Your activity: 17 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Toxicity of molecularly targeted antiangiogenic agents: Non-cardiovascular effects

Toxicity of molecularly targeted antiangiogenic agents: Non-cardiovascular effects
Authors:
Toni K Choueiri, MD
Guru P Sonpavde, MD
Section Editors:
Michael B Atkins, MD
Richard M Goldberg, MD
Richard A Larson, MD
Deputy Editors:
Diane MF Savarese, MD
Jennifer S Tirnauer, MD
Literature review current through: Dec 2022. | This topic last updated: Nov 01, 2022.

INTRODUCTION — The use of angiogenesis inhibitors in cancer therapy is expanding, following the recognition of the role of angiogenesis in promoting tumor growth [1]. Multiple trials have shown that angiogenesis inhibitors yield incremental improvements in outcomes for a variety of advanced solid tumors.

Several classes of agents are available:

Bevacizumab is a monoclonal antibody against vascular endothelial growth factor (VEGF) that inhibits binding of the normal VEGF ligand to its receptor. In the United States (US), the approval of bevacizumab for metastatic colorectal cancer (mCRC) by the US Food and Drug Administration (FDA) ushered in the modern era of antiangiogenic therapy. The European Medicines Agency (EMA) granted approval for bevacizumab in mCRC in January 2006. Subsequently, bevacizumab has been approved by the FDA and the EMA for a wide variety of advanced solid tumors:

(See "Systemic therapy for nonoperable metastatic colorectal cancer: Selecting the initial therapeutic approach", section on 'Efficacy and toxicity of bevacizumab and biosimilars'.)

(See "Systemic chemotherapy for advanced non-small cell lung cancer", section on 'Bevacizumab'.)

(See "Antiangiogenic and molecularly targeted therapy for advanced or metastatic clear cell renal carcinoma", section on 'Bevacizumab plus interferon alfa'.)

(See "Management of recurrent high-grade gliomas", section on 'Bevacizumab'.)

(See "Management of recurrent or metastatic cervical cancer", section on 'Chemotherapy plus bevacizumab as first-line treatment'.)

Aflibercept, another VEGF ligand inhibitor, is a VEGF receptor (VEGFR) fusion molecule that binds to and inhibits VEGF binding to all classes of VEGFR, as well as placenta growth factor (PlGF) binding to VEGFR-1; it is approved in both the US and Europe in combination with chemotherapy for recurrent mCRC. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'Role of aflibercept'.)

Ramucirumab is a recombinant monoclonal antibody of the immunoglobulin G1 (IgG1) class that binds to the VEGFR-2, blocking receptor activation. It is approved for use in advanced gastric cancer and NSCLC in both the United States and Europe, and for mCRC in the United States. (See "Progressive, locally advanced unresectable, and metastatic esophageal and gastric cancer: Approach to later lines of systemic therapy", section on 'Ramucirumab with or without paclitaxel' and "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'Ramucirumab' and "Systemic chemotherapy for advanced non-small cell lung cancer", section on 'Ramucirumab plus docetaxel'.)

Multiple orally active tyrosine kinase inhibitors (TKIs) that block angiogenesis by inhibiting the actions of VEGF and other growth factors (eg, platelet-derived growth factor) are available, including sunitinib, sorafenib, pazopanib, vandetanib, cabozantinib, axitinib, ponatinib, lenvatinib, regorafenib, and tivozanib. These drugs have received approval for treatment of a variety of tumors, including RCC, hepatocellular cancer (HCC), gastrointestinal stromal tumors (GIST), thyroid cancer, pancreatic neuroendocrine tumors (PNET), soft tissue sarcomas (STS), refractory chronic myelogenous leukemia, and refractory mCRC both in the United States and Europe [2].

(See "Antiangiogenic and molecularly targeted therapy for advanced or metastatic clear cell renal carcinoma", section on 'Preferred VEGFR inhibitors'.)

(See "Systemic treatment for advanced hepatocellular carcinoma", section on 'Sorafenib'.)

(See "Tyrosine kinase inhibitor therapy for advanced gastrointestinal stromal tumors", section on 'Alternative therapies'.)

(See "Medullary thyroid cancer: Systemic therapy and immunotherapy", section on 'Kinase inhibitors'.)

(See "Differentiated thyroid cancer refractory to standard treatment: Systemic therapy", section on 'Mutation not identified'.)

(See "Metastatic well-differentiated gastrointestinal neuroendocrine (carcinoid) tumors: Systemic therapy options to control tumor growth", section on 'Treatments targeting tumor angiogenesis'.)

(See "Systemic treatment of metastatic soft tissue sarcoma".)

(See "Treatment of chronic myeloid leukemia in chronic phase after failure of initial therapy", section on 'Ponatinib'.)

(See "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'Regorafenib'.)

With the expanding use of agents that target the VEGF signaling pathway in cancer therapy, it is increasingly recognized that they are associated with a wide spectrum of toxicities which, in a small number of cases, may be fatal [3,4]. While some adverse effects are shared with conventional chemotherapeutic agents (which were designed to target cell division), many are unique and not typically observed with conventional cytotoxics:

Class-effects of VEGF axis inhibition by both VEGF-ligand and VEGFR TKIs include cardiovascular effects (hypertension, thromboembolism, left ventricular dysfunction) and non-cardiovascular effects (proteinuria, bleeding, delayed wound healing, gastrointestinal perforation, fatigue, and dysphonia).

Other rare class-effects of VEGF axis inhibition include reversible posterior leukoencephalopathy (RPLS), osteonecrosis of the jaw (ONJ), and microangiopathic hemolysis.

