Skip to main content
Erschienen in: Medical Oncology 8/2019

Open Access 01.08.2019 | Review Article

Anti-cancer drugs-induced arterial injury: risk stratification, prevention, and treatment

verfasst von: Edit Gara, Kristóf György Csikó, Zoltán Ruzsa, Gábor Földes, Béla Merkely

Erschienen in: Medical Oncology | Ausgabe 8/2019

Abstract

Vascular side effects of standard chemotherapeutic drugs and novel anti-tumor agents complicate treatment cycles, increase non-cancer-related mortality rates, and decrease the quality of life in cancer survivors. Arterial thromboembolic events (ATEE) are associated with most anti-cancer medications. Previous articles have reported a variety of vascular events including ST-segment elevation myocardial infarction as one of the most severe acute arterial attacks. Cardiologists should play an early role in identifying those at high risk for vascular complications and tailor anti-thrombotic therapies in keeping with thromboembolic and bleeding risks. Early preventive steps and individualized chemotherapy may decrease anti-tumor treatment-related vascular events. Here, we aim to provide an extensive review of anti-tumor drug-induced vascular injury (DIVI), pathomechanisms, and risk stratification underlining arterial events. We give a summary of clinical manifestations, treatment options, and possible preventive measures of DIVI. Additionally, the treatment of modifiable risk factors and tailored choice of chemotherapy must be considered in all oncology patients to prevent DIVI. We propose a complex tool for ATEE risk stratification which is warranted for early prediction leading to less frequent complications in cancer patients.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ATEE
Arterial thromboembolic events
BEP
Bleomycin–etoposide–cisplatin
CAD
Coronary arterial diseases
DIVI
Drug-induced vascular injury
PAD
Peripheral arterial diseases
TKI
Tyrosine kinase inhibitors
vWF
von Willebrand factor

Introduction

Severe cardiovascular adverse events related to anti-tumor treatments have long been discussed including arterial thromboembolic events (ATEE), coronary and peripheral arterial diseases, secondary myocardial dysfunction, and arrhythmias [1]. Chemotherapy-related major adverse events may have long-term effects on quality of life, especially in early ages at the time of treatment. For instance, testicular cancer is the most frequent solid tumor in men aged 15–35 years with an incident rate of 8/100,000–12/100,000 in developed countries [2]. Cisplatin, often used to treat testicular cancer, and other chemotherapeutic drugs also affect the vascular system and cause either acute (mainly thrombotic events) or chronic (e.g., arterial hypertension) arterial injury. A common regulatory or signaling pathway responsible for drug-induced vascular injury (DIVI) has not yet been discovered as each chemotherapeutic drug acts via different mechanisms. Treatment of DIVI, such as coronary interventions or hospitalization due to major adverse events, may interrupt or delay oncology therapy cycles. Additionally, cardiac function may be severely impaired if the side effects are recognized late in treatment (e.g., ischemic heart diseases) [3]. Therefore, oncology and cardiac specialists must cooperate so that mortality and survival rates of cancer patients can improve. Here, we aim to review and underpin the clinical importance of early risk stratification, prevention, and treatment of DIVI in arteries.

