep molecular responses, although cardiovascular risk and other comorbidities should nevertheless be considered in this subgroup of patients [113,114]. Attention should be paid, finally, to obese patients eligible for bariatric surgery, since it can alter TKIs’ absorption and metabolism, being able to influence patient outcomes. Despite this, no data support a change of dose in patients with a history of obesity or candidates for bariatric surgery.

3.1.6. Renal Function Impairment

Chronic Kidney Disease (CKD), defined as abnormalities in the structure or kidney function that persist for 3 months [117], is an important cardiovascular risk factor, with its mortality progressively increasing to triple when the estimated Glomerular Filtration Rate (eGFR) reaches 15 mL/min/1.73 m2 [101].
In CML, several studies in the literature have reported impaired glomerular function, especially in patients treated with imatinib. A median eGFR reduction of 2.77 mL/min/1.73 m2/years (p < 0.001) was reported in a cohort of 105 CML patients treated with imatinib, 12% of which developed CKD and 7% developed AKI [118]. In a retrospective analysis of 397 CML patients, 320 of them were treated with imatinib, 25 with dasatinib, and 53 with nilotinib. In the imatinib group, a median reduction in eGFR of 3 mL/min after 12 months of treatment was found. In addition, the authors reported an increased rate of CTR-CVTs in the imatinib cohort when compared to patients treated with 2G-TKIs [119]. A recent meta-analysis confirmed these data, reporting a significantly higher risk of renal impairment (CKD or AKI) in patients treated with imatinib than other TKIs (RR 2.7 IC 95% 1.49–4.91) [64].
An improvement in renal function was reported in 60 patients previously treated with imatinib that switched to a next-generation TKI [120], but nilotinib and dasatinib in the second- or third-line still seem to have mild effect on kidney function [66].
Nephrotoxicity was also explored by Cortes et al. in patients receiving bosutinib, both in the front-line and after one or more TKI. Like imatinib, bosutinib was associated with a median eGFR reduction of −15.62 mL/min/1.73 m2 from baseline to 48 months, with the use of loop diuretics, aminoglycosides, anti-hypertensive drugs, and grade 3–4 diarrhea being time-dependent prognostic factors for an eGFR of < 45 mL/min/1.73 m2 [65]. To note, renal dysfunction during the simultaneous intake of imatinib or bosutinib and anti-hypertensive therapy was reported in another experience, with rates of eGFR reduction of −10.1%/year (−12.3, −7.9; p = 0.04) and −5.7%/year (−6.6; −4.9; p < 0.01), especially when treated with RASi [68].
Recommendations: Based on the aforementioned evidence, TKIs have been shown to be safe when administered at standard doses in patients with modest levels of renal dysfunction. Imatinib and, to a lesser extent, bosutinib, have been associated with renal dysfunction in long-term management [64,65]. According to expert opinions, patients can be treated with the most suitable TKI and subjected to careful monitoring of their kidney function, with dose optimization if necessary. Moreover, the eGFR should be carefully monitored in patients simultaneously receiving RASi and imatinib or bosutinib [68].

3.2. Non-Modifiable Risk Factors

3.2.1. Age and Frailty

Age is considered to be a major driver of CVD risk, as incidence increases with ageing, both in men and women, exceeding 75% in people between 60 and 79 years and more than 86% in those aged >80 years. The high prevalence of CVD in older people has been linked to increased oxidative stress, inflammation, apoptosis, and myocardial dysfunction and deterioration [101,121].
The median age for CML diagnosis is between 50 and 60 years, with about 50% of cases being diagnosed in people aged ≥ 60 years [122]. Advanced age in CML patients can profoundly impact their management, as it can be a problem for the progressive deterioration of performance, co-morbidity, polypharmacy, and outcomes [123]. Finally, frailty, defined as a reduced resistance of the individual to stressors, is crucial to consider at diagnosis before starting therapy. It is also important to include functional capacity, co-morbidity burden, level of social support, and nutritional status [124]. In a retrospective analysis of 810 elderly patients with CML, with a median age of 75 (range 70–80), 26% had an underlying CVD and 86% had at least one CV risk factor. In the front-line, 63.1% received imatinib, 23.1% dasatinib, 12.5% nilotinib, and less than 1.4% bosutinib or ponatinib. Overall, 65.3% and 21.1% of patients received one or two lines of therapy, respectively. A multivariate analysis showed that the first-line patients treated with dasatinib were more likely to switch to a second line than those with imatinib (HR 1.9, IC 95% 1.35–2.66, p < 0.01). In addition, patients with more comorbidities were less likely to change from a first-line TKI than those without comorbidity (HR 0.66, IC 95% 0.46–0.95, p = 0.02) [125].
Recommendations: Based on specific evidence derived from the CML population and expert opinions, starting treatment with TKIs is crucial even in elderly patients, as they were all proved effective when elderly patients were compared to younger patients. Nevertheless, when selecting TKI therapy, special attention should be paid to comorbidities, polypharmacy, and frailty, since they can affect survival. Thus, imatinib seems to be the best choice to date for initial therapy in elderly patients with CML, due to its good cardiovascular toxicity profile. Dasatinib should be avoided, where possible, in patients with underlying lung disorders (e.g., Chronic Obstructive Pulmonary Disease or Pulmonary Hypertension), while nilotinib and ponatinib should be avoided in elderly patients with CVD risk factors [126].

3.2.2. Gender

The current international guidelines recommend integrating sex, gender, and gender identity considerations into individuals’ risk assessment and clinical management. In fact, not only do women present unique CVD risk factors (e.g., Polycystic Ovaric Syndrome or pregnancy-associated conditions), but seem to present different CVD manifestations and different responses to treatments [127].
Studies after introducing TKIs have shown no gender differences in the overall survival or response to TKIs. However, few studies have investigated the role of gender in CTR-CVTs during TKI therapy. Among them, a case–control study of AEs recorded in the FDA Adverse Events Reporting System (FAERS) database found that the CTR-CVTs rate in CML was higher in men than women exposed to TKIs (52.9% vs. 36.2%), compared to patients exposed to other anticancer drugs [128]. Karantanos et al., investigating the biological mechanism behind sex-related differences in Chronic Myeloid Neoplasm, including CML, reported a higher mutation frequency in DNMT3A and TET2 in women, which are known to be correlated with a higher CVD risk, but also a higher incidence in men of non-specific-MPN mutations, especially in ASXL1, IDH1/2, U2AF1, SRSF2, and EZH2, which are associated with worse outcomes in MPN patients [129].
Recommendations: Currently, there is no evidence to suggest a different CTR-CVTs risk prevention gender-based approach in the CML population. Rather, considering the data presented, it would be interesting to carry out an ad hoc study to assess the impact of gender on the risk of CTR-CVTs in patients treated with TKIs.

4. Conclusions

CTR-CVTs represent one of the main issues related to TKI treatment, thus limiting the available therapeutic options and leading to a dramatic worsening of prognoses. Moreover, CTR-CVTs have a strong impact on mortality, independent from CML. The current manuscript highlights the importance of multidisciplinary evaluations in CML patients, balancing the need to pursue TKI therapy with the specific burden of CVD and its risk. Finally, we emphasize tailored cardiovascular risk stratification in every CML patient. Accurate prevention protocols, the availability of safer agents, and dose optimization strategies may be the key to reducing the mobility and mortality in these patients.

