Vascular Complications of Cancer and Cancer Therapy


Patient characteristics

Risk score

Site of cancer

  Very high risk (stomach, pancreas)

  High risk (lung, lymphoma, gynecologic, bladder, testicular)

2

1

Prechemotherapy platelet count 350,000/mm3 or more

1

Hemoglobin level less than 10 g/dL or use of red cell growth factors

1

Prechemotherapy leukocyte count more than 11,000/mm3

1

Body mass index 35 kg/m2 or more

1


High-risk score ≥3

Intermediate-risk score = 1–2

Low-risk score = 0





Consequences of Venous Thromboembolism in Patients with Cancer


VTE in patients with cancer is associated with several adverse consequences including early mortality [2, 2326]. Additional serious clinical complications include recurrent VTE, major bleeding associated with anticoagulation , and interruption of optimal cancer treatment along with an impact on quality of life and healthcare costs [27, 28]. Importantly, the risk of recurrence, bleeding, and mortality in cancer patients with incidental or unsuspected VTE appears to be similar to those with symptomatic VTE [29]. The majority of patients with unsuspected pulmonary embolism (PE) identified on staging computerized tomography scans are symptomatic with similar clinical consequences [20, 3032]. Clinical symptoms of chest pain, shortness of breath, and fatigue are often attributed to the underlying malignancy [20, 31, 32].


Prevention and Treatment of Cancer-Associated Venous Thromboembolism


There is experimental evidence that the heparins may interfere with cancer cell proliferation, angiogenesis, and the formation of metastases [33]. Several RCTs in patients with cancer but without VTE have evaluated whether anticoagulants improve overall survival with varied results [3440]. In a meta-analysis of 11 RCTs of patients with cancer receiving anticoagulants or no anticoagulants, a significant reduction in 1-year mortality with LMWHs but not with warfarin was observed with relative risks for all-cause mortality of 0.88 [95 % CI, 0.79–0.98; P = 0.015] and 0.94 [95 % CI, 0.85–1.04; P = 0.239], respectively [41]. Greater risk of major bleeding was reported in patients receiving anticoagulation reaching statistical significance in warfarin studies [41]. Therefore, while anticoagulation is not recommended in this situation due to the recognized limitations of these trials and the increased risk for bleeding, a number of clinical practice guidelines address the appropriate role of thromboprophylaxis in the treatment and preventions of VTE in patients with cancer [4246]. As summarized in Table 9.2, recommendations cover treatment and prevention of VTE in hospitalized medical and surgical cancer patients, the current limited role of prophylaxis in the ambulatory setting, and secondary prophylaxis of patients with established VTE. These guidelines also recommend that patients with cancer be educated about the symptoms and signs of VTE and that VTE risk be assessed at the time of chemotherapy initiation and periodically over the course of treatment.


Table 9.2
VTE treatment and prophylaxis recommendations [46, 124]



















ASCO recommendations

Inpatient

  1.1 Hospitalized patients who have active malignancy with acute medical illness or reduced mobility should receive pharmacologic thromboprophylaxis in the absence of bleeding or other contraindications

  1.2 Hospitalized patients who have active malignancy without additional risk factors may be considered for pharmacologic thromboprophylaxis in the absence of bleeding or other contraindications

  1.3 Data are inadequate to support routine thromboprophylaxis in patients admitted for minor procedures or brief infusional chemotherapy or in patients undergoing stem cell/bone marrow transplantation

Outpatient

  2.1 Routine pharmacologic thromboprophylaxis is not recommended in cancer outpatients

  2.2 Based on limited RCT data, clinicians may consider LMWH prophylaxis on a case-by-case basis in highly selected outpatients with solid tumors receiving chemotherapy. Consideration of such therapy should be accompanied by a discussion with the patient about the uncertainty concerning benefits and harms, as well as dose and duration of prophylaxis in this setting

  2.3 Patients with multiple myeloma receiving thalidomide- or lenalidomide-based regimens with chemotherapy and/or dexamethasone should receive pharmacologic thromboprophylaxis with either aspirin or LMWH for low-risk patients and LMWH for high-risk patients

Perioperative

  3.1 All patients with malignant disease undergoing major surgical intervention should be considered for pharmacologic thromboprophylaxis with either UFH or LMWH unless contraindicated because of active bleeding or a high risk of bleeding with the procedure