Alkylating agents and platinum compounds

Platinum-based treatments develop chemical bonds on cellular phospholipid membranes and cause direct cytotoxicity. ATEE are the most frequent vascular side effects (Table 1) induced via elevated coagulant state and direct endothelial toxic effects in response to alkylating agents [4]. During bleomycin–etoposide–cisplatin (BEP) treatment, levels of von Willebrand factor (vWF), intercellular adhesion molecule-1 (ICAM-1), plasminogen activator inhibitor-1 (PAI-1), and tissue plasminogen activator (tPA), which are principal regulators of blood clotting, all increased [5]. Of note, peroxisome proliferator-activated receptor (PPAR) agonists such as rosiglitazone and troglitazone, and insulin sensitisers, were demonstrated to protect against cisplatin-induced DIVI [6, 7]. Moore et al. conducted a retrospective analysis, including 932 patients treated with cisplatin-based chemotherapy, and reported an 18.1% incidence rate of early DIVI which happened either during treatment or within 4 weeks after the last dose [6].
Table 1
Suggested pathomechanisms and possible treatments of arterial DIVI
Drug class
Pathomechanisms
Clinical manifestations
Prevention/treatment
Alkylating agents
Direct endothelial toxic effect
DNA crosslinking
Decreased plasma VEGF
Pulmonary arterial thrombosis
Pulmonary arterial hypertension
Serous pericarditis
Pulmonary fibrosis
Amifostineb
 Cyclophosphamide, Ifosfamide
Platinum compounds
Direct endothelial damage
Increased ICAM-1 expression
Increased tPA and PAI-1
Platelet activation
Elevated plasma levels of vWF, FVIII
Increased PAI-1/tPA ratio
Increased production of pro-inflammatory endothelial micro particles
Enhancement of thrombin generation
Induced caspase 3, 7 activity
Decreased NOS activity
Increased intima-media thickness
Acute ischemic stroke
Acute myocardial infarction
Bilateral retinal ischemia
Lower limb digital necrosis
Raynaud’s phenomenon
Abdominal aortic thrombosis
Celiac trunk thrombosis and splenic infarction
PPAR agonists: rosiglitazonea
troglitazonea
LMWH2
ASAb
 Cisplatin
Topoisomerase inhibitors
DNA (TopIIb) damage
ROS generation
Decreased endothelium-mediated relaxation
Induced caspase 3, 7 activity, apoptosis
Angiopathy
Microvascular obstruction
Raynaud’s phenomenon
Zofenoprilata
Dexrazoxanec
 Anthracyclines: doxorubicin, epirubicin
Anti-microtubule agents
Impaired of endothelial migration and proliferation
Apoptosis induction
Downregulation of thrombomodulin
Increased TF expression
Impaired NO-mediated vasodilation
Acute coronary syndrome
Vasoconstriction induced by paclitaxel-eluting stents
Late stent thrombosis
Dual anti-platelet therapyc
 Taxanes: docetaxel, paclitaxel
 Vinca alkaloids: vincristine, vinblastine, vinorelbine
Inhibition of endothelial migration and proliferation
Increased endothelial cell death
Increased ROS production
Microangiopathy
N-acetylcysteineb
Anti-metabolites
Direct endothelial damage
PKC-mediated vasoconstriction
ROS formation
Inhibition of VEGF-mediated angiogenesis
Increased level of endothelin-1
Apoptosis induction
Elevated homocysteine levels
Angina
Arrhythmias
Myocardial ischemia Myocardial infarction
Probucolb
Dalteparinb
Nitro-glycerinea,b,c
N-acetylcysteinea
Calcium channel blockersc
Folic acidc
Curcumina
 5-fluorouracil, capecitabine, methotrexate
 Targeted therapeutics
Impaired VEGF signaling (endothelial survival)
Reduced NO production
Increased ET-1 release
Inhibition of PDGFR
Hypertension
Myocardial ischemia and infarction
Cerebrovascular attack
Peripheral arterial ischemia
Hypertension
Takotsubo cardiomyopathy
Anti-platelet drugsc
LMWHc
Low-dose acetylsalicylatec
 VEGF signaling pathway inhibitors: bevacizumab (mAb)
sunitinib, sorafenib (TKIs)
 Anti-HER2: bevacizumab, trastuzumab
Anti-CD20 (rituximab)
Vasospasm
Arterial thrombosis
Anti-platelet drugs
 BCR-ABL1 inhibitor TKIs: Nilotinib, dasatinib, ponatinib
Inhibition of TIE-2/TEK
Increased endothelial of ICAM-1, VCAM-1, E-selectin
Induction of casp-3/7 mediated apoptosis
Inhibition of PDGFR
Inhibition of VEGFR-2
Vasoconstriction
PAD
Acute coronary syndromes
Ischemic cerebrovascular events
Pulmonary arterial hypertension
Systemic arterial hypertension
Acute ischemic stroke
Anti-platelet drugsc
Hormonal therapies
Endothelial damage
Hypercholesterolemia
Myocardial infarction
Thrombotic vascular events
Anti-platelet drugs
 Aromatase inhibitor, tamoxifen
Immunomodulatory drugs
Vasoconstriction
Immune-mediated reaction
Angiopathy
LMWHc
Acetylsalicylatec
Ipilimumab
Antibiotics with anti-cancer effect
FAS ligand upregulation
Increased production of ROS: bleomycin-Fe(II)
NOS inhibition
Increased ICAM-1 expression
Increased amounts of tPA and PAI-1
Limb ischemia
Raynaud’s phenomenon
PPAR agonists: rosiglitazoneb troglitazoneb
bleomycin
ASA acetylsalicylate, DNA deoxyribonucleic acid, ET-1 endothelin-1, FVIII factor VIII, ICAM-1 intercellular adhesion molecule-1, LMWH low-molecular-weight heparin, NO nitric oxide, NOS nitric oxide synthase, PAD peripheral artery disease, PAI-1 plasminogen activator inhibitor-1, PDGFR platelet-derived growth factor receptor, PKC protein kinase C, PPAR peroxisome proliferator-activated receptors, ROS reactive oxygen species, TF tissue factor, TIE-2, TEK angiopoietin-1 receptor, tPA tissue plasminogen activator, VCAM vascular cell adhesion molecule-1, VEGF vascular endothelial growth factor, VEGFR-2 vascular endothelial growth factor receptor-2, vWF von Willebrand Factor
aIn vitro preclinical
bIn vivo preclinical
cIn guidelines
Half of these vascular events were presented as deep vein thrombosis (50%), followed by pulmonary embolisms (25%), or a combination of the two (13.6%). About 2% of the patients developed arterial thrombosis, including cerebrovascular attack, and myocardial infarction. As well as causing early ATEE, endothelial dysfunction may lead to chronic vascular diseases. For instance, arterial hypertension usually occurs 10–20 years after cisplatin treatment [7]. Another study observed long-term incidences (2 years or more after diagnosis) of DIVI, and the authors reported that bleomycin–etoposide–cisplatin (BEP) has been associated with coronary artery diseases (CAD), a 4.3-fold higher risk in multivariate analysis, and with peripheral artery disease (PAD), a 4.1-fold higher risk in multivariate analysis [8]. Cyclophosphamides often damage pulmonary endothelial, mesothelial, and lymphatic endothelial cells and affect serous tissue of the pericardial sac, leading to pericarditis and pericardial effusion [9].

Topoisomerase inhibitors

Although anthracycline effects are fully characterized in the development of cardiomyopathy, their vascular side effects are not deeply understood. Anthracyclines cause late-onset and dose-dependent cardiomyopathy by overloading the reactive oxygen species (ROS), sarco/endoplasmic reticulum (SERCA) dysfunction, and inactivation of cytoprotective pathways [10]. Furthermore, impaired endothelial-myocyte coupling was proposed as a possible mechanism of cardiotoxicity. Endothelial neuregulin-1 acts protectively and maintains endothelial-myocyte coupling, in vitro [11]. Direct DIVI and acute vascular events with anthracyclines are less frequently reported. An in vitro study emphasized that epirubicin treatment interferes with intracellular cAMP levels and lipid-peroxidation, and this results in endothelial injury [12]. Caspase-3-mediated apoptosis has also been observed in endothelial cells, in vitro. Small animal models have demonstrated microthrombus formation in coronary arterioles after doxorubicin treatment [13].

Anti-microtubule agents

Taxanes and vinca alkaloids target microtubules, suppress microtubule dynamics, and block proliferation through cell cycle arrest [14]. Furthermore, they induce tumor cell death by anti-angiogenic effects. Multiple cases have reported taxane-induced angina pectoris, myocardial ischemia, and myocardial infarction [14, 15]. In these reports, vasospasm was suggested as a potential mechanism behind these ischemic events. Enhanced TNF-α-induced tissue factor expression and downregulation of thrombomodulin were suggested as possible pathologic steps [11]. Other cardiovascular side effects of taxanes comprise sinus node dysfunction, bradycardia, and conduction abnormalities, mainly atrioventricular block [15]. Second-generation vinca alkaloid vinorelbine causes local toxicities (irritation, pain, phlebitis) in 30% of patients [16]. Vascular injury develops due to increased ROS production and depleted glutathione levels. In vitro studies have addressed these mechanisms and showed the advantages of glutathione and N-acetylcysteine substitution without clear clinical evidence of benefit [17].