Author Contributions

Conceptualization, A.C. and O.M.; writing—original draft preparation, A.C., R.P. and O.M.; writing—review and editing, A.C., R.P., O.M., G.C., F.M., F.T. and G.L.N.; supervision, G.L.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. García-Gutiérrez, V.; Hernández-Boluda, J.C. Tyrosine Kinase Inhibitors Available for Chronic Myeloid Leukemia: Efficacy and Safety. Front. Oncol. 2019, 9, 603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Bower, H.; Björkholm, M.; Dickman, P.W.; Höglund, M.; Lambert, P.C.; Andersson, T.M.-L. Life Expectancy of Patients with Chronic Myeloid Leukemia Approaches the Life Expectancy of the General Population. J. Clin. Oncol. 2016, 34, 2851–2857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lipton, J.H.; Chuah, C.; Guerci-Bresler, A.; Rosti, G.; Simpson, D.; Assouline, S.; Etienne, G.; Nicolini, F.E.; le Coutre, P.; Clark, R.E.; et al. Ponatinib versus Imatinib for Newly Diagnosed Chronic Myeloid Leukaemia: An International, Randomised, Open-Label, Phase 3 Trial. Lancet Oncol. 2016, 17, 612–621. [Google Scholar] [CrossRef] [PubMed]
  4. Dorer, D.J.; Knickerbocker, R.K.; Baccarani, M.; Cortes, J.E.; Hochhaus, A.; Talpaz, M.; Haluska, F.G. Impact of Dose Intensity of Ponatinib on Selected Adverse Events: Multivariate Analyses from a Pooled Population of Clinical Trial Patients. Leuk. Res. 2016, 48, 84–91. [Google Scholar] [CrossRef] [Green Version]
  5. Giles, F.J.; Rea, D.; Rosti, G.; Cross, N.C.P.; Steegmann, J.L.; Griskevicius, L.; le Coutre, P.; Coriu, D.; Petrov, L.; Ossenkoppele, G.J.; et al. Impact of Age on Efficacy and Toxicity of Nilotinib in Patients with Chronic Myeloid Leukemia in Chronic Phase: ENEST1st Subanalysis. J. Cancer Res. Clin. Oncol. 2017, 143, 1585–1596. [Google Scholar] [CrossRef] [Green Version]
  6. Li, S.; He, J.; Zhang, X.; Cai, Y.; Liu, J.; Nie, X.; Shi, L. Cardiovascular Adverse Events in Chronic Myeloid Leukemia Patients Treated with Nilotinib or Imatinib: A Systematic Review, Meta-Analysis and Integrative Bioinformatics Analysis. Front. Cardiovasc. Med. 2022, 9, 966182. [Google Scholar] [CrossRef]
  7. Cortes, J.; Mauro, M.; Steegmann, J.L.; Saglio, G.; Malhotra, R.; Ukropec, J.A.; Wallis, N.T. Cardiovascular and Pulmonary Adverse Events in Patients Treated with BCR-ABL Inhibitors: Data from the FDA Adverse Event Reporting System. Am. J. Hematol. 2015, 90, E66–E72. [Google Scholar] [CrossRef]
  8. Jain, P.; Kantarjian, H.; Jabbour, E.; Gonzalez, G.N.; Borthakur, G.; Pemmaraju, N.; Daver, N.; Gachimova, E.; Ferrajoli, A.; Kornblau, S.; et al. Ponatinib as First-Line Treatment for Patients with Chronic Myeloid Leukaemia in Chronic Phase: A Phase 2 Study. Lancet Haematol. 2015, 2, e376–e383. [Google Scholar] [CrossRef] [Green Version]
  9. Shah, N.P.; Wallis, N.; Farber, H.W.; Mauro, M.J.; Wolf, R.A.; Mattei, D.; Guha, M.; Rea, D.; Peacock, A. Clinical Features of Pulmonary Arterial Hypertension in Patients Receiving Dasatinib. Am. J. Hematol. 2015, 90, 1060–1064. [Google Scholar] [CrossRef]
  10. Rea, D.; Mauro, M.J.; Hochhaus, A.; Boquimpani, C.; Lomaia, E.; Voloshin, S.; Turkina, A.G.; Kim, D.-W.; Apperley, J.; Cortes, J.E.; et al. Efficacy and Safety Results from ASCEMBL, a Phase 3 Study of Asciminib versus Bosutinib (BOS) in Patients (Pts) with Chronic Myeloid Leukemia in Chronic Phase (CML-CP) after ≥2 Prior Tyrosine Kinase Inhibitors (TKIs): Week 96 Update. J. Clin. Oncol. 2022, 40, 7004. [Google Scholar] [CrossRef]
  11. Cortes, J.E.; Saikia, T.; Kim, D.-W.; Alvarado, Y.; Nicolini, F.E.; Rathnam, K.; Khattry, N.; Apperley, J.F.; Deininger, M.W.; de Lavallade, H.; et al. An Update of Safety and Efficacy Results from Phase 1 Dose-Escalation and Expansion Study of Vodobatinib, a Novel Oral BCR-ABL1 Tyrosine Kinase Inhibitor (TKI), in Patients with Chronic Myeloid Leukemia (CML) and Philadelphia Chromosome Positive Acute Lymphoblastic Leukemia (Ph+ ALL) Failing Prior TKI Therapies. Blood 2021, 138, 309. [Google Scholar] [CrossRef]
  12. Cortes, J.E.; Saikia, T.; Kim, D.-W.; Alvarado, Y.; Nicolini, F.E.; Rathnam, K.; Khattry, N.; Punatar, S.; Apperley, J.F.; Charbonnier, A.; et al. Efficacy and Safety of Vodobatinib in Patients (Pts) with Chronic Phase Philadelphia Positive Chronic Myeloid Leukemia (Ph+ CML): A Sub Group Analysis by Lines of Tyrosine Kinase Inhibitor (TKI) Therapy. Blood 2022, 140, 205–207. [Google Scholar] [CrossRef]
  13. Jiang, Q.; Li, Z.; Hou, Y.; Hu, Y.; Li, W.; Liu, X.; Xu, N.; Zhang, Y.; Song, Y.; Meng, L.; et al. Updated Results of Pivotal Phase 2 Trials of Olverembatinib (HQP1351) in Patients (Pts) with Tyrosine Kinase Inhibitor (TKI)-Resistant Chronic- and Accelerated-Phase Chronic Myeloid Leukemia (CML-CP and CML-AP) with T315I Mutation. Blood 2022, 140, 203–204. [Google Scholar] [CrossRef]
  14. Lyon, A.R.; López-Fernández, T.; Couch, L.S.; Asteggiano, R.; Aznar, M.C.; Bergler-Klein, J.; Boriani, G.; Cardinale, D.; Cordoba, R.; Cosyns, B.; et al. 2022 ESC Guidelines on Cardio-Oncology Developed in Collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur. Heart J. 2022, 43, 4229–4361. [Google Scholar] [CrossRef]
  15. Baseline Cardiovascular Risk Assessment in Cancer Patients Scheduled to Receive Cardiotoxic Cancer Therapies: A Position Statement and New Risk Assessment Tools from the Cardio-Oncology Study Group of the Heart Failure Association of the European Society of Cardiology in Collaboration with the International Cardio-Oncology Society—PubMed. Available online: https://pubmed.ncbi.nlm.nih.gov/32463967/ (accessed on 8 April 2023).
  16. Cohen, P. The Role of Protein Phosphorylation in Human Health and Disease: Delivered on June 30th 2001 at the FEBS Meeting in Lisbon. Eur. J. Biochem. 2001, 268, 5001–5010. [Google Scholar] [CrossRef]
  17. Rea, D. Management of Adverse Events Associated with Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia. Ann. Hematol. 2015, 94, 149–158. [Google Scholar] [CrossRef]
  18. Moslehi, J.J.; Deininger, M. Tyrosine Kinase Inhibitor–Associated Cardiovascular Toxicity in Chronic Myeloid Leukemia. J. Clin. Oncol. 2015, 33, 4210–4218. [Google Scholar] [CrossRef]