The antiangiogenic TKIs have additional class effects, including gastrointestinal events (diarrhea, nausea), thyroid dysfunction, fatigue, stomatitis, myelosuppression, and cutaneous effects (including hand-foot syndrome). In addition, severe and occasionally fatal hepatic toxicities have been described with sunitinib, sorafenib, ponatinib, and pazopanib. Rare effects include pancreatic enzyme elevations, hypoglycemia, and QTc prolongation.

Some of these side effects may reflect the promiscuity of kinase inhibitors, which inhibit multiple other receptors in addition to VEGFRs (referred to as off-target activity).

Some toxicities are mostly reported with certain agents, such as nasal septal perforation with bevacizumab, and sarcopenia with sorafenib.

Many of these adverse effects are serious, and some may be fatal. (See 'Risk of fatality' below.)

This topic review will cover the non-cardiovascular adverse effects of the antiangiogenic agents. Cardiovascular toxicities from angiogenesis inhibitors and thrombotic complications of thalidomide, lenalidomide, and pomalidomide, drugs that have immunomodulatory as well as some antiangiogenic activity, are discussed in detail separately. (See "Toxicity of molecularly targeted antiangiogenic agents: Cardiovascular effects" and "Multiple myeloma: Prevention of venous thromboembolism in patients receiving immunomodulatory drugs (thalidomide, lenalidomide, and pomalidomide)".)

A discussion of infusion reactions to therapeutic monoclonal antibodies is also presented separately. (See "Infusion-related reactions to therapeutic monoclonal antibodies used for cancer therapy".)

RISK OF FATALITY — Meta-analyses have demonstrated a small risk of fatal adverse events (approximately 1.5 to 2.5 percent, relative risk [RR] 1.5-2.2) with both antiangiogenic tyrosine kinase inhibitors (TKIs) and bevacizumab [3-5]. In one analysis, bevacizumab was associated with an increased risk of fatal events in combination with taxanes or platinum agents (RR 3.49) but not in combination with other agents (RR 0.85) [3].

In two meta-analyses, hemorrhage was the most common fatal adverse event with both classes of agents; other causes of treatment-related death were cardiac, gastrointestinal tract perforation, hepatic dysfunction, infection, and cerebrovascular events [3,4]. In another meta-analysis examining fatal events with antiangiogenic TKIs, heart failure, pulmonary emboli, hepatic failure, intestinal perforation, and pneumonia/respiratory failure were numerically higher on the antiangiogenic TKIs treatment arms [5]. In this study, the increased RR for death with the antiangiogenic TKIs was statistically significant for renal cell carcinoma (RCC) but not for lung cancer. However, the increased risk seen in patients treated for RCC may be ascribed partly to increased exposure time to the antiangiogenic TKIs in these patients relative to those with lung cancer.

CLASS SIDE EFFECTS OF VEGF INHIBITORS

Proteinuria/nephrotic syndrome

Incidence and mechanism — Both vascular endothelial growth factor (VEGF) ligand-inhibiting agents (bevacizumab, aflibercept) and the small molecule antiangiogenic tyrosine kinase inhibitors (TKIs) are associated with proteinuria, which is rarely in the nephrotic range (>3.5 g/24 hours) and even more rarely associated with the nephrotic syndrome [6-10]. Hypertension frequently accompanies proteinuria (see below). Proteinuria is usually an asymptomatic event detected only through laboratory analysis. (See "Toxicity of molecularly targeted antiangiogenic agents: Cardiovascular effects", section on 'Hypertension' and "Overview of heavy proteinuria and the nephrotic syndrome", section on 'Introduction'.)

The overall incidence of mild proteinuria in patients treated with bevacizumab ranges from 21 to up to 63 percent, but grade 3 or 4 proteinuria (defined as 3+ on dipstick, >3.5 g of protein/24 hours, or the nephrotic syndrome) affects approximately 2 percent of treated patients [11]. The incidence is not higher in patients who receive shorter bevacizumab infusions (ie, 10 versus 90 minutes) [12].

Fewer data are available for aflibercept, but in a phase III trial, proteinuria developed in 62 percent of patients treated with aflibercept plus chemotherapy (versus 41 percent of those treated with chemotherapy alone), and it was severe (grade 3 or 4) in 7.8 versus 1.2 percent [13].

Among patients treated with ramucirumab, the risk of proteinuria may be lower. In a meta-analysis of six placebo-controlled randomized trials, the incidence of all-grade proteinuria for ramucirumab versus placebo was 9.4 versus 3.1 percent, while the risk of severe (grade 3 or 4) proteinuria was 1.1 versus 0.04 percent [14].

Among patients with cancer treated with antiangiogenic TKIs, the incidence of mild and asymptomatic proteinuria also ranges from 21 to 63 percent, but heavy proteinuria is reported in up to 6.5 percent of patients [9]. In a meta-analysis, the incidence of all-grade and high-grade proteinuria with VEGF receptor (VEGFR) TKIs was 18.7 and 2.4 percent, respectively, with a corresponding increased risk of all-grade (odds ratio [OR] 2.92, 95% CI: 1.09-7.82, p = 0.033) and high-grade (OR 1.97, 95% CI: 1.01-3.84, p = 0.046) proteinuria when compared with controls [15]. The incidence of proteinuria with regorafenib may be lower than with other agents (7 percent all grade in one trial, 1 percent grade 3 or 4) [16].

Although proteinuria appears to be an effect common to all agents targeting the VEGF pathway, the factors associated with the occurrence and severity of proteinuria in patients treated with the individual agents are incompletely characterized:

Preexisting renal disease (including higher baseline urine protein levels and hypertension) and a diagnosis of renal cell carcinoma (RCC), as compared with other malignant diseases, may be predisposing factors for all agents [10,17-19].