  3.2 Prophylaxis should be commenced preoperatively

  3.3 Mechanical methods may be added to pharmacologic thromboprophylaxis, but should not be used as monotherapy for VTE prevention unless pharmacologic methods are contraindicated because of active bleeding or high bleeding risk

  3.4 A combined regimen of pharmacologic and mechanical prophylaxis may improve efficacy, especially in the highest-risk patients

  3.5 Pharmacologic thromboprophylaxis should be continued for at least 7–10 days in all patients. Extended prophylaxis with LMWH for up to 4 weeks postoperatively should be considered for patients undergoing major abdominal or pelvic surgery for cancer who have high-risk features such as restricted mobility, obesity, history of VTE, or with additional risk factors

Treatment and secondary prophylaxis

  4.1 LMWH is preferred over UFH for the initial 5–10 days of anticoagulation for the cancer patient with newly diagnosed VTE who does not have severe renal impairment (defined as creatinine clearance <30 mL/min)

  4.2 For long-term anticoagulation, LMWH for at least 6 months is preferred due to improved efficacy over vitamin K antagonists. Vitamin K antagonists are an acceptable alternative for long-term therapy if LMWH is not available

  4.3 Anticoagulation with LMWH or vitamin K antagonist beyond the initial 6 months may be considered for select patients with active cancer, such as those with metastatic disease or those receiving chemotherapy

  4.4 The insertion of a vena cava filter is only indicated for patients with contraindications to anticoagulant therapy. It may be considered as an adjunct to anticoagulation in patients with progression of thrombosis (recurrent VTE or extension of existing thrombus) despite maximal therapy with LMWH

  4.5 For patients with central nervous system malignancies, anticoagulation is recommended for established VTE as described for other patients with cancer. Careful monitoring is necessary to limit the risk of hemorrhagic complications

  4.6 Use of novel oral anticoagulants for either prevention or treatment of VTE in cancer patients is not recommended at this time

  4.7 Incidental PE and DVT should be treated in the same manner as symptomatic VTE. Treatment of splanchnic or visceral vein thrombi diagnosed incidentally should be considered on a case-by-case basis, considering potential benefits and risks of anticoagulation

Anticoagulation and survival

  5.1 Anticoagulants are not recommended to improve survival in patients with cancer without VTE

  5.2 Patients with cancer should be encouraged to participate in clinical trials designed to evaluate anticoagulant therapy as an adjunct to standard anticancer therapies

Risk assessment

  6.1 Cancer patients should be assessed for VTE risk at the time of chemotherapy initiation and periodically thereafter

    6.1a In the outpatient setting, risk assessment can be conducted based on a validated risk assessment tool

    6.1b Solitary risk factors, including biomarkers or cancer site, do not reliably identify cancer patients at high risk of VTE

  6.2 Oncologists should educate patients regarding VTE, particularly in settings that increase risk such as major surgery, hospitalization, and while receiving systemic antineoplastic therapy. Patient education should at least include a discussion of the warning signs and symptoms of VTE, including leg swelling or pain, sudden-onset chest pain, and shortness of breath


Treatment of VTE in Patients with Cancer


The initial treatment of established VTE in cancer patients is generally patterned after therapeutic approaches in other non-cancer settings. However, the duration of therapy to prevent early recurrence is often extended in cancer patients with persistent disease or continuing on cancer treatment [47]. Current recommendations call for low molecular weight heparin (LMWH) for the initial 5–10 days of anticoagulation in cancer patients with established VTE, as well as for secondary prevention of recurrence for at least 6 months. Patients with unsuspected or incidental VTE should be treated the same as symptomatic VTE. High-risk patients on systemic therapy for persistent malignancy should be considered for extended anticoagulation to prevent VTE recurrence. The development of a number of new oral and parenteral antithrombotic agents is likely to have future application to patients with cancer [48, 49].


Thromboprophylaxis of Hospitalized Medical or Surgical Patients with Cancer


Although reported rates vary considerably, VTE is a common cause of death in hospitalized cancer patients [2, 4, 5053]. Although three large randomized controlled trials (RCTs) have demonstrated that thromboprophylaxis reduces VTE risk in hospitalized patients with acute medical illness, cancer represented only a small proportion of the study populations [5457]. Nevertheless, due to the increased risk of VTE associated with malignancy, prophylactic anticoagulation of most hospitalized patients with major medical illnesses including cancer or reduced mobility is recommended in the absence of a serious bleeding risk with anticoagulation.