  19. Force, T.; Kolaja, K.L. Cardiotoxicity of Kinase Inhibitors: The Prediction and Translation of Preclinical Models to Clinical Outcomes. Nat. Rev. Drug Discov. 2011, 10, 111–126. [Google Scholar] [CrossRef]
  20. Rix, U.; Hantschel, O.; Dürnberger, G.; Remsing Rix, L.L.; Planyavsky, M.; Fernbach, N.V.; Kaupe, I.; Bennett, K.L.; Valent, P.; Colinge, J.; et al. Chemical Proteomic Profiles of the BCR-ABL Inhibitors Imatinib, Nilotinib, and Dasatinib Reveal Novel Kinase and Nonkinase Targets. Blood 2007, 110, 4055–4063. [Google Scholar] [CrossRef]
  21. Chen, W.; Du, B.; Liu, K.; Yu, Z.; Wang, X.; Yang, P. Nilotinib Related Acute Myocardial Infarction with Nonobstructive Coronary Arteries: A Case Report and Literature Review. BMC Cardiovasc. Disord. 2022, 22, 46. [Google Scholar] [CrossRef]
  22. Hamadi, A.; Grigg, A.P.; Dobie, G.; Burbury, K.L.; Schwarer, A.P.; Kwa, F.A.; Jackson, D.E. Ponatinib Tyrosine Kinase Inhibitor Induces a Thromboinflammatory Response. Thromb. Haemost. 2019, 119, 1112–1123. [Google Scholar] [CrossRef] [PubMed]
  23. Emir, H.; Albrecht-Schgoer, K.; Huber, K.; Grebien, F.; Eisenwort, G.; Schgoer, W.; Kaun, C.; Herndlhofer, S.; Theurl, M.; Cerny-Reiterer, S.; et al. Nilotinib Exerts Direct Pro-Atherogenic and Anti-Angiogenic Effects on Vascular Endothelial Cells: A Potential Explanation For Drug-Induced Vasculopathy In CML. Blood 2013, 122, 257. [Google Scholar] [CrossRef]
  24. Alhawiti, N.; Burbury, K.L.; Kwa, F.A.; O’Malley, C.J.; Shuttleworth, P.; Alzard, M.; Hamadi, A.; Grigg, A.P.; Jackson, D.E. The Tyrosine Kinase Inhibitor, Nilotinib Potentiates a Prothrombotic State. Thromb. Res. 2016, 145, 54–64. [Google Scholar] [CrossRef] [PubMed]
  25. Schmaier, A.H.; Merkulova, A.A.; Mitchell, S.; Stavrou, E.X. Ponatinib and Cardiovascular Complications. Blood 2016, 128, 3055. [Google Scholar] [CrossRef]
  26. Hadzijusufovic, E.; Albrecht-Schgoer, K.; Huber, K.; Hoermann, G.; Grebien, F.; Eisenwort, G.; Schgoer, W.; Herndlhofer, S.; Kaun, C.; Theurl, M.; et al. Nilotinib-Induced Vasculopathy: Identification of Vascular Endothelial Cells as a Primary Target Site. Leukemia 2017, 31, 2388–2397. [Google Scholar] [CrossRef] [Green Version]
  27. Mori, H.; Sukegawa, M.; Fukatsu, M.; Sano, T.; Takahashi, H.; Harada, K.; Kimura, S.; Ohkawara, H.; Nakamura, K.; Mita, M.; et al. The Link between Interleukin-1β and Acute Myocardial Infarction in Chronic Myeloid Leukemia Patients Treated with Nilotinib: Cross-Sectional Study. Ann. Hematol. 2020, 99, 359–361. [Google Scholar] [CrossRef]
  28. Libby, P. Interleukin-1 Beta as a Target for Atherosclerosis Therapy: Biological Basis of CANTOS and Beyond. J. Am. Coll. Cardiol. 2017, 70, 2278–2289. [Google Scholar] [CrossRef]
  29. Jaiswal, S.; Libby, P. Clonal Haematopoiesis: Connecting Ageing and Inflammation in Cardiovascular Disease. Nat. Rev. Cardiol. 2020, 17, 137–144. [Google Scholar] [CrossRef]
  30. Madonna, R.; Pieragostino, D.; Cufaro, M.C.; Doria, V.; Del Boccio, P.; Deidda, M.; Pierdomenico, S.D.; Dessalvi, C.C.; De Caterina, R.; Mercuro, G. Ponatinib Induces Vascular Toxicity through the Notch-1 Signaling Pathway. J. Clin. Med. 2020, 9, 820. [Google Scholar] [CrossRef] [Green Version]
  31. Tousif, S.; Singh, A.P.; Umbarkar, P.; Galindo, C.; Wheeler, N.; Toro Cora, A.; Zhang, Q.; Prabhu, S.D.; Lal, H. Ponatinib Drives Cardiotoxicity by S100A8/A9-NLRP3-IL-1β Mediated Inflammation. Circ. Res. 2023, 132, 267–289. [Google Scholar] [CrossRef]
  32. Camarda, N.; Travers, R.; Yang, V.K.; London, C.; Jaffe, I.Z. VEGF Receptor Inhibitor-Induced Hypertension: Emerging Mechanisms and Clinical Implications. Curr. Oncol. Rep. 2022, 24, 463–474. [Google Scholar] [CrossRef]
  33. Caocci, G.; Deidda, M.; Noto, A.; Greco, M.; Simula, M.P.; Mulas, O.; Cocco, D.; Fattuoni, C.; Mercuro, G.; La Nasa, G.; et al. Metabolomic Analysis of Patients with Chronic Myeloid Leukemia and Cardiovascular Adverse Events after Treatment with Tyrosine Kinase Inhibitors. J. Clin. Med. 2020, 9, 1180. [Google Scholar] [CrossRef] [Green Version]
  34. Herrmann, J. Vascular Toxic Effects of Cancer Therapies. Nat. Rev. Cardiol. 2020, 17, 503–522. [Google Scholar] [CrossRef]
  35. Mouton, A.J.; Li, X.; Hall, M.E.; Hall, J.E. Obesity, Hypertension, and Cardiac Dysfunction. Circ. Res. 2020, 126, 789–806. [Google Scholar] [CrossRef]
  36. Cheng, M.; Yang, F.; Liu, J.; Yang, D.; Zhang, S.; Yu, Y.; Jiang, S.; Dong, M. Tyrosine Kinase Inhibitors-Induced Arrhythmias: From Molecular Mechanisms, Pharmacokinetics to Therapeutic Strategies. Front. Cardiovasc. Med. 2021, 8, 758010. [Google Scholar] [CrossRef]
  37. Cui, X.; Sun, J.; Li, C.; Qiu, S.; Shi, C.; Ma, J.; Xu, Y. Downregulation of HERG Channel Expression by Tyrosine Kinase Inhibitors Nilotinib and Vandetanib Predominantly Contributes to Arrhythmogenesis. Toxicol. Lett. 2022, 365, 11–23. [Google Scholar] [CrossRef]
  38. Humbert, M.; Kovacs, G.; Hoeper, M.M.; Badagliacca, R.; Berger, R.M.F.; Brida, M.; Carlsen, J.; Coats, A.J.S.; Escribano-Subias, P.; Ferrari, P.; et al. 2022 ESC/ERS Guidelines for the Diagnosis and Treatment of Pulmonary Hypertension: Developed by the Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS). Endorsed by the International Society for Heart and Lung Transplantation (ISHLT) and the European Reference Network on Rare Respiratory Diseases (ERN-LUNG). Eur. Heart J. 2022, 43, 3618–3731. [Google Scholar] [CrossRef]
  39. Özgür Yurttaş, N.; Eşkazan, A.E. Dasatinib-Induced Pulmonary Arterial Hypertension. Br. J. Clin. Pharmacol. 2018, 84, 835–845. [Google Scholar] [CrossRef] [Green Version]
  40. Guignabert, C.; Phan, C.; Seferian, A.; Huertas, A.; Tu, L.; Thuillet, R.; Sattler, C.; Le Hiress, M.; Tamura, Y.; Jutant, E.-M.; et al. Dasatinib Induces Lung Vascular Toxicity and Predisposes to Pulmonary Hypertension. J. Clin. Investig. 2016, 126, 3207–3218. [Google Scholar] [CrossRef] [Green Version]
  41. Breccia, M.; Alimena, G. The Metabolic Consequences of Imatinib Mesylate: Changes on Glucose, Lypidic and Bone Metabolism. Leuk. Res. 2009, 33, 871–875. [Google Scholar] [CrossRef]