Anti-metabolites

Pyrimidine analogues, mainly 5-fluorouracil (5-FU) and its oral form capecitabine, cause cardiac toxicity in 120% of patients. Most DIVI present as angina, hypotension, myocardial infarction, cardiogenic shock, and heart failure due to acute ischemic events [18]. A recent prospective study of 644 patients reported a low (1%) incidence for both myocardial ischemia and infarction [19]; however, silent (asymptomatic) ischemia was reported in 56% of the patients. Large-scale factors are responsible including coronary vasospasm, direct endothelial toxicity, increased oxidative stress, and altered rheology [20]. Gemcitabine causes peripheral ischemia and Raynaud’s phenomenon in resistance arteries [21]. Furthermore, Takotsubo cardiomyopathy and microvascular angiopathy were associated with capecitabine administration [22]. The anti-folate drug methotrexate (MTX) is known to cause vascular and endothelial injury [23]. During MTX treatment, the availability of tetrahydrofolate reductase leads to hyperhomocysteinemia, a risk factor for vascular events [23]. Concomitant folic acid (leucovorin) supplementation normalizes plasma homocysteine levels during MTX treatment [23].

Targeted therapeutics

Monoclonal antibodies may cause systemic side effects by significant cytokine release upon administration [24]. These range from mild erythema to severe bronchospasm and anaphylactic shock [25]. VEGF signaling pathway inhibitors have an increased risk for arterial thrombosis as they decrease endothelial cell survival and proliferation [1, 26]. The anti-VEGF-A monoclonal antibody bevacizumab, trastuzumab, and tyrosine kinase inhibitor (TKI) sorafenib cause vasospasm and arterial thrombosis in 2.6% and 1.4% of patients, respectively [27, 28]. Clinically, the ATEE presented as myocardial infarction or cerebral ischemic attacks [27, 28]. A decrease in nitric oxide (NO) synthase activity, inhibition of the PDGF receptor, and consequent rarefaction of pericytes are suggested to be involved in sorafenib toxicity [11, 28]. Of note, a group of anti-VEGFR-TKIs: sunitinib, sorafenib, pazopanib, and vandetanib were shown not to increase the risk of DIVI [29].
Hypertension is a common side effect of VEGF-inhibitors with an incidence above 20%. Hypertensive response can occur from initiation (within hours) until 1 year after treatment [29]. Proposed mechanisms include NO pathway inhibition, reduced microvascular capacity and flow, oxidative stress, and glomerular injury [28, 29]. BCR-ABL1 TKIs have unique vascular safety profiles, which differ from other TKIs. The first-generation BCR-ABL1 inhibitor, imatinib, has been described as a safe drug regarding cardiovascular toxicities. The second-generation drug dasatinib is primarily associated with pulmonary arterial hypertension, and nilotinib is associated with increased risk of peripheral arterial disease, myocardial and cerebrovascular ischemia, and metabolic changes (hyperglycemia and dyslipidemia). The third-generation drug ponatinib is associated with hypertension, and ATEE [29]. Endothelial damage leading to pro-atherogenic and anti-angiogenic effects are the suggested pathomechanisms [2729] (Table 1).

Hormonal therapies

Aromatase inhibitors and tamoxifen are used in post-menopausal hormone receptor-positive breast cancer treatment. The use of aromatase inhibitors has increased the relative risk of myocardial infarction and other vascular side effects compared to tamoxifen [30, 31]. Aromatase inhibitor therapy has also been associated with increased prevalence of hypercholesterolemia and hypertension [31, 32]. Tamoxifen has been associated with a higher risk of ATEE compared to aromatase inhibitors [31].

Immune checkpoint inhibitors

Monoclonal antibodies target immune checkpoints to facilitate natural killer T cells and other innate immunity-related anti-tumor responses. These checkpoint inhibitors may lead to severe vascular side effects which mainly comprise inflammatory responses, mesothelial, and lymphatic endothelial injury: vasculitis, pericarditis, and myocarditis [33]. Ipilimumab is a CTLA-4 inhibitor which targets cytotoxic T cells. Side effects, such as diffuse rash and itching, can relate to microangiopathy [34]. Thalidomide and lenalidomide are immunomodulatory agents that reduce the level of TNF-α and consequently have anti-angiogenic activity and also inhibit leukocyte migration. They are used in the treatment of myeloma multiplex [35]. The anti-tumor anti-biotic bleomycin is known to cause limb ischemia and Raynaud’s phenomenon after the administration of the first dose [36, 37].