Likewise, patients undergoing major cancer surgery are at an increased risk for VTE as well as for bleeding complications [58]. Patients undergoing major surgical procedures for cancer should receive thromboprophylaxis unless contraindicated, while combined mechanical prophylaxis and anticoagulation may be considered in high-risk patients [59]. Prophylactic anticoagulation should be initiated preoperatively when possible and continued for at least 7–10 days. Extended prophylaxis for up to 4 weeks postoperatively should also be considered in high-risk patients including those with restricted mobility, obesity, or a history of VTE. Of note, there remain inadequate data to support routine thromboprophylaxis in those with short admissions for chemotherapy or for minor procedures [46, 60].


Thromboprophylaxis of Ambulatory Patients with Cancer


The risk of VTE in ambulatory cancer patients varies widely with the type of cancer and treatment and associated comorbid conditions. As discussed further in this chapter, the emergence of more aggressive interventions and a number of new cancer therapies and supportive care agents with an increased risk of VTE has resulted in increased interest in the potential value of VTE prophylaxis in this setting [8, 6172]. Several RCTs of thromboprophylaxis in ambulatory cancer patients have been reported including nine with LMWHs. The greatest impact on the absolute risk of VTE has been observed in patients with advanced pancreatic cancer receiving specified chemotherapy [73, 74]. A meta-analysis has estimated a relative risk for symptomatic VTE across studies of 0.47 (0.36–0.61; P < 0.001) but with only an absolute reduction in VTE risk of 2.8 % (1.8–3.7 %; P < 0.001) [75]. Due to the small incremental benefit observed in most trials of ambulatory cancer patients, routine thromboprophylaxis is not recommended with the exception of patients with multiple myeloma receiving thalidomide or lenalidomide with chemotherapy and dexamethasone where the risk of VTE is very high. However, real-world studies in unselected ambulatory cancer patients receiving cancer chemotherapy have suggested rates of VTE twofold to threefold greater than those reported in selected patients in reported RCTs (Fig. 9.1) [76]. Therefore, thromboprophylaxis may be considered on an individual basis in selected high-risk patients with solid tumors receiving chemotherapy balancing the potential benefits and harms [46, 77].

A323316_1_En_9_Fig1_HTML.gif


Fig. 9.1
Cumulative risk for VTE in patients with cancer undergoing chemotherapy. VTE venous thromboembolism [76]


Vascular Complications of Endocrine Cancer Therapies


VTE is a recognized adverse event associated with estrogens as well as certain estrogen-like agents such as tamoxifen and other selective estrogen receptor modulating agents (SERMS) [7881]. Increased risk of VTE with these agents has been observed both in patients receiving endocrine treatment for cancer but also in those receiving SERMs as chemoprevention to reduce cancer risk [82]. Despite an apparent lower risk , an increased risk of VTE has also been associated with endocrine treatment with the aromatase inhibitors [83].


Vascular Complications Associated with Targeted Cancer Therapeutics


While several traditional cytotoxic chemotherapeutic agents including 5-fluorouracil and cisplatin have been associated with vascular toxicities such as coronary vasospasm and arterial thrombotic events [84, 85], newer targeted biological agents, including monoclonal antibodies and tyrosine kinase inhibitors, are also associated with a significant risk of vascular complications. In the following sections, we will provide an overview of these cardiovascular toxicities and specifically focus on the vascular complications of newer anti-VEGF (vascular endothelial growth factor) therapies, tyrosine kinase inhibitors, and immunomodulatory therapies.