  42. Gottschalk, S.; Anderson, N.; Hainz, C.; Eckhardt, S.G.; Serkova, N.J. Imatinib (STI571)-Mediated Changes in Glucose Metabolism in Human Leukemia BCR-ABL-Positive Cells. Clin. Cancer Res. 2004, 10, 6661–6668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Markovits, N.; Kurnik, D.; Friedrich, C.; Gueta, I.; Halkin, H.; David, S.; Lomnicky, Y.; Topol, Y.; Tirosh, A.; Loebstein, R. Effects of Imatinib on Glycemic and Lipid Profiles: A Retrospective Cohort Study. Leuk. Lymphoma 2022, 63, 2224–2232. [Google Scholar] [CrossRef] [PubMed]
  44. Kumar, V.; Singh, P.; Gupta, S.K.; Ali, V.; Jyotirmayee; Verma, M. Alterations in Cellular Metabolisms after Imatinib Therapy: A Review. Med. Oncol. 2022, 39, 95. [Google Scholar] [CrossRef] [PubMed]
  45. Fitter, S.; Vandyke, K.; Gronthos, S.; Zannettino, A.C.W. Suppression of PDGF-Induced PI3 Kinase Activity by Imatinib Promotes Adipogenesis and Adiponectin Secretion. J. Mol. Endocrinol. 2012, 48, 229–240. [Google Scholar] [CrossRef] [Green Version]
  46. Breccia, M.; Muscaritoli, M.; Cannella, L.; Stefanizzi, C.; Frustaci, A.; Alimena, G. Fasting Glucose Improvement under Dasatinib Treatment in an Accelerated Phase Chronic Myeloid Leukemia Patient Unresponsive to Imatinib and Nilotinib. Leuk. Res. 2008, 32, 1626–1628. [Google Scholar] [CrossRef]
  47. Iizuka, K.; Niwa, H.; Kato, T.; Takeda, J. Dasatinib Improves Insulin Sensitivity and Affects Lipid Metabolism in a Patient with Chronic Myeloid Leukaemia. Case Rep. 2016, 2016, bcr2015214284. [Google Scholar] [CrossRef] [Green Version]
  48. Ono, K.; Suzushima, H.; Watanabe, Y.; Kikukawa, Y.; Shimomura, T.; Furukawa, N.; Kawaguchi, T.; Araki, E. Rapid Amelioration of Hyperglycemia Facilitated by Dasatinib in a Chronic Myeloid Leukemia Patient with Type 2 Diabetes Mellitus. Intern. Med. 2012, 51, 2763–2766. [Google Scholar] [CrossRef] [Green Version]
  49. Agostino, N.M.; Chinchilli, V.M.; Lynch, C.J.; Koszyk-Szewczyk, A.; Gingrich, R.; Sivik, J.; Drabick, J.J. Effect of the Tyrosine Kinase Inhibitors (Sunitinib, Sorafenib, Dasatinib, and Imatinib) on Blood Glucose Levels in Diabetic and Nondiabetic Patients in General Clinical Practice. J. Oncol. Pharm. Pract. 2011, 17, 197–202. [Google Scholar] [CrossRef]
  50. Salaami, O.; Kuo, C.-L.; Drake, M.T.; Kuchel, G.A.; Kirkland, J.L.; Pignolo, R.J. Antidiabetic Effects of the Senolytic Agent Dasatinib. Mayo Clin. Proc. 2021, 96, 3021–3029. [Google Scholar] [CrossRef]
  51. Palmer, A.K.; Xu, M.; Zhu, Y.; Pirtskhalava, T.; Weivoda, M.M.; Hachfeld, C.M.; Prata, L.G.; van Dijk, T.H.; Verkade, E.; Casaclang-Verzosa, G.; et al. Targeting Senescent Cells Alleviates Obesity-Induced Metabolic Dysfunction. Aging Cell 2019, 18, e12950. [Google Scholar] [CrossRef]
  52. Murakami, T.; Inagaki, N.; Kondoh, H. Cellular Senescence in Diabetes Mellitus: Distinct Senotherapeutic Strategies for Adipose Tissue and Pancreatic β Cells. Front. Endocrinol. 2022, 13, 869414. [Google Scholar] [CrossRef]
  53. Yu, L.; Liu, J.; Huang, X.; Jiang, Q. Adverse Effects of Dasatinib on Glucose-Lipid Metabolism in Patients with Chronic Myeloid Leukaemia in the Chronic Phase. Sci. Rep. 2019, 9, 17601. [Google Scholar] [CrossRef] [Green Version]
  54. Iurlo, A.; Orsi, E.; Cattaneo, D.; Resi, V.; Bucelli, C.; Orofino, N.; Sciumè, M.; Elena, C.; Grancini, V.; Consonni, D.; et al. Effects of First- and Second-Generation Tyrosine Kinase Inhibitor Therapy on Glucose and Lipid Metabolism in Chronic Myeloid Leukemia Patients: A Real Clinical Problem? Oncotarget 2015, 6, 33944–33951. [Google Scholar] [CrossRef] [Green Version]
  55. Racil, Z.; Koritakova, E.; Sacha, T.; Klamova, H.; Belohlavkova, P.; Faber, E.; Rea, D.; Malaskova, L.; Prochazkova, J.; Zackova, D.; et al. Insulin Resistance Is an Underlying Mechanism of Impaired Glucose Metabolism during Nilotinib Therapy. Am. J. Hematol. 2018, 93, E342–E345. [Google Scholar] [CrossRef] [Green Version]
  56. Racil, Z.; Razga, F.; Drapalova, J.; Buresova, L.; Zackova, D.; Palackova, M.; Semerad, L.; Malaskova, L.; Haluzik, M.; Mayer, J. Mechanism of Impaired Glucose Metabolism during Nilotinib Therapy in Patients with Chronic Myelogenous Leukemia. Haematologica 2013, 98, e124–e126. [Google Scholar] [CrossRef]
  57. Kosmas, C.E.; Bousvarou, M.D.; Kostara, C.E.; Papakonstantinou, E.J.; Salamou, E.; Guzman, E. Insulin Resistance and Cardiovascular Disease. J. Int. Med. Res. 2023, 51, 03000605231164548. [Google Scholar] [CrossRef]
  58. Sicuranza, A.; Ferrigno, I.; Abruzzese, E.; Iurlo, A.; Galimberti, S.; Gozzini, A.; Luciano, L.; Stagno, F.; Russo Rossi, A.; Sgherza, N.; et al. Pro-Inflammatory and Pro-Oxidative Changes During Nilotinib Treatment in CML Patients: Results of a Prospective Multicenter Front-Line TKIs Study (KIARO Study). Front. Oncol. 2022, 12, 835563. [Google Scholar] [CrossRef]
  59. Wages, P.A.; Kim, H.-Y.H.; Korade, Z.; Porter, N.A. Identification and Characterization of Prescription Drugs That Change Levels of 7-Dehydrocholesterol and Desmosterol. J. Lipid Res. 2018, 59, 1916–1926. [Google Scholar] [CrossRef] [Green Version]
  60. Zhang, X.; McDonald, J.G.; Aryal, B.; Canfrán-Duque, A.; Goldberg, E.L.; Araldi, E.; Ding, W.; Fan, Y.; Thompson, B.M.; Singh, A.K.; et al. Desmosterol Suppresses Macrophage Inflammasome Activation and Protects against Vascular Inflammation and Atherosclerosis. Proc. Natl. Acad. Sci. USA 2021, 118, e2107682118. [Google Scholar] [CrossRef]
  61. Spann, N.J.; Garmire, L.X.; McDonald, J.G.; Myers, D.S.; Milne, S.B.; Shibata, N.; Reichart, D.; Fox, J.N.; Shaked, I.; Heudobler, D.; et al. Regulated Accumulation of Desmosterol Integrates Macrophage Lipid Metabolism and Inflammatory Responses. Cell 2012, 151, 138–152. [Google Scholar] [CrossRef] [Green Version]
  62. Snodgrass, R.G.; Benatzy, Y.; Schmid, T.; Namgaladze, D.; Mainka, M.; Schebb, N.H.; Lütjohann, D.; Brüne, B. Efferocytosis Potentiates the Expression of Arachidonate 15-Lipoxygenase (ALOX15) in Alternatively Activated Human Macrophages through LXR Activation. Cell Death Differ. 2021, 28, 1301–1316. [Google Scholar] [CrossRef]