Risk assessment

The European Society of Cardiology (ESC) published a consensus paper to evaluate the cardiovascular risk of patients receiving anti-tumor treatment [38]. The ESC position paper is comprised of a cardiovascular risk assessment, which is recommended by the American Society of Clinical Oncology (ASCO), the Canadian Cardiovascular Society, and the European Society of Medical Oncology (ESMO) [39, 40]. However, the Khorana score is a recommended tool to evaluate the risk of venous thrombosis, no risk score is designed to assess the risk of ATEE [41]. Limitations of the Khorana score include the presence of platinum and pyrimidine analogue therapy, where risk stratification was not sensitive enough in the early stages of tumors. Therefore, the initiative of a “vascular fingerprint,” a risk calculation score, has been suggested to detect risk of adverse arterial events among patients with anti-tumor therapy (Table 2) [10, 12, 42]. Cisplatin treatment, type and stage of primary malignancy, age, presence of cardiovascular diseases, elevated LDH, retroperitoneal metastatic lymph nodes in germ cell tumors, and high Khorana score, but not pre-chemotherapy platelet number and BMI, are independent risk factors for DIVI [8].
Table 2
Complex risk stratification for arterial thromboembolic events [10, 12, 42]
Khorana score
Cancer type, testicular
Body mass index (BMI) > 25 kg/m2
Pre-chemo platelet count > 350 ×  109/L
Hemoglobin level < 10 g/dL
Pre-chemo leukocyte count ≥ 11 ×  109/L
Vascular fingerprint
BMI > 25 kg/m2
Current smoking
Blood pressure > 140/90 mm Hg (or use of anti-hypertensive drugs)
Dyslipidemia (total cholesterol > 5.1 mmol/L, LDL > 2.5 mmol/L), or using lipid-lowering drugs)
Fasting glucose ≥ 7 mmol/L (or using blood glucose lowering drugs)
Independent risk factors
Cisplatin treatment
Testicular cancer
Age
Presence of CVD
Elevated LDH (333 U/L, ULN 451 U/L)
Retroperitoneal metastatic lymph nodes

Prevention and treatment

In accordance with the latest ASCO guidelines, the ESC does not recommend routine prophylactic treatment for preventing ATEE among cancer patients [38, 39]. Low-molecular-weight heparin (LMWH) prophylaxis may be proposed individually for high-risk patients (Khorana Score ≥ 3). However, Moore et al. suggest that all patients treated with cisplatin, including patients with a low Khorana score, should receive prophylaxis, if bleeding risk is not elevated [6]. In a retrospective study involving germ cell tumor, patients treated with only cisplatin, prophylactic LMWH treatment, halved the number of venous DIVI [43]. The greatest reduction of ATEE by nadroparin versus placebo was shown in patients receiving either cisplatin, carboplatin, or gemcitabine alone or in combination [4446]. Multiple myeloma patients receiving thalidomide or lenalidomide combined with chemotherapy or dexamethasone should be treated with low-dose acetylsalicylate (low risk) or LMWH (high risk) [35]. According to the findings of the PROTECHT trial, nadroparin reduced the incidence of DIVI by nearly 50% among patients with advanced oncological diseases [47]. Clinical trials are underway to investigate different preventive strategies. Factor Xa inhibitor apixaban vs placebo (NCT02048865), low-dose acetylsalicylate vs simvastatin vs placebo (NCT02285738), and enoxaparin vs acetylsalicylate (NCT01763606) are being evaluated.
The use of nitrates and/or calcium channel inhibitors can be considered as a possible regular treatment in the instance of coronary vasospasm (ECG changes or angiography). Close follow-up and anti-platelet drugs should be considered in asymptomatic PAD cancer patients. In the case of severe PAD at baseline or during cancer therapy, revascularization strategy should be evaluated and discussed by a multidisciplinary team. ACE inhibitors and beta-receptor blockers should also be administered to prevent or reverse myocardial and vascular remodeling [38].
Given the short of clinical evidence in prevention and treatment of ATEE, large registries are urgently needed to better understand vascular side effects and therapies with mortality benefit. Additionally, extensive in vitro studies are warranted to model pathomechanisms of arterial DIVI, such as altered regulation of vascular tone, disturbances in endothelial inflammatory responses, barrier function, and hemostasis.

Conclusion and outlook

Mortality rates of cancer patients have had noticeable decreases due to the latest anti-tumor drugs and innovative surgical techniques. In parallel, the number of treatment-related side effects is increasing. Various studies and case reports have been focusing on cardiovascular toxicities induced by cisplatin and other chemotherapeutic drugs. The risk of incident myocardial infarction was significantly increased after cisplatin-based treatment, hazard ratio 3.1 vs control group [48]. Significantly, a large study involving 15,006 patients with testicular non-seminoma cancer quantified the short-term risk of death due to DIVI. Surprisingly, a 4.8-fold increased risk of death was reported within 1 year of chemotherapy (mostly BEP) compared to the control group (non-seminoma patients treated with surgery only) [45, 49]. Chemotherapeutic drugs have diverse mechanisms of action and extensive clinical presentations of major vascular adverse events.
Clinical scenarios are frequently complicated by radiation-induced side effects which mainly occur during long-term follow-up [50]. Radiation itself rarely causes ATEE, but rather generates increased arterial stiffness, hypertension, and fibrosis of myocardial and connective tissues. Considering the high number of ATEE among cancer patients, more sensitive and earlier risk stratification tools are warranted. We suggest developing and implementing a combined score including the Grace Score, Khorana score, and vascular fingertip score.
To put this into clinical practice, once the combined risk for ATEE exceeds the risk of bleeding, low-dose anti-coagulation would be recommended, preferably with unfractionated heparin. This can be safely tailored if patient condition deteriorates, bleeding develops, or surgery proceeds. Prophylaxis may be administered during all chemo-cycles and 5–10 days post-chemo. The risk stratification strategy, followed by an aggressive treatment of modifiable risk factors, should be introduced in conjunction with planning oncology treatment. Treatment of modifiable risk factors comprises quitting smoking, healthy diet, lipid lowering, blood pressure-lowering, and anti-diabetic medications. Despite the vast amount of research and clinical focus, there is still an urgent need to develop specific guidelines on the prevention and management of ATEE. Specifically, individual and sensitive risk stratification is warranted. Guidelines may include the localized site of primary tumor and drug-specific recommendations to predict risk and prevent ATEE.

Acknowledgements

Open access funding provided by Semmelweis University (SE).