Monoclonal Antibodies: VEGF Inhibitors


The relationship between vascular endothelial growth factor and tumor angiogenesis was first introduced in the 1970s by Dr. Judah Folkman, when an association between solid tumor growth and vascular supply was observed. He identified a soluble factor released from tumors (“tumor angiogenesis factor”), which promoted neovascularization, and he proposed that inhibition of this factor could halt tumor neo-angiogenesis [86]. Dr. Napoleone Ferrara’s group subsequently sequenced this factor, now known as VEGF, and since then this signaling pathway has been the target of multiple pharmacotherapies for the treatment of various malignancies [86]. Bevacizumab (Avastin™), a humanized monoclonal antibody that binds and neutralizes VEGF, was the first VEGF inhibitor approved by the FDA in 2004 after a landmark study demonstrated that the addition of bevacizumab to fluorouracil-based combination chemotherapy resulted in improved survival among patients with metastatic colorectal cancer [87]. It has since been approved as monotherapy for the treatment of advanced non-squamous non-small cell lung cancer, metastatic renal cell carcinoma, and recurrent glioblastoma [8688]. VEGF, while crucial to tumor angiogenesis, is also fundamental for endothelial cell function and proliferation and thus the formation of new vessels. As might be expected, VEGF inhibition has been associated with a significant increase in vascular complications, in particular arterial thrombotic events and bleeding [86, 87].


Mechanism of Action and Toxicity


VEGF is essential for tumor angiogenesis and increased VEGF expression is associated with increased tumor invasiveness, metastatic ability, and recurrence [87, 88]. There are three receptors (VEGFR-1/Flt-1, VEGFR-2/Flk-1/KDR, VEGFR-3/Flt-4). The ligand VEGF-A, generally referred to as VEGF, binds to VEGFR-2 on endothelial cells leading to pro-angiogenic effects [89]. By binding to the VEGF receptor, bevacizumab disrupts downstream VEGF signaling, preventing tumor angiogenesis and increasing the delivery of cancer therapy to tumor cells [90]. It is hypothesized that the vascular toxicities of bevacizumab, which include both thrombosis and bleeding, occur through similar mechanisms and are the result of an imbalance in the tightly regulated hemostatic system. This system includes a balance of pro- and anticoagulant proteins, platelet-activating and platelet-inhibiting factors, and pro- and antifibrinolytic products [12, 90]. Inhibition of VEGF results in decreased endothelial cell survival, platelet aggregation and thrombosis, increased platelet reactivity, and downregulation of several factors. The vasculature is susceptible to damage induced by trauma [12, 90], and disruption of the endothelial barrier results in exposure of the subendothelial von Willebrand factor and tissue factor, platelet aggregation, and thrombus formation [86, 91]. The increase in platelet-endothelial interactions may further precipitate thrombosis [92]. Finally, downregulation of several factors that are modulated by VEGF including nitric oxide, prostacyclin, and thrombolytic serine proteases (u-PA and t-PA) contributes to increased vascular toxicity such as vasoconstriction, endothelial cell apoptosis, and increased arterial stiffness [86, 90]. Several other mechanisms have been proposed as contributing to VEGF inhibitor-induced vascular toxicity. Drug-induced hypertension generating high shear stress at plaque sites may accelerate atherothrombosis [88, 91]. Associated inflammation and cell lysis have also been shown to increase thrombogenicity, and apoptotic cell blebs may sustain vascular inflammation and activate the complement cascade [88, 91]. Anti-angiogenic drugs are known to hinder the recognized insulin anti-atherogenic actions, including glucose uptake, lipogenesis, and antilipolysis, ultimately producing a thrombophilic hyperglycemic , atherogenic lipoprotein prone to thrombosis [9]. Finally, VEGF inhibition may result in lack of protective growth factors, potentially resulting in plaque instability and thrombosis [93]. Figure 9.2 illustrates the proposed mechanisms for the vascular toxic effects of bevacizumab.