  63. Lin, Z.; Lin, X.; Lai, Y.; Han, C.; Fan, X.; Tang, J.; Mo, S.; Su, J.; Liang, S.; Shang, J.; et al. Ponatinib Modulates the Metabolic Profile of Obese Mice by Inhibiting Adipose Tissue Macrophage Inflammation. Front. Pharmacol. 2022, 13, 1040999. [Google Scholar] [CrossRef] [PubMed]
  64. Singh, A.K.; Hussain, S.; Ahmed, R.; Agrawal, N.; Bhurani, D.; Klugar, M.; Sharma, M. Impact of Imatinib Treatment on Renal Function in Chronic Myeloid Leukaemia Patients. Nephrology 2022, 27, 318–326. [Google Scholar] [CrossRef] [PubMed]
  65. Cortes, J.E.; Gambacorti-Passerini, C.; Kim, D.-W.; Kantarjian, H.M.; Lipton, J.H.; Lahoti, A.; Talpaz, M.; Matczak, E.; Barry, E.; Leip, E.; et al. Effects of Bosutinib Treatment on Renal Function in Patients with Philadelphia Chromosome-Positive Leukemias. Clin. Lymphoma Myeloma Leuk. 2017, 17, 684–695.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Ren, X.; Qin, Y.; Huang, X.; Zuo, L.; Jiang, Q. Assessment of Chronic Renal Injury in Patients with Chronic Myeloid Leukemia in the Chronic Phase Receiving Tyrosine Kinase Inhibitors. Ann. Hematol. 2019, 98, 1627–1640. [Google Scholar] [CrossRef]
  67. Al-Kali, A.; Farooq, S.; Tfayli, A. Tumor Lysis Syndrome after Starting Treatment with Gleevec in a Patient with Chronic Myelogenous Leukemia. J. Clin. Pharm. Ther. 2009, 34, 607–610. [Google Scholar] [CrossRef]
  68. Tsuda, M.; Hirata, A.; Tokunaga, S.; Masuda, T.; Haji, S.; Kimura, D.; Nojiri, C.; Nakashima, Y.; Shiratsuchi, M.; Kato, K.; et al. Rapid Decrease in EGFR with Concomitant Use of Tyrosine Kinase Inhibitors and Renin–Aldosterone–Angiotensin System Inhibitors in Patients with Chronic Myelogenous Leukemia. Int. J. Hematol. 2022, 116, 863–870. [Google Scholar] [CrossRef]
  69. Vidal-Petiot, E.; Rea, D.; Serrano, F.; Stehlé, T.; Gardin, C.; Rousselot, P.; Peraldi, M.-N.; Flamant, M. Imatinib Increases Serum Creatinine by Inhibiting Its Tubular Secretion in a Reversible Fashion in Chronic Myeloid Leukemia. Clin. Lymphoma Myeloma Leuk. 2016, 16, 169–174. [Google Scholar] [CrossRef]
  70. Abumiya, M.; Takahashi, N.; Takahashi, S.; Yoshioka, T.; Kameoka, Y.; Miura, M. Effects of SLC22A2 808G>T Polymorphism and Bosutinib Concentrations on Serum Creatinine in Patients with Chronic Myeloid Leukemia Receiving Bosutinib Therapy. Sci. Rep. 2021, 11, 6362. [Google Scholar] [CrossRef]
  71. Ozkurt, S.; Temiz, G.; Acikalin, M.F.; Soydan, M. Acute Renal Failure under Dasatinib Therapy. Ren. Fail. 2010, 32, 147–149. [Google Scholar] [CrossRef]
  72. Ochiai, S.; Sato, Y.; Minakawa, A.; Fukuda, A.; Fujimoto, S. Dasatinib-Induced Nephrotic Syndrome in a Patient with Chronic Myelogenous Leukemia: A Case Report. BMC Nephrol. 2019, 20, 87. [Google Scholar] [CrossRef]
  73. Quintas-Cardama, A.; Han, X.; Kantarjian, H.; Cortes, J. Dasatinib-Induced Platelet Dysfunction. Blood 2007, 110, 2941. [Google Scholar] [CrossRef]
  74. Sener, Y.; Okay, M.; Aydin, S.; Buyukasik, Y.; Akbiyik, F.; Dikmen, Z.G. TKI-Related Platelet Dysfunction Does Not Correlate with Bleeding in Patients with Chronic Phase-Chronic Myeloid Leukemia with Complete Hematological Response. Clin. Appl. Thromb. Hemost. 2019, 25, 1076029619858409. [Google Scholar] [CrossRef] [Green Version]
  75. Fox, L.C.; Cummins, K.D.; Costello, B.; Yeung, D.; Cleary, R.; Forsyth, C.; Tatarczuch, M.; Burbury, K.; Motorna, O.; Shortt, J.; et al. The Incidence and Natural History of Dasatinib Complications in the Treatment of Chronic Myeloid Leukemia. Blood Adv. 2017, 1, 802–811. [Google Scholar] [CrossRef]
  76. Piepoli, M.F.; Hoes, A.W.; Agewall, S.; Albus, C.; Brotons, C.; Catapano, A.L.; Cooney, M.-T.; Corrà, U.; Cosyns, B.; Deaton, C.; et al. 2016 European Guidelines on Cardiovascular Disease Prevention in Clinical Practice. Eur. Heart J. 2016, 37, 2315–2381. [Google Scholar] [CrossRef]
  77. Caocci, G.; Mulas, O.; Abruzzese, E.; Luciano, L.; Iurlo, A.; Attolico, I.; Castagnetti, F.; Galimberti, S.; Sgherza, N.; Bonifacio, M.; et al. Arterial Occlusive Events in Chronic Myeloid Leukemia Patients Treated with Ponatinib in the Real-Life Practice Are Predicted by the Systematic Coronary Risk Evaluation (SCORE) Chart. Hematol. Oncol. 2019, 37, 296–302. [Google Scholar] [CrossRef]
  78. Caocci, G.; Mulas, O.; Capodanno, I.; Bonifacio, M.; Annunziata, M.; Galimberti, S.; Luciano, L.; Tiribelli, M.; Martino, B.; Castagnetti, F.; et al. Low-Density Lipoprotein (LDL) Levels and Risk of Arterial Occlusive Events in Chronic Myeloid Leukemia Patients Treated with Nilotinib. Ann. Hematol. 2021, 100, 2005–2014. [Google Scholar] [CrossRef]
  79. Chipayo-Gonzales, D.; Ramakrishna, H.; Nuñez-Gil, I.J. Score2: A New Updated Algorithm to Predict Cardiovascular Disease Risk in Europe. J. Cardiothorac. Vasc. Anesth. 2022, 36, 18–21. [Google Scholar] [CrossRef]
  80. Di Lisi, D.; Madaudo, C.; Alagna, G.; Santoro, M.; Rossetto, L.; Siragusa, S.; Novo, G. The New HFA/ICOS Risk Assessment Tool to Identify Patients with Chronic Myeloid Leukaemia at High Risk of Cardiotoxicity. ESC Heart Fail. 2022, 9, 1914–1919. [Google Scholar] [CrossRef]
  81. Zhou, B.; Bentham, J.; Di Cesare, M.; Bixby, H.; Danaei, G.; Cowan, M.J.; Paciorek, C.J.; Singh, G.; Hajifathalian, K.; Bennett, J.E.; et al. Worldwide Trends in Blood Pressure from 1975 to 2015: A Pooled Analysis of 1479 Population-Based Measurement Studies with 19·1 Million Participants. Lancet 2017, 389, 37–55. [Google Scholar] [CrossRef] [Green Version]
  82. Jain, M.; Townsend, R.R. Chemotherapy Agents and Hypertension: A Focus on Angiogenesis Blockade. Curr. Hypertens. Rep. 2007, 9, 320–328. [Google Scholar] [CrossRef] [PubMed]
  83. Mulas, O.; Caocci, G.; Mola, B.; La Nasa, G. Arterial Hypertension and Tyrosine Kinase Inhibitors in Chronic Myeloid Leukemia: A Systematic Review and Meta-Analysis. Front. Pharmacol. 2021, 12, 674748. [Google Scholar] [CrossRef] [PubMed]
  84. Kantarjian, H.M.; Hughes, T.P.; Larson, R.A.; Kim, D.-W.; Issaragrisil, S.; le Coutre, P.; Etienne, G.; Boquimpani, C.; Pasquini, R.; Clark, R.E.; et al. Long-Term Outcomes with Frontline Nilotinib versus Imatinib in Newly Diagnosed Chronic Myeloid Leukemia in Chronic Phase: ENESTnd 10-Year Analysis. Leukemia 2021, 35, 440–453. [Google Scholar] [CrossRef] [PubMed]