Compliance with ethical standards

Conflict of interest

The authors declare that they have no conflict of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Publisher's Note

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

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Cameron AC, Touyz RM, Lang NN. Vascular complications of cancer chemotherapy. Can J Cardiol. 2016;32:852–62.CrossRef Cameron AC, Touyz RM, Lang NN. Vascular complications of cancer chemotherapy. Can J Cardiol. 2016;32:852–62.CrossRef
2.
Zurück zum Zitat Hayes-Lattin B, Nichols CR. Testicular cancer: a prototypic tumor of young adults. Semin Oncol. 2009;36:432–8.CrossRef Hayes-Lattin B, Nichols CR. Testicular cancer: a prototypic tumor of young adults. Semin Oncol. 2009;36:432–8.CrossRef
3.
Zurück zum Zitat Bellinger AM, Arteaga CL, Force T, Humphreys BD, Demetri GD, Druker BJ, et al. Cardio-oncology. Circulation. 2015;132:2248–58.CrossRef Bellinger AM, Arteaga CL, Force T, Humphreys BD, Demetri GD, Druker BJ, et al. Cardio-oncology. Circulation. 2015;132:2248–58.CrossRef
4.
Zurück zum Zitat Dieckmann KP, Struss WJ, Budde U. Evidence for acute vascular toxicity of cisplatin-based chemotherapy in patients with germ cell tumour. Anticancer Res. 2011;31:4501–5.PubMed Dieckmann KP, Struss WJ, Budde U. Evidence for acute vascular toxicity of cisplatin-based chemotherapy in patients with germ cell tumour. Anticancer Res. 2011;31:4501–5.PubMed
5.
Zurück zum Zitat Dieckmann K-P, Gerl A, Witt J, Hartmann J-T. Myocardial infarction and other major vascular events during chemotherapy for testicular cancer. Ann Oncol. 2010;21:1607–11.CrossRef Dieckmann K-P, Gerl A, Witt J, Hartmann J-T. Myocardial infarction and other major vascular events during chemotherapy for testicular cancer. Ann Oncol. 2010;21:1607–11.CrossRef
6.
Zurück zum Zitat Moore RA, Adel N, Riedel E, Bhutani M, Feldman DR, Tabbara NE, et al. High incidence of thromboembolic events in patients treated with cisplatin-based chemotherapy: a large retrospective analysis. J Clin Oncol. 2011;29:3466–73.CrossRef Moore RA, Adel N, Riedel E, Bhutani M, Feldman DR, Tabbara NE, et al. High incidence of thromboembolic events in patients treated with cisplatin-based chemotherapy: a large retrospective analysis. J Clin Oncol. 2011;29:3466–73.CrossRef
7.
Zurück zum Zitat de Haas EC, Altena R, Boezen HM, Zwart N, Smit AJ, Bakker SJL, et al. Early development of the metabolic syndrome after chemotherapy for testicular cancer. Ann Oncol. 2013;24:749–55.CrossRef de Haas EC, Altena R, Boezen HM, Zwart N, Smit AJ, Bakker SJL, et al. Early development of the metabolic syndrome after chemotherapy for testicular cancer. Ann Oncol. 2013;24:749–55.CrossRef
8.
Zurück zum Zitat Haugnes HS, Wethal T, Aass N, Dahl O, Klepp O, Langberg CW, et al. Cardiovascular risk factors and morbidity in long-term survivors of testicular cancer: a 20-year follow-up study. J Clin Oncol. 2010;28:4649–57.CrossRef Haugnes HS, Wethal T, Aass N, Dahl O, Klepp O, Langberg CW, et al. Cardiovascular risk factors and morbidity in long-term survivors of testicular cancer: a 20-year follow-up study. J Clin Oncol. 2010;28:4649–57.CrossRef
9.
Zurück zum Zitat Kachel DL, Martin WJ. Cyclophosphamide-induced lung toxicity: mechanism of endothelial cell injury. J Pharmacol Exp Ther. 1994;268:42–6.PubMed Kachel DL, Martin WJ. Cyclophosphamide-induced lung toxicity: mechanism of endothelial cell injury. J Pharmacol Exp Ther. 1994;268:42–6.PubMed
10.
Zurück zum Zitat Renu K, Abilash VG, Tirupathi TP, Arunachalam S. Molecular mechanism of doxorubicin-induced cardiomyopathy—An update. Eur J Pharmacol. 2018;818:241–53.CrossRef Renu K, Abilash VG, Tirupathi TP, Arunachalam S. Molecular mechanism of doxorubicin-induced cardiomyopathy—An update. Eur J Pharmacol. 2018;818:241–53.CrossRef
11.
Zurück zum Zitat Wood SC, Tang X, Tesfamariam B. Paclitaxel potentiates inflammatory cytokine-induced prothrombotic molecules in endothelial cells. J Cardiovasc Pharmacol. 2010;55:276–85.CrossRef Wood SC, Tang X, Tesfamariam B. Paclitaxel potentiates inflammatory cytokine-induced prothrombotic molecules in endothelial cells. J Cardiovasc Pharmacol. 2010;55:276–85.CrossRef
12.
Zurück zum Zitat Herrmann J, Yang EH, Iliescu CA, Cilingiroglu M, Charitakis K, Hakeem A, et al. Vascular toxicities of cancer therapies: the old and the new - an evolving avenue. Circulation. 2016;133:1272–89.CrossRef Herrmann J, Yang EH, Iliescu CA, Cilingiroglu M, Charitakis K, Hakeem A, et al. Vascular toxicities of cancer therapies: the old and the new - an evolving avenue. Circulation. 2016;133:1272–89.CrossRef
13.
Zurück zum Zitat Ben Aharon I, Bar Joseph H, Tzabari M, Shenkman B, Farzam N, Levi M, et al. Doxorubicin-induced vascular toxicity - targeting potential pathways may reduce procoagulant activity. PLoS ONE. 2013;20:8. Ben Aharon I, Bar Joseph H, Tzabari M, Shenkman B, Farzam N, Levi M, et al. Doxorubicin-induced vascular toxicity - targeting potential pathways may reduce procoagulant activity. PLoS ONE. 2013;20:8.
14.