Others suggest a dose dependency of proteinuria in patients treated with bevacizumab and an increase in risk when bevacizumab is combined with chemotherapy [11,17].

Proteinuria has been described as a late effect of cabozantinib, associated with previous chemotherapy and/or treatment with other TKIs and prolonged treatment with cabozantinib [20].

However, the relationship between treatment duration and proteinuria and whether the development of proteinuria serves as a surrogate marker of antitumor efficacy for most antiangiogenic agents is unknown [9].

The exact mechanism underlying proteinuria is not known. Reports of renal biopsies among patients with proteinuria receiving VEGF-targeted agents are sparse; when reported, the most common causative agent was bevacizumab. Histologic findings have included thrombotic microangiography (TMA), collapsing glomerulopathy, and isolated reports of cryoglobulinemic and immune complex glomerulonephritis [9,21-25]. (See "Toxicity of molecularly targeted antiangiogenic agents: Cardiovascular effects", section on 'Agent-specific effects' and "Pathophysiology of TTP and other primary thrombotic microangiopathies (TMAs)".)

Studies in rodents have demonstrated that VEGF plays a major role in endothelial development and the maintenance of a fenestrated endothelium [26] and in repair of glomerular endothelial injury [27]. Renal TMA has been recapitulated in a mouse model in which VEGF was locally ablated within the kidney [21]. It is hypothesized, although not proven, that renal TMA leads to glomerular capillary endothelial injury, which in turn, is responsible for proteinuria. Hypertension, which commonly accompanies proteinuria, could also contribute to endothelial injury.

Management — The implications of asymptomatic proteinuria from VEGF inhibitors are unknown and it is possible that the vast majority of cases have no clinical consequences. However, proteinuria has been linked to adverse cardiovascular outcomes and progression to end stage renal disease in patients with chronic kidney disease, and as such, proteinuria is identified as a target for treatment in kidney diseases in general. (See "Secondary factors and progression of chronic kidney disease", section on 'Albuminuria'.)

Evidence-based guidelines for management of proteinuria in patients receiving VEGF-targeted agents are lacking. The United States (US) prescribing information for bevacizumab recommends intermittent monitoring for the development of proteinuria but does not provide specific recommendations, except temporary withholding of the drug if protein excretion if >2 g/24 hours, and permanent discontinuation for patients who develop the nephrotic syndrome. However, this complication is uncommon and many institutions do not routinely dipstick urine prior to each dose of bevacizumab. In a report of 243 patients receiving bevacizumab for a metastatic solid tumor, the development of proteinuria affected treatment decisions in only 2 percent of cases [28].

Baseline and periodic urinalysis is also recommended during treatment with pazopanib, lenvatinib, and axitinib, with treatment interruption or discontinuation for patients who develop moderate to severe proteinuria (defined as ≥3 g/24 hours for pazopanib, ≥2 g/24 hours for lenvatinib and axitinib) until resolution. There are no guidelines for sorafenib, sunitinib, vandetanib, and cabozantinib; however, good clinical practice dictates baseline and periodic assessment for proteinuria (every eight weeks; more frequently if significant proteinuria is detected) for these agents also.

Although discontinuation of the anti-VEGF agent results in significant reduction in proteinuria, persistence is common [29]. For patients with persisting proteinuria, in the absence of specific therapy directed against the underlying disease, lowering of intraglomerular pressure, which may reduce protein excretion, may be achieved by the administration of an angiotensin converting enzyme inhibitor or angiotensin receptor blocker. There are no controlled studies, however, evaluating the benefit of these agents in patients with proteinuria related to therapy with a VEGF inhibitor. (See "Moderately increased albuminuria (microalbuminuria) in type 2 diabetes mellitus", section on 'ACE inhibitors and ARBs' and "Moderately increased albuminuria (microalbuminuria) in type 1 diabetes mellitus", section on 'Angiotensin inhibition' and "Overview of heavy proteinuria and the nephrotic syndrome", section on 'Proteinuria'.)

Management of drug-induced TMA is presented separately. (See "Drug-induced thrombotic microangiopathy (DITMA)".)

Other kidney problems — Less commonly, nephritic syndrome, acute kidney injury, and proliferative glomerulonephritis have been reported with bevacizumab [30]. Renal insufficiency and diabetes insipidus have been reported in clinical trials of vandetanib in medullary thyroid cancer and lung cancer, although causality has not been proven [31,32].

Alterations in red blood cell production — In mouse models, very high-grade VEGF inhibition is associated with reversible erythrocytosis, reticulocytosis, and increases in red blood cell mass [33]. Cyclic changes in hemoglobin levels have been noted in patients with metastatic renal cell cancer receiving sunitinib [34,35]. In addition, sunitinib administration is frequently associated with macrocytosis with an elevated mean corpuscular volume (MCV) in the absence of folate deficiency.

Although the mechanism underlying erythrocytosis is not established, it could be due either to temporary loss of intravascular fluid volume (ie, relative polycythemia) caused by inhibition of VEGFR-2 and subsequent reduction in nitric oxide, or secondarily by an increase in erythropoiesis driven by hepatic erythropoietin synthesis [34,36].

Secondary erythrocytosis has also been described in patients treated with sorafenib, axitinib, and bevacizumab [37,38]. Intriguingly, in a study of 10 patients with RCC who developed secondary erythrocytosis with bevacizumab, the peak increase of hemoglobin correlated with longer progression-free survival.

Bleeding — All VEGF-targeted agents have been associated with an increased risk of hemorrhage. Perturbation of endothelial cell function by targeting molecules expressed on the endothelial cell surface, such as VEGFRs, may heighten susceptibility to bleeding. In addition, direct antitumor activity leading to cavitation in an area of tumor that contains poorly developed neovessels that lack a well-formed musculature has been postulated to cause pulmonary hemorrhage, particularly in squamous cell lung cancer [22]. Finally, coexisting thrombocytopenia may aggravate bleeding.