  85. Del Corpo, O.; Harnois, M.; Busque, L.; Assouline, S. Real-Life Use of Nilotinib for Chronic Phase CML Demonstrates Similar Efficacy and Rate of Cardiovascular Events as Enestnd. Blood 2021, 138, 4602. [Google Scholar] [CrossRef]
  86. Cortes, J.E.; Kim, D.-W.; Pinilla-Ibarz, J.; le Coutre, P.D.; Paquette, R.; Chuah, C.; Nicolini, F.E.; Apperley, J.F.; Khoury, H.J.; Talpaz, M.; et al. Ponatinib Efficacy and Safety in Philadelphia Chromosome–Positive Leukemia: Final 5-Year Results of the Phase 2 PACE Trial. Blood 2018, 132, 393–404. [Google Scholar] [CrossRef]
  87. Cortes, J.; Apperley, J.; Lomaia, E.; Moiraghi, B.; Undurraga Sutton, M.; Pavlovsky, C.; Chuah, C.; Sacha, T.; Lipton, J.H.; Schiffer, C.A.; et al. Ponatinib Dose-Ranging Study in Chronic-Phase Chronic Myeloid Leukemia: A Randomized, Open-Label Phase 2 Clinical Trial. Blood 2021, 138, 2042–2050. [Google Scholar] [CrossRef]
  88. Cortes, J.E.; Deininger, M.W.; Lomaia, E.; Moiraghi, B.; Sutton, M.U.; Pavlovsky, C.; Chuah, C.; Sacha, T.; Lipton, J.H.; McCloskey, J.; et al. Three-Year Update from the Optic Trial: A Dose-Optimization Study of 3 Starting Doses of Ponatinib. Blood 2022, 140, 1495–1497. [Google Scholar] [CrossRef]
  89. Breccia, M.; Luciano, L.; Annunziata, M.; Attolico, I.; Malato, A.; Abruzzese, E.; Bonifacio, M.; Scortechini, A.R.; Cascavilla, N.; Di Renzo, N.; et al. Multicenter, Prospective and Retrospective Observational Cohort Study of Ponatinib in Patients with CML in Italy: Primary Analysis of the Oiti Trial. Blood 2021, 138, 3603. [Google Scholar] [CrossRef]
  90. Hughes, T.P.; Mauro, M.J.; Cortes, J.E.; Minami, H.; Rea, D.; DeAngelo, D.J.; Breccia, M.; Goh, Y.-T.; Talpaz, M.; Hochhaus, A.; et al. Asciminib in Chronic Myeloid Leukemia after ABL Kinase Inhibitor Failure. N. Engl. J. Med. 2019, 381, 2315–2326. [Google Scholar] [CrossRef]
  91. Hochhaus, A.; Réa, D.; Boquimpani, C.; Minami, Y.; Cortes, J.E.; Hughes, T.P.; Apperley, J.F.; Lomaia, E.; Voloshin, S.; Turkina, A.; et al. Asciminib vs Bosutinib in Chronic-Phase Chronic Myeloid Leukemia Previously Treated with at Least Two Tyrosine Kinase Inhibitors: Longer-Term Follow-up of ASCEMBL. Leukemia 2023, 37, 617–626. [Google Scholar] [CrossRef]
  92. Mancia, G.; Rosei, E.A.; Azizi, M.; Burnier, M.; Clement, D.L.; Coca, A.; de Simone, G.; Dominiczak, A.; Kahan, T.; Mahfoud, F.; et al. 2018 ESC/ESH Guidelines for the Management of Arterial Hypertension. Kardiol. Pol. 2019, 77, 71–159. [Google Scholar]
  93. Sayitoglu, M.; Haznedaroğlu, I.; Hatirnaz, O.; Erbilgin, Y.; Aksu, S.; Koca, E.; Adiguzel, C.; Bayik, M.; Akalin, I.; Gülbas, Z.; et al. Effects of Imatinib Mesylate on Renin–Angiotensin System (RAS) Activity during the Clinical Course of Chronic Myeloid Leukaemia. J. Int. Med. Res. 2009, 37, 1018–1028. [Google Scholar] [CrossRef]
  94. Mulas, O.; Caocci, G.; Stagno, F.; Bonifacio, M.; Annunziata, M.; Luciano, L.; Orlandi, E.M.; Abruzzese, E.; Sgherza, N.; Martino, B.; et al. Renin Angiotensin System Inhibitors Reduce the Incidence of Arterial Thrombotic Events in Patients with Hypertension and Chronic Myeloid Leukemia Treated with Second- or Third-Generation Tyrosine Kinase Inhibitors. Ann. Hematol. 2020, 99, 1525–1530. [Google Scholar] [CrossRef]
  95. Ference, B.A.; Ginsberg, H.N.; Graham, I.; Ray, K.K.; Packard, C.J.; Bruckert, E.; Hegele, R.A.; Krauss, R.M.; Raal, F.J.; Schunkert, H.; et al. Low-Density Lipoproteins Cause Atherosclerotic Cardiovascular Disease. 1. Evidence from Genetic, Epidemiologic, and Clinical Studies. A Consensus Statement from the European Atherosclerosis Society Consensus Panel. Eur. Heart J. 2017, 38, 2459–2472. [Google Scholar] [CrossRef] [Green Version]
  96. Rea, D.; Mirault, T.; Cluzeau, T.; Gautier, J.-F.; Guilhot, F.; Dombret, H.; Messas, E. Early Onset Hypercholesterolemia Induced by the 2nd-Generation Tyrosine Kinase Inhibitor Nilotinib in Patients with Chronic Phase-Chronic Myeloid Leukemia. Haematologica 2014, 99, 1197–1203. [Google Scholar] [CrossRef] [Green Version]
  97. Devos, T.; Havelange, V.; Theunissen, K.; Meers, S.; Benghiat, F.S.; Gadisseur, A.; Vanstraelen, G.; Vellemans, H.; Bailly, B.; Granacher, N.; et al. P699: Real-life outcomes of ponatinib treatment in patients with chronic myeloid leukemia (cml) or philadelphia chromosome-positive acute lymphoblastic leukemia (ph+all): 5-year-data from a belgian registry. HemaSphere 2022, 6, 594. [Google Scholar] [CrossRef]
  98. Mela Osorio, M.J.; Osycka, M.V.; Moiraghi, B.; Pavlovsky, M.A.; Varela, A.I.; Fernandez, I.; Sackmann Massa, F.; Ferrari, L.; Giere, I.; Sighel, C.; et al. P715: Multicenter observational study of ponatinib in cml patients in Argentina. Real-world experience. HemaSphere 2022, 6, 610–611. [Google Scholar] [CrossRef]
  99. Nichols, E.D.; Jabbour, E.; Jammal, N.; Chew, S.; Bryan, J.; Issa, G.; Garcia-Manero, G.; Sasaki, K.; DiPippo, A.; Kantarjian, H. Real-Life Incidence of Thrombotic Events in Leukemia Patients Treated with Ponatinib. Am. J. Hematol. 2022, 97, E350–E352. [Google Scholar] [CrossRef]
  100. Caocci, G.; Mulas, O.; Capodanno, I.; Abruzzese, E.; Iurlo, A.; Luciano, L.; Albano, F.; Annunziata, M.; Tiribelli, M.; Bonifacio, M.; et al. Low Low-Density Lipoprotein (LDL), Cholesterol and Triglycerides Plasma Levels Are Associated with Reduced Risk of Arterial Occlusive Events in Chronic Myeloid Leukemia Patients Treated with Ponatinib in the Real-Life. A Campus CML Study. Blood Cancer J. 2020, 10, 66. [Google Scholar] [CrossRef]
  101. Visseren, F.L.J.; Mach, F.; Smulders, Y.M.; Carballo, D.; Koskinas, K.C.; Bäck, M.; Benetos, A.; Biffi, A.; Boavida, J.-M.; Capodanno, D.; et al. 2021 ESC Guidelines on Cardiovascular Disease Prevention in Clinical Practice. Eur. Heart J. 2021, 42, 3227–3337. [Google Scholar] [CrossRef]
  102. Breccia, M.; Pregno, P.; Spallarossa, P.; Arboscello, E.; Ciceri, F.; Giorgi, M.; Grossi, A.; Mallardo, M.; Nodari, S.; Ottolini, S.; et al. Identification, Prevention and Management of Cardiovascular Risk in Chronic Myeloid Leukaemia Patients Candidate to Ponatinib: An Expert Opinion. Ann. Hematol. 2017, 96, 549–558. [Google Scholar] [CrossRef] [PubMed]