Zurück zum Zitat Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer. 2004;4:253–65.CrossRef Jordan MA, Wilson L. Microtubules as a target for anticancer drugs. Nat Rev Cancer. 2004;4:253–65.CrossRef
15.
Zurück zum Zitat Rowinsky EK, McGuire WP, Guarnieri T, Fisherman JS, Christian MC, Donehower RC. Cardiac disturbances during the administration of taxol. J Clin Oncol. 1991;9:1704–12.CrossRef Rowinsky EK, McGuire WP, Guarnieri T, Fisherman JS, Christian MC, Donehower RC. Cardiac disturbances during the administration of taxol. J Clin Oncol. 1991;9:1704–12.CrossRef
16.
Zurück zum Zitat Yoh K, Niho S, Goto K, Ohmatsu H, Kubota K, Kakinuma R, et al. Randomized trial of drip infusion versus bolus injection of vinorelbine for the control of local venous toxicity. Lung Cancer. 2007;55:337–41.CrossRef Yoh K, Niho S, Goto K, Ohmatsu H, Kubota K, Kakinuma R, et al. Randomized trial of drip infusion versus bolus injection of vinorelbine for the control of local venous toxicity. Lung Cancer. 2007;55:337–41.CrossRef
17.
Zurück zum Zitat Yamada T, Egashira N, Imuta M, Yano T, Yamauchi Y, Watanabe H, et al. Role of oxidative stress in vinorelbine-induced vascular endothelial cell injury. Free Radic Biol Med. 2010;48:120–7.CrossRef Yamada T, Egashira N, Imuta M, Yano T, Yamauchi Y, Watanabe H, et al. Role of oxidative stress in vinorelbine-induced vascular endothelial cell injury. Free Radic Biol Med. 2010;48:120–7.CrossRef
18.
Zurück zum Zitat Polk A, Vaage-Nilsen M, Vistisen K, Nielsen DL. Cardiotoxicity in cancer patients treated with 5-fluorouracil or capecitabine: a systematic review of incidence, manifestations and predisposing factors. Cancer Treat Rev. 2013;39:974–84.CrossRef Polk A, Vaage-Nilsen M, Vistisen K, Nielsen DL. Cardiotoxicity in cancer patients treated with 5-fluorouracil or capecitabine: a systematic review of incidence, manifestations and predisposing factors. Cancer Treat Rev. 2013;39:974–84.CrossRef
19.
Zurück zum Zitat Kosmas C, Kallistratos MS, Kopterides P, Syrios J, Skopelitis H, Mylonakis N, et al. Cardiotoxicity of fluoropyrimidines in different schedules of administration: a prospective study. J Cancer Res Clin Oncol. 2008;134:75–82.CrossRef Kosmas C, Kallistratos MS, Kopterides P, Syrios J, Skopelitis H, Mylonakis N, et al. Cardiotoxicity of fluoropyrimidines in different schedules of administration: a prospective study. J Cancer Res Clin Oncol. 2008;134:75–82.CrossRef
20.
Zurück zum Zitat Polk A, Vistisen K, Vaage-Nilsen M, Nielsen DL. A systematic review of the pathophysiology of 5-fluorouracil-induced cardiotoxicity. BMC Pharmacol Toxicol. 2014;4:15–47. Polk A, Vistisen K, Vaage-Nilsen M, Nielsen DL. A systematic review of the pathophysiology of 5-fluorouracil-induced cardiotoxicity. BMC Pharmacol Toxicol. 2014;4:15–47.
21.
Zurück zum Zitat Dasanu CA. Gemcitabine: vascular toxicity and prothrombotic potential. Expert Opin Drug Saf. 2008;7:703–16.CrossRef Dasanu CA. Gemcitabine: vascular toxicity and prothrombotic potential. Expert Opin Drug Saf. 2008;7:703–16.CrossRef
22.
Zurück zum Zitat Qasem A, Abdulhak AAB, Aly A, Moormeier J. Capecitabine-induced Takotsubo cardiomyopathy: a case report and literature review. Am J Ther. 2014;23:1188–92.CrossRef Qasem A, Abdulhak AAB, Aly A, Moormeier J. Capecitabine-induced Takotsubo cardiomyopathy: a case report and literature review. Am J Ther. 2014;23:1188–92.CrossRef
23.
Zurück zum Zitat Howard SC, McCormick J, Pui C-H, Buddington RK, Harvey RD. Preventing and managing toxicities of high-dose methotrexate. Oncologist. 2016;12:1–12. Howard SC, McCormick J, Pui C-H, Buddington RK, Harvey RD. Preventing and managing toxicities of high-dose methotrexate. Oncologist. 2016;12:1–12.
24.
Zurück zum Zitat Hansel TT, Kropshofer H, Singer T, Mitchell JA, George AJT. The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov. 2010;9:325–38.CrossRef Hansel TT, Kropshofer H, Singer T, Mitchell JA, George AJT. The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov. 2010;9:325–38.CrossRef
25.
Zurück zum Zitat Baldo BA. Adverse events to monoclonal antibodies used for cancer therapy focus on hypersensitivity responses. Oncoimmunology. 2013;10:2. Baldo BA. Adverse events to monoclonal antibodies used for cancer therapy focus on hypersensitivity responses. Oncoimmunology. 2013;10:2.
26.
Zurück zum Zitat Kamba T, McDonald DM. Mechanisms of adverse effects of anti-VEGF therapy for cancer. Br J Cancer. 2007;96:1788–95.CrossRef Kamba T, McDonald DM. Mechanisms of adverse effects of anti-VEGF therapy for cancer. Br J Cancer. 2007;96:1788–95.CrossRef
27.
Zurück zum Zitat Choueiri TK, Schutz FAB, Je Y, Rosenberg JE, Bellmunt J. Risk of arterial thromboembolic events with sunitinib and sorafenib: a systematic review and meta-analysis of clinical trials. J Clin Oncol. 2010;28:2280–5.CrossRef Choueiri TK, Schutz FAB, Je Y, Rosenberg JE, Bellmunt J. Risk of arterial thromboembolic events with sunitinib and sorafenib: a systematic review and meta-analysis of clinical trials. J Clin Oncol. 2010;28:2280–5.CrossRef
28.