Bevacizumab and aflibercept — Meta-analyses have demonstrated an increase in the risk of bleeding with bevacizumab; most commonly this is grade 1 epistaxis, but serious and, in some cases, fatal hemorrhagic events, including hemoptysis, gastrointestinal bleeding, hematemesis, intracerebral hemorrhage, epistaxis, and vaginal bleeding, have occurred.

In one trial-level meta-analysis, the risk of major bleeding in patients with advanced solid tumors who were treated with bevacizumab (at any dose) was 2.8 percent (95% CI 2.1-3.6) [39]. The overall relative risk (RR) of high-grade bleeding was 1.60 and was lower at 2.5 mg/kg/week (RR 1.27) compared with 5 mg/kg per week (RR 3.02). Higher risks were observed in patients with non-small cell lung cancer (NSCLC; RR 3.41), RCC (RR 6.37), and colorectal cancer (RR 9.11) receiving bevacizumab 5 mg/kg per week.

In another trial-level meta-analysis, the incidence of all-grade hemorrhage with bevacizumab was 30 percent, and 3.5 percent were high grade [40]. The overall RR for any grade bleeding was 2.48, with RRs of 3.02 and 2.01 for 5 and 2.5 mg/kg/week, respectively. Most hemorrhages occurred within the first five months of treatment. The most common type of hemorrhage was epistaxis, although hemoptysis, gastrointestinal bleeding, intracerebral hemorrhage, and intratumoral hemorrhage also occurred. The RR of high-grade bleeding was 1.91 and the risk of fatal bleeding was low (0.8 percent) and significantly elevated only in lung cancer (RR 5.02).

In contrast to these two studies, in an individual patient level meta-analysis evaluating the risk of arterial thromboembolism, the risk of serious bleeding was modestly increased with bevacizumab but the difference was not statistically significant [41]. Grade 3 and 4 bleeding events occurred in 3.7 percent of bevacizumab-treated patients versus 1.8 percent of control patients. The rate of bleeding events per 100 person-years was 5.3 with bevacizumab and 3.3 for controls (ratio = 1.6, 95% CI 0.86 to 2.97; p = 0.13). Baseline or on-study aspirin use was associated with a modest 1.3-fold increase in the risk of grade 3 or 4 bleeding events in both treatment groups (from 3.6 to 4.7 percent in bevacizumab-treated patients and from 1.7 to 2.2 percent for control subjects).

In contrast, the risk of severe (grade 3 or 4) bleeding with bevacizumab was not increased in patients receiving primary anticoagulant prophylaxis for venous thromboembolic disease, including low-dose aspirin in an analysis of the BRiTE observational registry [42].

Less information is available for aflibercept. In a phase III trial, epistaxis was noted in 28 percent of aflibercept-treated patients (versus 7 percent with chemotherapy alone), and the rate of grade 3 or 4 hemorrhage was 3 versus 1.7 percent in the control group [13].

Ramucirumab — Although severe and sometimes fatal hemorrhage has occurred in patients treated with ramucirumab, the risk appears to be low. In a 2017 meta-analysis of individual patient safety data from six placebo-controlled randomized trials in a variety of malignancies, the risk of all-grade bleeding was 38 versus 19 percent, but it was not significantly elevated for severe (≥grade 3) bleeding (2.7 versus 2.8 percent) [14]. The product labeling for ramucirumab includes a Boxed Warning to permanently discontinue ramucirumab in patients who experience severe bleeding [43].

Antiangiogenic tyrosine kinase inhibitors — An increased risk of bleeding has also been reported for the antiangiogenic TKIs:

In a meta-analysis of 27 randomized trials, patients treated with an antiangiogenic TKI (vandetanib, sunitinib, sorafenib, axitinib, pazopanib, or regorafenib) had an overall incidence of all-grade and high-grade bleeding events of 9.1 and 1.3 percent, respectively [44]. The RR for all-grade bleeding was higher for patients receiving a TKI than controls (RR 1.67; 95% CI 1.19-2.33). The risk of all-grade hemorrhage varied significantly by tumor type and the specific TKI. The risk of all-grade hemorrhage was highest in patients with gastrointestinal stromal tumors, while for high-grade hemorrhage, it was highest for melanoma; in both cases, rates were lowest for lung cancer. The TKIs with the highest risk of all-grade hemorrhage were sorafenib, sunitinib, and pazopanib.

As noted above, in two separate meta-analyses that examined fatal adverse events with antiangiogenic TKIs and bevacizumab in advanced solid tumors, hemorrhage was the most common toxic cause of death with both classes of VEGF inhibitors [3,4]. (See 'Risk of fatality' above.)

Special categories of bleeding

Intracranial bleeding — Concerns have been raised about a potential increase in the risk of intracerebral hemorrhage (ICH) in patients treated with bevacizumab who have brain metastases. In a phase I study, bevacizumab was associated with fatal ICH in a patient with an unsuspected intracerebral metastasis from hepatocellular carcinoma [45]. Based on this single anecdotal report, patients with brain metastases have been excluded from most clinical trials evaluating bevacizumab. However, subsequent accumulation of data has demonstrated that the risk of ICH in patients with treated, nonhemorrhagic brain metastases or with previously undiagnosed brain metastases or with treatment-emergent brain metastases does not appear to be significantly greater than that of patients with brain metastases who are not treated with bevacizumab [46-54]:

A safety analysis analyzed the incidence of ICH with bevacizumab in approximately 13,000 patients from randomized and nonrandomized trials conducted in patients with breast, non-small cell lung, pancreatic, renal cell, or colorectal cancer [46]. Patients with central nervous system (CNS) metastases were at similar risk of developing ICH, independent of bevacizumab therapy. Among 187 patients who were found to have occult brain metastases, 3 of 91 bevacizumab treated patients developed grade 4 ICH, compared with one fatal ICH among 96 controls.