  103. Cosentino, F.; Grant, P.J.; Aboyans, V.; Bailey, C.J.; Ceriello, A.; Delgado, V.; Federici, M.; Filippatos, G.; Grobbee, D.E.; Hansen, T.B.; et al. 2019 ESC Guidelines on Diabetes, Pre-Diabetes, and Cardiovascular Diseases Developed in Collaboration with the EASD: The Task Force for Diabetes, Pre-Diabetes, and Cardiovascular Diseases of the European Society of Cardiology (ESC) and the European Association for the Study of Diabetes (EASD). Eur. Heart J. 2020, 41, 255–323. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Yusuf, S.; Hawken, S.; Ôunpuu, S.; Dans, T.; Avezum, A.; Lanas, F.; McQueen, M.; Budaj, A.; Pais, P.; Varigos, J.; et al. Effect of Potentially Modifiable Risk Factors Associated with Myocardial Infarction in 52 Countries (the INTERHEART Study): Case-Control Study. Lancet 2004, 364, 16. [Google Scholar] [CrossRef] [PubMed]
  105. Teo, K.K.; Rafiq, T. Cardiovascular Risk Factors and Prevention: A Perspective from Developing Countries. Can. J. Cardiol. 2021, 37, 733–743. [Google Scholar] [CrossRef]
  106. Musselman, J.R.B.; Blair, C.K.; Cerhan, J.R.; Nguyen, P.; Hirsch, B.; Ross, J.A. Risk of Adult Acute and Chronic Myeloid Leukemia with Cigarette Smoking and Cessation. Cancer Epidemiol. 2013, 37, 410–416. [Google Scholar] [CrossRef] [Green Version]
  107. Kabat, G.C.; Wu, J.W.; Moore, S.C.; Morton, L.M.; Park, Y.; Hollenbeck, A.R.; Rohan, T.E. Lifestyle and Dietary Factors in Relation to Risk of Chronic Myeloid Leukemia in the NIH-AARP Diet and Health Study. Cancer Epidemiol. Biomark. Prev. 2013, 22, 848–854. [Google Scholar] [CrossRef] [Green Version]
  108. Lauseker, M.; Hasford, J.; Saussele, S.; Kremers, S.; Kraemer, D.; Lindemann, W. Smokers with Chronic Myeloid Leukemia Are at a Higher Risk of Disease Progression and Premature Death. Cancer 2017, 123, 2467–2471. [Google Scholar] [CrossRef] [Green Version]
  109. Messner, B.; Bernhard, D. Smoking and Cardiovascular Disease. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 509–515. [Google Scholar] [CrossRef] [Green Version]
  110. Caligiuri, S.P.B.; Pierce, G.N.; Ravandi, A.; Aukema, H.M. The Plasma Oxylipidome Links Smoking Status to Peripheral Artery Disease. Metabolites 2022, 12, 627. [Google Scholar] [CrossRef]
  111. Strom, S.S.; Yamamura, Y.; Kantarijian, H.M.; Cortes-Franco, J.E. Obesity, Weight Gain, and Risk of Chronic Myeloid Leukemia. Cancer Epidemiol. Biomark. Prev. 2009, 18, 1501–1506. [Google Scholar] [CrossRef] [Green Version]
  112. Larsson, S.C.; Wolk, A. Overweight and Obesity and Incidence of Leukemia: A Meta-Analysis of Cohort Studies. Int. J. Cancer 2008, 122, 1418–1421. [Google Scholar] [CrossRef]
  113. Breccia, M.; Loglisci, G.; Salaroli, A.; Serrao, A.; Mancini, M.; Diverio, D.; Latagliata, R.; Alimena, G. Delayed Cytogenetic and Major Molecular Responses Associated to Increased BMI at Baseline in Chronic Myeloid Leukemia Patients Treated with Imatinib. Cancer Lett. 2013, 333, 32–35. [Google Scholar] [CrossRef]
  114. Molica, M.; Canichella, M.; Colafigli, G.; Latagliata, R.; Diverio, D.; Alimena, G.; Foà, R.; Breccia, M. Body Mass Index Does Not Impact on Molecular Response Rate of Chronic Myeloid Leukaemia Patients Treated Frontline with Second Generation Tyrosine Kinase Inhibitors. Br. J. Haematol. 2018, 182, 427–429. [Google Scholar] [CrossRef] [Green Version]
  115. Eisenberg, D.; Shikora, S.A.; Aarts, E.; Aminian, A.; Angrisani, L.; Cohen, R.V.; Luca, M.D.; Faria, S.L.; Goodpaster, K.P.S.; Haddad, A.; et al. 2022 American Society for Metabolic and Bariatric Surgery (ASMBS) and International Federation for the Surgery of Obesity and Metabolic Disorders (IFSO): Indications for Metabolic and Bariatric Surgery. Surg. Obes. Relat. Dis. 2022, 18, 1345–1356. [Google Scholar] [CrossRef]
  116. Haddad, F.G.; Kantarjian, H.M.; Bidikian, A.; Jabbour, E.J.; Short, N.J.; Ning, J.; Xiao, L.; Pemmaraju, N.; DiNardo, C.D.; Kadia, T.M.; et al. Association between Bariatric Surgery and Outcomes in Chronic Myeloid Leukemia. Cancer 2023, 129, 1866–1872. [Google Scholar] [CrossRef]
  117. Chapter 1: Definition and Classification of CKD. Kidney Int. Suppl. 2013, 3, 19–62. [CrossRef] [Green Version]
  118. Marcolino, M.S.; Boersma, E.; Clementino, N.C.D.; Macedo, A.V.; Marx-Neto, A.D.; Silva, M.H.C.R.; van Gelder, T.; Akkerhuis, K.M.; Ribeiro, A.L. Imatinib Treatment Duration Is Related to Decreased Estimated Glomerular Filtration Rate in Chronic Myeloid Leukemia Patients. Ann. Oncol. 2011, 22, 2073–2079. [Google Scholar] [CrossRef]
  119. Molica, M.; Scalzulli, E.; Colafigli, G.; Fegatelli, D.A.; Massaro, F.; Latagliata, R.; Foà, R.; Breccia, M. Changes in Estimated Glomerular Filtration Rate in Chronic Myeloid Leukemia Patients Treated Front Line with Available TKIs and Correlation with Cardiovascular Events. Ann. Hematol. 2018, 97, 1803–1808. [Google Scholar] [CrossRef]
  120. Hino, A.; Yoshida, H.; Tada, Y.; Koike, M.; Minami, R.; Masaie, H.; Ishikawa, J. Changes from Imatinib Mesylate to Second Generation Tyrosine Kinase Inhibitors Improve Renal Impairment with Imatinib Mesylate in Chronic Myelogenous Leukemia. Int. J. Hematol. 2016, 104, 605–611. [Google Scholar] [CrossRef]
  121. Rodgers, J.L.; Jones, J.; Bolleddu, S.I.; Vanthenapalli, S.; Rodgers, L.E.; Shah, K.; Karia, K.; Panguluri, S.K. Cardiovascular Risks Associated with Gender and Aging. J. Cardiovasc. Dev. Dis. 2019, 6, 19. [Google Scholar] [CrossRef] [Green Version]
  122. Hoffmann, V.S.; Baccarani, M.; Hasford, J.; Lindoerfer, D.; Burgstaller, S.; Sertic, D.; Costeas, P.; Mayer, J.; Indrak, K.; Everaus, H.; et al. The EUTOS Population-Based Registry: Incidence and Clinical Characteristics of 2904 CML Patients in 20 European Countries. Leukemia 2015, 29, 1336–1343. [Google Scholar] [CrossRef] [PubMed]
  123. Iurlo, A.; Ubertis, A.; Artuso, S.; Bucelli, C.; Radice, T.; Zappa, M.; Cattaneo, D.; Mari, D.; Cortelezzi, A. Comorbidities and Polypharmacy Impact on Complete Cytogenetic Response in Chronic Myeloid Leukaemia Elderly Patients. Eur. J. Intern. Med. 2014, 25, 63–66. [Google Scholar] [CrossRef] [PubMed]