Zurück zum Zitat Qi WX, Shen Z, Tang LN, Yao Y. Bevacizumab increases the risk of gastrointestinal perforation in cancer patients: a meta-analysis with a focus on different subgroups. Eur J Clin Pharmacol. 2014;70:893–906.CrossRef Qi WX, Shen Z, Tang LN, Yao Y. Bevacizumab increases the risk of gastrointestinal perforation in cancer patients: a meta-analysis with a focus on different subgroups. Eur J Clin Pharmacol. 2014;70:893–906.CrossRef
29.
Zurück zum Zitat Moslehi JJ, Deininger M. Tyrosine kinase inhibitor-associated cardiovascular toxicity in chronic myeloid leukemia. J Clin Oncol. 2015;10:4210–8.CrossRef Moslehi JJ, Deininger M. Tyrosine kinase inhibitor-associated cardiovascular toxicity in chronic myeloid leukemia. J Clin Oncol. 2015;10:4210–8.CrossRef
30.
Zurück zum Zitat Amir E, Seruga B, Niraula S, Carlsson L, Ocaña A. Toxicity of adjuvant endocrine therapy in postmenopausal breast cancer patients: a systematic review and meta-analysis. J Natl Cancer Inst. 2011;103:1299–309.CrossRef Amir E, Seruga B, Niraula S, Carlsson L, Ocaña A. Toxicity of adjuvant endocrine therapy in postmenopausal breast cancer patients: a systematic review and meta-analysis. J Natl Cancer Inst. 2011;103:1299–309.CrossRef
31.
Zurück zum Zitat Burstein HJ, Prestrud AA, Seidenfeld J, Anderson H, Buchholz TA, Davidson NE, et al. American Society of Clinical Oncology clinical practice guideline: update on adjuvant endocrine therapy for women with hormone receptor-positive breast cancer. J Clin Oncol. 2010;4:3784–96.CrossRef Burstein HJ, Prestrud AA, Seidenfeld J, Anderson H, Buchholz TA, Davidson NE, et al. American Society of Clinical Oncology clinical practice guideline: update on adjuvant endocrine therapy for women with hormone receptor-positive breast cancer. J Clin Oncol. 2010;4:3784–96.CrossRef
32.
Zurück zum Zitat Abdel-Qadir H, Amir E, Fischer HD, Fu L, Austin PC, Harvey PJ, et al. The risk of myocardial infarction with aromatase inhibitors relative to tamoxifen in post-menopausal women with early stage breast cancer. Eur J Cancer. 2016;68:11–21.CrossRef Abdel-Qadir H, Amir E, Fischer HD, Fu L, Austin PC, Harvey PJ, et al. The risk of myocardial infarction with aromatase inhibitors relative to tamoxifen in post-menopausal women with early stage breast cancer. Eur J Cancer. 2016;68:11–21.CrossRef
33.
Zurück zum Zitat Varricchi G, Marone G, Mercurio V, Bonaduce D, Tocchetti CG. Immune checkpoint inhibitors and cardiac toxicity: an emerging issue. Curr Med Chem. 2017;25(11):1327–39.CrossRef Varricchi G, Marone G, Mercurio V, Bonaduce D, Tocchetti CG. Immune checkpoint inhibitors and cardiac toxicity: an emerging issue. Curr Med Chem. 2017;25(11):1327–39.CrossRef
34.
Zurück zum Zitat Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;15:50. Darvin P, Toor SM, Sasidharan Nair V, Elkord E. Immune checkpoint inhibitors: recent progress and potential biomarkers. Exp Mol Med. 2018;15:50.
35.
Zurück zum Zitat Palumbo A, Rajkumar SV, Dimopoulos MA, Richardson PG, San Miguel J, Barlogie B, et al. Prevention of thalidomide- and lenalidomide-associated thrombosis in myeloma. Leukemia. 2008;22:414–23.CrossRef Palumbo A, Rajkumar SV, Dimopoulos MA, Richardson PG, San Miguel J, Barlogie B, et al. Prevention of thalidomide- and lenalidomide-associated thrombosis in myeloma. Leukemia. 2008;22:414–23.CrossRef
36.
Zurück zum Zitat McGrath SE, Webb A, Walker-Bone K. Bleomycin-induced Raynaud’s phenomenon after single-dose exposure: risk factors and treatment with intravenous iloprost infusion. J Clin Oncol. 2013;31(4):e51–2.CrossRef McGrath SE, Webb A, Walker-Bone K. Bleomycin-induced Raynaud’s phenomenon after single-dose exposure: risk factors and treatment with intravenous iloprost infusion. J Clin Oncol. 2013;31(4):e51–2.CrossRef
37.
Zurück zum Zitat Grünwald V, Bolte O, Wiebe S, Ganser A, Schöffski P. Acral necrosis after inadequate excessive administration of bleomycin in a testicular cancer patient. Onkologie. 2005;28(1):41–3.PubMed Grünwald V, Bolte O, Wiebe S, Ganser A, Schöffski P. Acral necrosis after inadequate excessive administration of bleomycin in a testicular cancer patient. Onkologie. 2005;28(1):41–3.PubMed
38.
Zurück zum Zitat Zamorano JL, Lancellotti P, Rodriguez Muñoz D, Aboyans V, Asteggiano R, Galderisi M, et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines. Eur Heart J. 2016;74:2768–801.CrossRef Zamorano JL, Lancellotti P, Rodriguez Muñoz D, Aboyans V, Asteggiano R, Galderisi M, et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines. Eur Heart J. 2016;74:2768–801.CrossRef
39.
Zurück zum Zitat Lyman GH, Khorana AA, Kuderer NM, Lee AY, Arcelus JI, Balaban EP, et al. Venous thromboembolism prophylaxis and treatment in patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol. 2013;31:2189–204.CrossRef Lyman GH, Khorana AA, Kuderer NM, Lee AY, Arcelus JI, Balaban EP, et al. Venous thromboembolism prophylaxis and treatment in patients with cancer: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol. 2013;31:2189–204.CrossRef
40.