Clinical Presentation and Epidemiology


The major cardiovascular toxicities of bevacizumab are hypertension, hemorrhage, perforation, and thrombosis [90]. Thrombotic events include thrombosis and thromboembolism, generally in the form of acute coronary syndrome, stroke, and peripheral vascular disease, although coronary ischemia is the most frequent arterial thrombotic event (ATE) [94]. While initial trials of bevacizumab only reported an insignificant increase in thrombotic or bleeding events, multiple subsequent larger clinical studies have demonstrated an elevated risk of vascular events [87]. A meta-analysis of 1745 patients from five randomized trials showed combination treatment with bevacizumab and chemotherapy, compared with chemotherapy alone, was associated with an increased risk of ATE (HR 2.0, 95 % CI 1.05–3.75, P = 0.031), but not venous thromboembolism [10]. A meta-analysis published in 2010 to evaluate the incidence of arterial thromboembolic events associated with bevacizumab included over 12,500 patients with various advanced solid tumors and found a 3.3 % incidence of ATE (95 % CI 2.0–5.6) and a 2.0 % incidence of high-grade arterial thrombotic events (95 % CI 1.7–2.5), defined as acute coronary syndrome, transient ischemic attack, stroke, life-threatening peripheral arterial thrombosis or requiring surgery, and death [94]. Compared to controls, bevacizumab was associated with a relative risk of ATE of 1.44 (95 % CI 1.08–1.91, p < 0.013). In particular, this meta-analysis found that bevacizumab was associated with a significantly increased risk of cardiac ischemia (RR 2.14, 95 % CI 1.12–4.08, p < 0.021) [94]. Vascular events have been noted early after starting VEGF inhibitor therapy, with a median time of event occurrence after drug initiation of 7 months (1–12 months is general range) [88, 95]. Moreover, the risk of ATE associated with bevacizumab may differ by tumor type. One meta-analysis found a significantly higher risk of high-grade ATE in patients with renal cell carcinoma (RR 5.14 95 % CI 1.35–19.64) [94]. Patients with pre-existing cardiovascular disease are at an increased high risk for thrombotic complications [96, 97]. Specifically, patients older than 65 years of age, with diabetes, with known atherosclerosis, or with a history of a prior cardiovascular event, appear to be at a higher risk for bevacizumab-associated ATE [97].


Management


Traditionally arterial thrombotic events have been associated with grave prognosis in cancer patients [98]. Once an arterial thrombotic event develops in a patient receiving VEGF inhibitor therapy, it is generally recommended therapy be permanently stopped and the ATE treated as per guidelines for the particular event, such as the American College of Cardiology/American Heart Association (ACC/AHA) recommendations for acute coronary events [99, 100]. Some clinicians advocate for avoiding anti-VEGF therapies in patients with a history of coronary or peripheral vascular disease. Others recommend prophylactic antiplatelet therapy such as aspirin or clopidogrel [86]. Aspirin therapy has been shown to be associated with a significant improvement in short-term cardiovascular outcomes [10, 88]. Theoretically, statins and angiotensin-converting-enzyme inhibitors (ACE-inhibitors) may also exert antioxidant and anti-inflammatory effects resulting in a decrease in atherothrombotic risk [10]. However, the use of anticoagulants and antiplatelet agents in patients receiving VEGF inhibitors remains controversial, especially in the setting of thromboembolic events given their association with bleeding.

Despite the increased frequency of thromboembolic events in cancer patients, there are limited data on the use of bevacizumab concurrently in patients being treated with therapeutic anticoagulation . A post hoc analysis of three phase III clinical trials of bevacizumab compared to control evaluated the incidence of thrombotic and bleeding in patients receiving therapeutic anticoagulation (warfarin and low molecular weight heparin) [95]. The incidence of thrombotic adverse events for patients receiving bevacizumab compared to the placebo group was primarily venous and on the order of 9.6–17.3 %. The overall rates of severe bleeding for all patients in the control group were 2.5 % versus 3.3 % in the bevacizumab group. The authors concluded that combining bevacizumab with therapeutic anticoagulation did not substantially increase the risk of bleeding beyond the risk of bleeding expected from therapeutic anticoagulation alone [95]. Similarly, a prospective observational cohort study including 1953 patients to assess the safety of bevacizumab for treatment of metastatic colorectal cancer (BRiTE study) showed serious bleeding events were comparable among patients on prophylactic anticoagulation therapy versus not [101]. In the absence of formal guidelines, it is generally recommended that patients be treated for thrombotic events with anticoagulation albeit cautiously with close monitoring for adverse bleeding events. There are insufficient data and therefore no standard recommendations to use prophylactic anticoagulation in this setting. Of course, an individual approach for patients should be conducted based on comorbidities and prior thrombotic and bleeding events. Furthermore, a cardiovascular risk assessment is recommended in all patients undergoing such therapy, with consideration for additional testing in certain patient populations.


Small Molecule Tyrosine Kinase Inhibitors: VEGF Inhibitors


Tyrosine kinase inhibitors (TKIs) are a growing and widely used group of therapies for various malignancies, and many TKIs have anti-angiogenic properties. Examples include, but are not limited to, sunitinib (Sutent™) , sorafenib (Nexavar™) , and pazopanib (Votrient™) . These agents, used widely in metastatic renal cell cancer, as well as for the treatment of other malignancies, have been associated with coronary ischemia and small vessel disease [12, 86].