In a prospective study, 115 patients with previously treated brain metastases were given bevacizumab in combination with systemic chemotherapy or erlotinib as first or second-line therapy for non-small cell lung cancer (NSCLC) [47]. At a median on-study duration of six months, there were no reported episodes of ICH (95% CI 0.0-3.3 percent). Eighty percent had received prior whole-brain radiation therapy with or without radiosurgery and/or neurosurgery, and 19.1 percent had received radiosurgery alone, and one patient (0.9 percent) underwent neurosurgery alone. A small proportion of patients were on enoxaparin (8.7 percent) and warfarin (7.0 percent), and 9.6 percent of patients were on daily aspirin. Of the five patients who developed a pulmonary or non-CNS, nonpulmonary hemorrhage while receiving bevacizumab, none had a risk factor for hemorrhage.

An evidence-based review of the incidence of CNS bleeding with anti-VEGF therapy in patients with NSCLC and brain metastases concluded that neither bevacizumab nor sunitinib/sorafenib increased the risk of ICH in patients with treatment-emergent, pretreated, or untreated occult brain metastases [54]. (See "Systemic chemotherapy for advanced non-small cell lung cancer", section on 'Bevacizumab'.)

Even in glioblastomas, which are highly vascular, ICH appears to be uncommon; this adverse event occurred in 0 to 3.8 percent of patients across studies of bevacizumab [50,51]. However, concurrent anticoagulation and bevacizumab therapy may increase the risk of hemorrhage in these patients (11 versus 3 percent overall rate of ICH in anticoagulated versus non-anticoagulated patients in one retrospective review [52]). (See "Management of recurrent high-grade gliomas", section on 'Side effects'.)

CNS metastases from renal cell cancer have a propensity to bleed. (See "Epidemiology, clinical manifestations, and diagnosis of brain metastases", section on 'Stroke'.)

An early report suggested a high incidence of ICH in patients with metastatic RCC and brain metastases who were treated with sunitinib or sorafenib [55]. In a retrospective review of 67 patients, five died of an ICH within two weeks after initiating therapy with sunitinib or sorafenib, four of whom had known brain metastases, all treated with RT. However, this could not be confirmed in an expanded access series analyzed the safety and efficacy of sunitinib treatment in 4564 patients with advanced RCC, of whom 321 had brain metastases [53]. Only one patient with a brain metastasis had a mild, treatment-related cerebral hemorrhage.

Data regarding ICH risk with other antiangiogenic TKIs used to treat RCC are lacking.

Taken together, these results suggest that patients with a history of treated nonhemorrhagic brain metastases probably should not be excluded from systemic therapy with a VEGF inhibitor as long as they are not on concurrent anticoagulation. In practice, antiangiogenic therapy is generally held during local therapy for brain metastases. (See "Systemic chemotherapy for advanced non-small cell lung cancer", section on 'Bevacizumab'.)

The decision to use bevacizumab in an anticoagulated patient with a recurrent primary brain tumor is more complex and must be based on a careful assessment of the risk to benefit ratio. This subject is discussed in detail elsewhere. (See "Management of recurrent high-grade gliomas", section on 'Side effects'.)

Pulmonary hemorrhage and cavitation — Pulmonary hemorrhage is a known complication of bevacizumab, especially in patients with squamous cell NSCLC [22,56,57]. In a phase II trial, 4 of 13 squamous carcinomas compared with 2 of 54 adenocarcinomas manifested severe hemorrhage [22]. As a result of this finding, subsequent phase III trials in NSCLC excluded squamous histology, and rates of pulmonary hemorrhage have been low. As examples (see "Systemic chemotherapy for advanced non-small cell lung cancer", section on 'Bevacizumab'):

In the Eastern Cooperative Oncology Group (ECOG) 4599 trial of paclitaxel plus carboplatin with or without bevacizumab, grade ≥3 pulmonary hemorrhage was reported in eight (1.9 percent) bevacizumab recipients (five of which were fatal), while only one grade 3 event (0.2 percent) occurred in the control group [56].

In the AVAiL trial of cisplatin plus gemcitabine with or without bevacizumab, grade ≥3 pulmonary hemorrhage was observed in five patients (1.5 percent) receiving bevacizumab 7.5 mg/kg, and three (0.9 percent) receiving 15.0 mg/kg, compared with two (0.6 percent) in the control group [58]. The incidences of fatal PH in these groups were 1.2, 0.9, and 0.3 percent, respectively.

Hemoptysis has not been seen in patients receiving bevacizumab for advanced colorectal or breast cancer.

Central tumor cavitation is common with the use of antiangiogenic agents, and is reported in 14 to 25 percent of cases of NSCLC during bevacizumab treatment [57,59]. In a retrospective study of phase II trials, the presence of baseline cavitation but not central tumor location was associated with a higher risk of hemorrhage [57,60].

Given the aforementioned data, bevacizumab is contraindicated in patients with squamous cell lung carcinoma and in any patient with hemoptysis (>2.5 mL of blood) within three months. Additionally, some trials have also excluded patients with NSCLC from receiving bevacizumab if their tumors invade or abut major blood vessels, but this is not widely practiced [58]. (See "Systemic chemotherapy for advanced non-small cell lung cancer", section on 'Bevacizumab'.)