Zurück zum Zitat Virani SA, Dent S, Brezden-Masley C, Clarke B, Davis MK, Jassal DS, et al. Canadian Cardiovascular Society guidelines for evaluation and management of cardiovascular complications of cancer therapy. Can J Cardiol. 2016;32:831–41.CrossRef Virani SA, Dent S, Brezden-Masley C, Clarke B, Davis MK, Jassal DS, et al. Canadian Cardiovascular Society guidelines for evaluation and management of cardiovascular complications of cancer therapy. Can J Cardiol. 2016;32:831–41.CrossRef
41.
Zurück zum Zitat Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis CW. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood. 2008;111:4902–7.CrossRef Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis CW. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood. 2008;111:4902–7.CrossRef
42.
Zurück zum Zitat Lubberts S, Boer H, Altena R, Meijer C, Van Roon AM, Zwart N, et al. Vascular fingerprint and vascular damage markers associated with vascular events in testicular cancer patients during and after chemotherapy. Eur J Cancer. 2016;63:180–8.CrossRef Lubberts S, Boer H, Altena R, Meijer C, Van Roon AM, Zwart N, et al. Vascular fingerprint and vascular damage markers associated with vascular events in testicular cancer patients during and after chemotherapy. Eur J Cancer. 2016;63:180–8.CrossRef
43.
Zurück zum Zitat Gizzi M, Oberic L, Massard C, Poterie A, Gwenael LT, Loriot Y, et al. Predicting and preventing thromboembolic events in patients receiving cisplatin-based chemotherapy for germ cell tumours. Eur J Cancer. 2016;69:151–7.CrossRef Gizzi M, Oberic L, Massard C, Poterie A, Gwenael LT, Loriot Y, et al. Predicting and preventing thromboembolic events in patients receiving cisplatin-based chemotherapy for germ cell tumours. Eur J Cancer. 2016;69:151–7.CrossRef
44.
Zurück zum Zitat Verso M, Agnelli G, Barni S, Gasparini G, LaBianca R. A modified Khorana risk assessment score for venous thromboembolism in cancer patients receiving chemotherapy: the Protecht score. Intern Emerg Med. 2012;7(3):291–2.CrossRef Verso M, Agnelli G, Barni S, Gasparini G, LaBianca R. A modified Khorana risk assessment score for venous thromboembolism in cancer patients receiving chemotherapy: the Protecht score. Intern Emerg Med. 2012;7(3):291–2.CrossRef
45.
Zurück zum Zitat Fung C, Fossa SD, Milano MT, Sahasrabudhe DM, Peterson DR, Travis LB. Cardiovascular disease mortality after chemotherapy or surgery for testicular nonseminoma: a population-based study. J Clin Oncol. 2015;33:3106–14.CrossRef Fung C, Fossa SD, Milano MT, Sahasrabudhe DM, Peterson DR, Travis LB. Cardiovascular disease mortality after chemotherapy or surgery for testicular nonseminoma: a population-based study. J Clin Oncol. 2015;33:3106–14.CrossRef
46.
Zurück zum Zitat Nuver J, Smit AJ, Van Der Meer J, Van Den Berg MP, Van Der Graaf WTA, Meinardi MT, et al. Acute chemotherapy-induced cardiovascular changes in patients with testicular cancer. J Clin Oncol. 2005;23:9130–7.CrossRef Nuver J, Smit AJ, Van Der Meer J, Van Den Berg MP, Van Der Graaf WTA, Meinardi MT, et al. Acute chemotherapy-induced cardiovascular changes in patients with testicular cancer. J Clin Oncol. 2005;23:9130–7.CrossRef
47.
Zurück zum Zitat Barni S, Labianca R, Agnelli G, Bonizzoni E, Verso M, Mandalà M, et al. Chemotherapy-associated thromboembolic risk in cancer outpatients and effect of nadroparin thromboprophylaxis: results of a retrospective analysis of the PROTECHT study. J Transl Med. 2011;9(1):79.CrossRef Barni S, Labianca R, Agnelli G, Bonizzoni E, Verso M, Mandalà M, et al. Chemotherapy-associated thromboembolic risk in cancer outpatients and effect of nadroparin thromboprophylaxis: results of a retrospective analysis of the PROTECHT study. J Transl Med. 2011;9(1):79.CrossRef
48.
Zurück zum Zitat Brouha ME, Bloemendal HJ, Kappelle LJ, Winter JB. Cerebral infarction and myocardial infarction due to cisplatin-containing chemotherapy. Ned Tijdschr Geneeskd. 2003;147:457–60.PubMed Brouha ME, Bloemendal HJ, Kappelle LJ, Winter JB. Cerebral infarction and myocardial infarction due to cisplatin-containing chemotherapy. Ned Tijdschr Geneeskd. 2003;147:457–60.PubMed
49.
Zurück zum Zitat Dieckmann KP, Gehrckens R. Thrombosis of abdominal aorta during cisplatin-based chemotherapy of testicular seminoma–a case report. BMC Cancer. 2009;9:459.CrossRef Dieckmann KP, Gehrckens R. Thrombosis of abdominal aorta during cisplatin-based chemotherapy of testicular seminoma–a case report. BMC Cancer. 2009;9:459.CrossRef
50.
Zurück zum Zitat Yusuf SW, Sami S, Daher IN. Radiation-induced heart disease: a clinical update. Cardiol Res Pract. 2011;2011:1–9. Yusuf SW, Sami S, Daher IN. Radiation-induced heart disease: a clinical update. Cardiol Res Pract. 2011;2011:1–9.
Metadaten
Titel
Anti-cancer drugs-induced arterial injury: risk stratification, prevention, and treatment
verfasst von
Edit Gara
Kristóf György Csikó
Zoltán Ruzsa
Gábor Földes
Béla Merkely
Publikationsdatum
01.08.2019
Verlag
Springer US
Erschienen in
Medical Oncology / Ausgabe 8/2019
Print ISSN: 1357-0560
Elektronische ISSN: 1559-131X
DOI
https://doi.org/10.1007/s12032-019-1295-8

Weitere Artikel der Ausgabe 8/2019

Medical Oncology 8/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.