Mechanisms of Action and Toxicity


Similar to bevacizumab, these small molecule TKIs work by blocking the intracellular domain of the VEGF receptor leading to inhibition of the VEGF pathway. Moreover, these TKIs affect multiple additional pathways. Sunitinib has activity against all three VEGFRs, platelet-derived growth factor receptor (PDGF) α and β, stem cell factor receptor (KIT), and fms-like kinase receptor 3 (FLT3) . It is approved for treatment of advanced renal cell carcinoma, gastrointestinal stromal tumors (GIST) , and advanced pancreatic neuroendocrine tumors [86, 88, 97]. Sorafenib has activity against all VEGFRs, PDGF-β, KIT, FLT3, and RET, as well as the intracellular kinases CRAF, BRAF, and mutant BRAF. It has been approved for the treatment of advanced hepatocellular carcinoma, thyroid cancer, and advanced renal cell carcinoma [86, 88, 97]. Pazopanib also has activity against VEGFRs, PDGFRs, fibroblast growth factor receptors (FGFRs) 1 and 3. It has been approved for the treatment of metastatic renal cell carcinoma and advanced soft tissue sarcomas that have received prior chemotherapy [86, 88, 97].

The mechanisms for vascular toxicity in these TKIs with VEGF pathway inhibiting characteristics may be similar to those proposed for bevacizumab. Individual response to VEGF inhibition and variation in response to the growth factor pathway blocking lends to variation in vascular complications and individual vulnerability to off-target effects [12, 86, 88, 97]. Figure 9.2 illustrates proposed mechanisms for vascular toxic effects of VEGF signaling pathway inhibitors.

A323316_1_En_9_Fig2_HTML.gif


Fig. 9.2
Proposed mechanisms for the “off-target” vascular toxicities from VEGF signaling pathway inhibitors [12, 86, 88, 9092, 97]


Clinical Presentation and Epidemiology


The major cardiac vascular toxicities of small molecule tyrosine kinase inhibitors with VEGF-inhibiting properties include hypertension, hemorrhage, perforation, and thrombosis [90]. As it relates to thrombosis , there is an increased incidence of acute coronary syndrome, cerebrovascular accidents, peripheral vascular disease, and bleeding observed with these anti-angiogenic TKIs.

Cardiac ischemia and related coronary artery disease has been a common manifestation of ATE associated with TKIs (Fig. 9.3). One observational study showed that among patients treated with sorafenib or sunitinib, there was an extremely high rate of cardiac events (33.8 %) leading to interruption or discontinuation of TKI therapy in 60 % of those patients [102]. Of the patients who experienced cardiac events, 52 % were symptomatic with angina, dyspnea, and dizziness while 48 % of the patients were asymptomatic with cardiac biomarker elevation or ECG changes [102]. In a large meta-analyses of over 38,000 patients evaluating the vascular complications of VEGF inhibitors, including both TKIs and bevacizumab , there was an increased risk of myocardial infarction , hypertension , and arterial thromboembolism [103]. Compared to the control group, recipients of VEGF inhibitors had significantly higher risk of myocardial infarction (RR 3.54, 95 % CI 1.61–7.80, I 2 = 0 %, tau2 = 0), arterial thrombotic events (RR 1.80, 95 % CI 1.24–2.59, I 2 = 0 %, tau2 = 0), and hypertension (RR 3.46, 95 % CI 2.89–4.15, I 2 = 58 %, tau2 = 0.16) [103]. In 2010, Choueiri et al. published a meta-analysis of clinical trials with sorafenib or sunitinib that included over 10,000 patients [91]. The rate of arterial thrombotic events was increased threefold (2 % absolute risk) with these drugs, and this finding was independent of type of malignancy or TKI [91]. Again, the most common type of ATE was coronary ischemia, followed by stroke [91].