Tumor cavitation and bleeding complications have been observed rarely with the use of sunitinib [61] and sorafenib [62] for NSCLC. However, pulmonary hemorrhage has not been described in patients receiving either drug for other malignancies, and neither agent is approved for treatment of lung cancer.

Management — The VEGF inhibitor needs to be discontinued in the setting of a severe bleed and supportive transfusions should be instituted. Given the short half-life of antiangiogenic TKIs (unlike bevacizumab), cessation of the drug may rapidly reverse bleeding in this context. Aspirin and anticoagulants should be discontinued, with careful consideration of the risk/benefit ratio before resumption.

Minor bleeding (eg, epistaxis) may be managed symptomatically with no discontinuation or only temporary cessation of the agent. (See "Approach to the adult with epistaxis".)

Bronchoscopic laser coagulation, electrocautery, argon plasma coagulation, hemostatic tamponade, and bronchial artery embolization may be useful in severe pulmonary hemorrhage [63,64]. Endobronchial irrigation with cold saline or epinephrine solution appears to have limited efficacy [65]. Radiation therapy and surgery may be necessary to salvage some cases. (See "Evaluation and management of life-threatening hemoptysis".)

The management of intracranial hemorrhage should follow good clinical practice guidelines. (See "Spontaneous intracerebral hemorrhage: Acute treatment and prognosis".)

Delayed wound healing

Bevacizumab — Bevacizumab has been associated with impaired wound healing in a variety of settings. This adverse effect is likely due to the critical role played by VEGF and angiogenesis during the early stages of wound healing. (See "Basic principles of wound healing".)

The incidence of wound healing problems with bevacizumab was addressed in a meta-analysis comparing fluorouracil (FU)-based chemotherapy with or without bevacizumab in patients who underwent surgery after beginning chemotherapy for metastatic colorectal cancer (mCRC) [66]. Wound healing complications were observed in a higher fraction of those receiving bevacizumab (3.4 [1 of 29] versus 13 [10 of 75] percent), although this difference was not statistically significant (p<0.28). Of the 10 patients who experienced wound healing complications after surgery with bevacizumab plus chemotherapy, the time interval between bevacizumab and surgery was 0 to 29 days in five patients, 30 to 59 days in five; no patient with more than 60 days between administration of bevacizumab and surgery had a wound complication.

Based on these data, and the long half-life of bevacizumab (20 days), at least 28 days (preferably six to eight weeks) should elapse between surgery and last dose of bevacizumab, when possible [66]. (See "Potentially resectable colorectal cancer liver metastases: Integration of surgery and chemotherapy", section on 'Issues related to bevacizumab'.)

The safety of surgery performed ≥6 weeks after the last dose of bevacizumab was confirmed in a retrospective study of three prospective trials of bevacizumab in patients with metastatic breast cancer [67]. There was a low risk of severe bleeding (0.1 to 0.9 percent) and a low risk of severe wound-healing complications (1.3 to 2.2 percent).

Hepatic metastasectomy — Impaired wound healing is especially germane to patients receiving bevacizumab prior to hepatic metastasectomy. Concerns about impaired wound healing and possibly impaired hepatic regeneration may affect the safety of metastasectomy, particularly if performed too soon after bevacizumab administration. It is generally recommended that six to eight weeks should elapse between the last dose of bevacizumab and elective hepatic resection. This subject is discussed in detail elsewhere. (See "Potentially resectable colorectal cancer liver metastases: Integration of surgery and chemotherapy".)

Recurrent glioma — Among patients treated for glioma, use of preoperative bevacizumab also appears to be associated with delayed wound healing or wound dehiscence [68,69]. In a series of 209 patients undergoing a second or third reoperation for recurrent glioblastoma, significantly more patients receiving preoperative bevacizumab developed wound healing complications than did non-bevacizumab-treated patients (35 versus 10 percent) [69]. Wound healing complications developed in only 6 percent of those who only received bevacizumab postoperatively. The incidence of wound complications was most striking for the third craniotomy (44 percent) and for a shorter delay between bevacizumab and surgery. Based on these results, the authors recommend that craniotomy not be performed until at least 28 days after the last dose of bevacizumab.

Chest wall port placement — Timing of administration also affects wound healing after chest wall port placement. In two studies, administration of bevacizumab within 10 to 14 days of chest wall placement of an implantable venous access device (port) was associated with a higher incidence of wound healing complications and wound dehiscence [70,71]. When possible, bevacizumab should be held for at least two weeks after placement of a port.

Tracheoesophageal fistula — The development of tracheoesophageal fistula has been reported in patients who were given bevacizumab with concomitant chemoradiotherapy or in the setting of prior thoracic irradiation:

Two independent phase II trials were discontinued for safety reasons after enrolling 29 patients with limited stage small cell lung cancer and five patients with advanced NSCLC, respectively [72]. Among the 34 patients, there were four confirmed cases of tracheoesophageal fistula and one suspected clinically. In all cases, patients were receiving bevacizumab with concomitant chemoradiotherapy.

There are at least two reported cases of tracheoesophageal fistula developing in patients treated with bevacizumab who have a prior history of mediastinal irradiation [73,74].

Bevacizumab should be used cautiously in patients who have received prior mediastinal irradiation and avoided altogether in patients receiving concomitant chemoradiotherapy.

Ramucirumab — There are few data on ramucirumab use in patients with serious or nonhealing wounds. In a 2017 meta-analysis of individual patient safety data from six randomized placebo-controlled trials of ramucirumab in a variety of malignancies (totaling 2748 ramucirumab-treated patients and 2248 placebo-treated patients), wound healing complications developed in only 14 patients treated with ramucirumab, compared with four patients in the control group (0.5 versus 0.2 percent). Nevertheless, because of the potential risk of delayed wound healing, ramucirumab should be withheld prior to surgery [75].