  124. Breccia, M.; Palandri, F.; Luciano, L.; Benevolo, G.; Bonifacio, M.; Caocci, G.; Castagnetti, F.; Palumbo, G.A.; Iurlo, A.; Landi, F. Identification and Assessment of Frailty in Older Patients with Chronic Myeloid Leukemia and Myelofibrosis, and Indications for Tyrosine Kinase Inhibitor Treatment. Ann. Hematol. 2018, 97, 745–754. [Google Scholar] [CrossRef]
  125. Shallis, R.M.; Wang, R.; Bewersdorf, J.P.; Zeidan, A.M.; Davidoff, A.J.; Huntington, S.F.; Podoltsev, N.A.; Ma, X. Contemporary Practice Patterns of Tyrosine Kinase Inhibitor Use among Older Patients with Chronic Myeloid Leukemia in the United States. Ther. Adv. Hematol. 2021, 12, 20406207211043404. [Google Scholar] [CrossRef] [PubMed]
  126. Luskin, M.R.; DeAngelo, D.J. How to Treat Chronic Myeloid Leukemia (CML) in Older Adults. J. Geriatr. Oncol. 2018, 9, 291–295. [Google Scholar] [CrossRef]
  127. Ferrannini, G.; Maldonado, J.M.; Raha, S.; Rao-Melacini, P.; Khatun, R.; Atisso, C.; Shurzinske, L.; Gerstein, H.C.; Rydén, L.; Bethel, M.A. Gender Differences in Cardiovascular Risk, Treatment, and Outcomes: A Post Hoc Analysis from the REWIND Trial. Scand. Cardiovasc. J. 2023, 57, 2166101. [Google Scholar] [CrossRef]
  128. Cirmi, S.; El Abd, A.; Letinier, L.; Navarra, M.; Salvo, F. Cardiovascular Toxicity of Tyrosine Kinase Inhibitors Used in Chronic Myeloid Leukemia: An Analysis of the FDA Adverse Event Reporting System Database (FAERS). Cancers 2020, 12, 826. [Google Scholar] [CrossRef] [Green Version]
  129. Karantanos, T.; Jain, T.; Moliterno, A.R.; Jones, R.J.; DeZern, A.E. Sex-Related Differences in Chronic Myeloid Neoplasms: From the Clinical Observation to the Underlying Biology. Int. J. Mol. Sci. 2021, 22, 2595. [Google Scholar] [CrossRef]
Figure 1. Mechanisms underlying off-target toxicities during TKIs treatment. AKI: Acute Kidney Injury; DHCR24: 24-Dehydrocholesterol Reductase; eGFR: estimated Glomerular Filtration Rate; hERG: human Ether-à-go-go-Related Gene; IL1β: Interleukin-1 β; IRS1: Insulin receptor substrate 1; LDLR: Low-density Lipoprotein Receptor; OCT2: Organic Cation Transporter 2; NLRP3: NOD-, LRR- Pyrin Domain-containing 3; oxLDL: oxidized Low-density Lipoprotein; PDGFR: Platelet-derived Growth Factor Receptor; ROS: reactive oxygen species; sCr: serum Creatinine; TLR4: Toll-like Receptor 4; VEGFR: Vascular Endothelial Growth Factor Receptor; and VLDLR: Very-Low-density Lipoprotein Receptor.
Figure 1. Mechanisms underlying off-target toxicities during TKIs treatment. AKI: Acute Kidney Injury; DHCR24: 24-Dehydrocholesterol Reductase; eGFR: estimated Glomerular Filtration Rate; hERG: human Ether-à-go-go-Related Gene; IL1β: Interleukin-1 β; IRS1: Insulin receptor substrate 1; LDLR: Low-density Lipoprotein Receptor; OCT2: Organic Cation Transporter 2; NLRP3: NOD-, LRR- Pyrin Domain-containing 3; oxLDL: oxidized Low-density Lipoprotein; PDGFR: Platelet-derived Growth Factor Receptor; ROS: reactive oxygen species; sCr: serum Creatinine; TLR4: Toll-like Receptor 4; VEGFR: Vascular Endothelial Growth Factor Receptor; and VLDLR: Very-Low-density Lipoprotein Receptor.
Hemato 04 00017 g001
Figure 2. Proposed algorithm for prevention and management of BCR::ABL1 TKIs cardiovascular toxicities according to the ESC Guidelines on CVR prevention and the most recent ESC guidelines on Cardioncology. The algorithm emphasizes the importance of mitigating CVR after adequate assessment of risk factor at baseline according to the new HFA/ICOS scoring system. With this aim, lifestyle intervention, implementation of cardiovascular therapy, tailored follow-up and TKIs dose-optimization strategies are crucial. Recommendation classes and levels of evidence are also reported where possible. ACE inhibitors: Angiotensin Converting Enzyme inhibitors; ARB: Angiotensin Receptor Blockers; ASCVD: Atherosclerotic Cardiovascular Disease; BP: Blood Pressure; CKD: Chronic Kidney Disease; EF: Ejection Fraction; eGFR: estimated Glomerular Filtration Rate; LDL: Low-density Lipoprotein; RAS inhibitors: Renin-Angiotensin System Inhibitors; SCORE2/OP: Systemic Coronary Risk Estimation/SCORE2 Older people; Tyrosine Kinase Inhibitors; TTE: transthoracic Echocardiogram; and T2D: Type 2 Diabetes.
Figure 2. Proposed algorithm for prevention and management of BCR::ABL1 TKIs cardiovascular toxicities according to the ESC Guidelines on CVR prevention and the most recent ESC guidelines on Cardioncology. The algorithm emphasizes the importance of mitigating CVR after adequate assessment of risk factor at baseline according to the new HFA/ICOS scoring system. With this aim, lifestyle intervention, implementation of cardiovascular therapy, tailored follow-up and TKIs dose-optimization strategies are crucial. Recommendation classes and levels of evidence are also reported where possible. ACE inhibitors: Angiotensin Converting Enzyme inhibitors; ARB: Angiotensin Receptor Blockers; ASCVD: Atherosclerotic Cardiovascular Disease; BP: Blood Pressure; CKD: Chronic Kidney Disease; EF: Ejection Fraction; eGFR: estimated Glomerular Filtration Rate; LDL: Low-density Lipoprotein; RAS inhibitors: Renin-Angiotensin System Inhibitors; SCORE2/OP: Systemic Coronary Risk Estimation/SCORE2 Older people; Tyrosine Kinase Inhibitors; TTE: transthoracic Echocardiogram; and T2D: Type 2 Diabetes.
Hemato 04 00017 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Costa, A.; Pittorru, R.; Caocci, G.; Migliore, F.; Tona, F.; Mulas, O.; La Nasa, G. The Direct and Indirect Effects of Tyrosine Kinase Inhibitors on the Cardiovascular System in Chronic Myeloid Leukemia. Hemato 2023, 4, 207-226. https://doi.org/10.3390/hemato4030017

AMA Style

Costa A, Pittorru R, Caocci G, Migliore F, Tona F, Mulas O, La Nasa G. The Direct and Indirect Effects of Tyrosine Kinase Inhibitors on the Cardiovascular System in Chronic Myeloid Leukemia. Hemato. 2023; 4(3):207-226. https://doi.org/10.3390/hemato4030017

Chicago/Turabian Style

Costa, Alessandro, Raimondo Pittorru, Giovanni Caocci, Federico Migliore, Francesco Tona, Olga Mulas, and Giorgio La Nasa. 2023. "The Direct and Indirect Effects of Tyrosine Kinase Inhibitors on the Cardiovascular System in Chronic Myeloid Leukemia" Hemato 4, no. 3: 207-226. https://doi.org/10.3390/hemato4030017

Article Metrics

Back to TopTop