A323316_1_En_9_Fig3_HTML.jpg


Fig. 9.3
Coronary angiogram of a 60-year-old male with metastatic renal cell cancer treated with multiple angiogenic inhibitors over a 9-year period. (a) Shows pre-percutaneous coronary intervention with occlusion of the left circumflex. (b) Shows post-percutaneous coronary intervention with restoration of flow in the left circumflex

Interestingly, a newer agent under investigation semaxanib, a potent and selective inhibitor of VEGFR-2/Flk-1/KDR, with activity against VEGFR-1, KIT, and FLT3, was aborted because of a remarkably high thrombosis rate of 42 % during a phase I trial [12, 88, 104]. The authors concluded that semaxanib likely causes endothelial cell activation, and in the setting of chemotherapy-induced triggered coagulation cascade, high thrombosis rates can occur. However, it is speculated that this high rate of thrombosis (both arterial and venous) is most likely related to the combination of semaxanib with cisplatin and gemcitabine , which are also associated with vascular events independently [104, 105].


Management


As with bevacizumab, there are no standardized recommendations on specifically how to manage these patients within the realm of cardio-oncology. Withdrawal of TKI therapy and treatment of arterial thrombosis as per oncology and cardiology guidelines is currently advised [99, 100]. The role of screening and prophylactic anticoagulation is not defined. Furthermore, large studies assessing the role of various risk stratification tools to evaluate patients vulnerable to the atherothrombotic effects of TKIs are lacking [12, 86].


Small Molecule Tyrosine Kinase Inhibitors: Bcr-Abl Inhibitors


Bcr-Abl tyrosine kinase inhibitors are also small molecule TKIs that appear to have anti-angiogenic activity, although they do not directly inhibit the VEGF pathway. Bcr-Abl TKIs are first-line therapy for treatment of chronic myelogenous leukemia (CML) . More than 90 % of CMLs are caused by a chromosomal abnormality with formation of a “Philadelphia chromosome” which is essentially the fusion between the Abelson (Abl) tyrosine kinase gene on chromosome 9 and the breakpoint cluster (Bcr) gene at chromosome 22 [106]. Therapy targeting this specific mutation has been developed and the first approved therapy was imatinib (Gleevec™) by the FDA in 2001 [107]. The therapeutic benefits from this targeted therapy have been astounding and truly revolutionized CML therapy. However, resistance has since emerged to imatinib in more than 20 % of patients. Therefore, second- and third-generation Bcr-Abl inhibitors (dasatinib (Sprycel™) , nilotinib (Tasigna™) , bosutinib (Bosulif™) , and ponatinib (Iclusig™) ) have been developed to overcome imatinib resistance. These are promising therapies from an oncologic standpoint, but they have been associated with significant vascular complications. Specifically these agents have been associated with acute coronary syndromes, stroke, and acute limb ischemia [96, 107].


Mechanisms of Action and Toxicity


The mutated fusion protein, Bcr-Abl, is expressed only on malignant cells, and thus Bcr-Abl-inhibiting TKIs are theoretically specific to leukemic cells. The TKIs bind to the amino acids of the Bcr-Abl tyrosine kinase ATP-binding site and stabilize the inactive form of the protein, preventing tyrosine autophosphorylation and downstream phosphorylation of its substrates [108]. Second- and third-generation Bcr-Abl-inhibiting TKIs target the genetic mutations leading to imatinib resistance. Ponatinib is an example and has activity against the specific T3151 mutation in Bcr-Abl implicated in many imatinib-resistant cases [107]. The mechanisms underlying the association between the second- and third-generation Bcr-Abl-inhibiting TKIs and vascular complications are largely unknown. Unlike other anti-angiogenic therapies , these small molecule TKIs do not have established VEGF receptor or pathway inhibiting properties. However, they are often referred to as “accidental” angiogenesis inhibitors as they likely still affect the angiogenesis pathway [88, 96, 109]. There is some speculation that the vascular toxicity associated with these agents may be related to unrecognized kinase targets being inhibited or at least preferentially blocked by the second- and third-generation TKIs which have demonstrated acute atherothrombosis and accelerated atherosclerosis [110]. These TKIs have been demonstrated to have effects on the discoidin domain receptor 1 (DDR1), which has been implicated in plaque formation in atherosclerosis [110]. Additional targets of these TKIs, KIT and PDGFR, appear to be involved in regulation of various vascular and perivascular cells, and thus disruption may lead to vascular events [110]. Some small prospective trials show evidence that nilotinib may also result in metabolic derangements including glucose intolerance and even diabetes, altered lipid profiles, and atherogenesis [110112].

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jun 25, 2017 | Posted by in CARDIOLOGY | Comments Off on Vascular Complications of Cancer and Cancer Therapy

Full access? Get Clinical Tree

Get Clinical Tree app for offline access