Antiangiogenic tyrosine kinase inhibitors — Impaired wound healing (and reopening of previously healed wounds) has also been observed following treatment with antiangiogenic TKIs [53,76-79]:

In a retrospective review of all patients undergoing cytoreductive nephrectomy at MD Anderson over a seven-year period, use of presurgical systemic targeted therapy with a broad range of VEGF inhibitors including TKIs was predictive of having a complication >90 days postoperatively, having multiple complications, and having wound complications [77].

Another study comparing perioperative complications and surgical outcomes among 14 patients undergoing surgery after neoadjuvant sunitinib or sorafenib versus a control group of 73 consecutively treated patients who underwent surgery in the absence of prior systemic therapy observed less hemorrhagic and wound healing issues but a significant increase in incidence and severity of intraoperative adhesions during debulking nephrectomy in patients who received preoperative therapy [53]. The median time from TKI discontinuation to surgery was two weeks.

Based on these findings, in our view, all antiangiogenic TKIs should be ideally held for one week prior to elective major surgery, unless otherwise specified in the United States Prescribing Information for these agents, and all should be held for two weeks postoperatively and/or until wounds are reasonably healed.

At many institutions, therapy with these agents is held for four weeks after major surgery and for at least two weeks after minor surgery, although there are no prospective data validating this approach. The decision to resume therapy following a major surgical intervention should be based on clinical judgment of recovery from surgery.

Intestinal perforation/fistula formation — All VEGF-targeted therapies can cause gastrointestinal perforation (GIP) and fistula formation, although this complication is best described in patients receiving bevacizumab. The mechanism by which these drugs contribute to GIP has not been proven, but proposed mechanisms include intestinal wall disruption (ulceration) in areas of tumor necrosis, disturbed platelet-endothelial cell homeostasis causing submucosal inflammation and subsequent ulcer formation, impaired healing of pathologic or surgical bowel injury, and mesenteric ischemia from thrombosis and/or vasoconstriction [80-82].

Bevacizumab — GIP is an infrequent but potentially fatal toxicity of bevacizumab. GIP may lead to peritonitis requiring emergency operative intervention, fistula formation [83], or intraabdominal abscess. GIP has been reported in patients treated with bevacizumab for a variety of malignancies, but are most often described in the setting of mCRC and epithelial ovarian cancer (EOC). Clinicians should maintain a high index of suspicion for GIP in patients who develop acute abdominal pain while receiving bevacizumab, even if they have no apparent risk factors.

Diseases other than ovarian cancer — The risk of GIP in patients treated with bevacizumab for conditions other than EOC has been addressed in the following studies:

In randomized trials of chemotherapy with and without bevacizumab in patients with mCRC and community-based cohort studies, the incidence of GIP in patients treated with bevacizumab has ranged from 1 to 4 percent. In a large community-based BRiTE observational cohort of patients treated with bevacizumab for mCRC, 37 of 1953 evaluable patients (1.9 percent) experienced GIP [84]. The majority of GIPs (26 of 37) occurred ≤6 months after starting bevacizumab (median, 3.35 months). In multivariate analysis, age ≥65 years was significantly associated with lower GIP risk (1.1 versus 2.6 percent), while intact primary tumor (3 versus 1.7 percent) and prior adjuvant radiation therapy (3.4 versus 1.7 percent) were associated with increased risk. Neither a prior history of peptic ulcer disease nor use of aspirin or other NSAIDs was a risk factor. No cumulative risk was observed with duration of bevacizumab exposure.

All patients receiving bevacizumab are at risk, regardless of the type of malignancy; however, risk is variable. In a meta-analysis of over 12,000 patients from 17 randomized trials conducted in a variety of tumor types (six in mCRC, three NSCLC, two RCC, two pancreatic, four breast cancer, no trials in ovarian cancer), the incidence of GIP was approximately 1 percent overall regardless of tumor type, and the mortality rate was 22 percent [85]. The RR compared with controls not receiving bevacizumab was 2.14 (95% CI 1.19-3.85; p = 0.011), and the risk was greater with higher bevacizumab doses (RRs for 5 and 2.5 mg/kg per week were 2.67 [95% CI 1.14-6.26] and 1.61 [95% CI 0.76-3.38], respectively). Risks were highest in patients treated for CRC (RR 3.10, 95% CI 1.26-7.63) and RCC (RR 5.67, 0.66-48.42). Among patients with NSCLC, risk was not significantly elevated compared with controls (RR 1.55, 95% CI 0.37-6.59).

Although several risk factors have been described for GIP during bevacizumab treatment, bowel perforation may occur even in the absence of predisposing risk factors [86], and it remains difficult to predict which patients will develop this complication. Many cases involve perforation of an in situ bowel primary [87]. However, surgical site complications can also occur at previously resected primary sites, often in the setting of previous irradiation or a prior anastomotic leak [83,88,89]. Perforation of an ileal neobladder has also been reported [90].

GI tract ulcers, possibly caused by bevacizumab, may predispose to perforation. One report described 18 patients with advanced colorectal cancer who participated in a phase III study and who were receiving oxaliplatin, capecitabine, and bevacizumab with or without cetuximab for mCRC who developed a GI ulcer (n = 6), a GIP (n = 8), or a perforated ulcer (n = 4) [91]. The occurrence of the perforations early in treatment, the established role of VEGF in ulcer healing [92], and the inhibitory effect of bevacizumab on wound healing support the causative role of bevacizumab in ulcerogenesis.

Accumulating data suggest a significantly increased risk of perforation in patients treated with bevacizumab for mCRC who subsequently undergo placement of an enteral stent. Thus, colonic stenting should not be performed in patients who are receiving bevacizumab. These data are addressed in detail elsewhere. (See "Enteral stents for the management of malignant colorectal obstruction", section on 'Stenting in the setting of adjunctive therapy'.)

Another potential risk factor for GIP in patients receiving bevacizumab for treatment of glioma is use of concurrent high-dose glucocorticoids [93]. (See "Management of recurrent high-grade gliomas", section on 'Bevacizumab'.)

Ovarian cancer — Although GIP has been reported in patients treated with bevacizumab for a variety of cancer primaries, it has attracted substantial attention in epithelial ovarian cancer (EOC) due to early reports noting high rates of GIP in these patients, and the common involvement of both the mesentery and the bowels in women with this disease.

Early studies suggested rates as high as 10 to 11 percent:

An early phase II study was terminated early when five GIPs were observed among 44 women (11 percent) treated with bevacizumab monotherapy for chemorefractory disease [94].

In another report of 112 patients with EOC who were treated with bevacizumab-containing regimens, 12 experienced a serious gastrointestinal event (10 [9 percent] had a GIP, and two a fistula) [95]. In this series the 30-day mortality rate was 50 percent, and 30 percent had died within one week of diagnosis. The only risk factor associated with GIP/fistula formation was rectovaginal nodularity (OR 3.64, 95% CI 1.1-12.1).

These studies lacked control groups for comparison, and they were conducted in populations of women with advanced, chemotherapy-refractory disease. It has been suggested that GIP may be more common in patients who are heavily pretreated with chemotherapy or who have diffuse peritoneal disease, significant small bowel disease, or bowel obstruction [80,96-98], and that the exclusion of such patients from bevacizumab therapy results in an underestimation of the true rate of GIP [99]. Even lower rates of GIP and fistula formation have in fact been reported in subsequent phase III trials comparing bevacizumab-containing versus non-bevacizumab chemotherapy for women with EOC, possibly because women were excluded who have evidence of GI tract obstruction, and many of the trials were undertaken in the first-line setting, or for women with recurrent, platinum-sensitive disease rather than in heavily pretreated women with advanced disease. As examples (see "First-line chemotherapy for advanced (stage III or IV) epithelial ovarian, fallopian tube, and peritoneal cancer", section on 'Incorporation of angiogenesis inhibitors' and "Medical treatment for relapsed epithelial ovarian, fallopian tube, or peritoneal cancer: Platinum-sensitive disease", section on 'Angiogenesis inhibitors'):

In Gynecologic Oncology Group trial 218, in which 1873 previously untreated women with ovarian cancer were randomly assigned to paclitaxel plus carboplatin with or without bevacizumab, the risk of GIP/fistula formation with and without bevacizumab was 3 versus 1 percent [100]. In a later analysis, GI tract fistulas of any grade were reported in 4 of the 625 patients treated with chemotherapy alone compared with 8 of the 1248 patients receiving bevacizumab; the corresponding numbers for perforation were 2 of 625 versus 18 of 1248 [101]. Specific risk factors for adverse GI events with bevacizumab included a history of treatment for inflammatory bowel disease and bowel resection at primary surgery.

Similarly, low rates of GIP were reported in the ICON7 trial of first line carboplatin plus paclitaxel with or without bevacizumab [102]. Among 1528 women enrolled to the trial, GIP occurred in 10 bevacizumab treated patients (versus 3 in the control group, 1.3 versus <1 percent), and fistula formation/abscess developed in 13 (versus 10 patients in the control group, 1.7 versus 1.3 percent).

The role of bevacizumab in recurrent platinum-sensitive EOC was studied in the OCEANS trial, in which 484 women were randomly assigned to carboplatin plus gemcitabine with or without bevacizumab [103]. There were no reported cases of GIP, and rates of fistula/abscess formation were also low in the bevacizumab group (4 versus 1 in the control group, 1.6 versus 0.4 percent).

Aflibercept — Fewer data are available for aflibercept. In a phase III trial conducted in 1226 patients with mCRC, GIP occurred in the same percentage of patients treated with chemotherapy alone or with aflibercept (0.5 percent) [13]. Rates of fistula formation of the GI tract with and without aflibercept were 1.1 versus 0.3 percent, and for fistulas from other than GI origin, rates were 0.3 versus 0.2 percent.

The elimination half-life of aflibercept is shorter than for bevacizumab (6 versus 20 days). There are no data addressing the safety of surgery in patients receiving treatment with this agent.

Ramucirumab — Rates of GIP are also low with ramucirumab. In a 2017 meta-analysis of individual patient safety data from six placebo-controlled trials of ramucirumab in a variety of malignancies, the incidence of GIP was 1.1, versus 0.3 percent with placebo [14].

Antiangiogenic tyrosine kinase inhibitors — Whether antiangiogenic TKIs increase the risk of GI perforation is uncertain. There are few cases reported in trials and case reports [104-109]. A systematic review of 5352 patients with a variety of solid tumors from 20 clinical trials found an overall 1.3 percent incidence of GI perforation among patients receiving antiangiogenic TKIs (with no statistically significant increase compared with controls and with a mortality of 29 percent) [110]. When compared with patients treated with a control medication in these trials, there was no significant increase in the risk of GI perforation with the use of antiangiogenic TKIs, although the confidence intervals were wide (OR 2.99, 95% CI 0.85-10.53). Subgroup analysis suggested no variability in the incidence of GI perforation according to tumor type, specific TKI, or treatment regimen.

Management — In order to minimize the risk of GIP and fistula formation, at least 28 days (preferably six to eight weeks) should elapse between surgery and last dose of bevacizumab, except in emergency situations [66]. Given the shorter half-life of the antiangiogenic TKIs, at least two weeks may suffice in this context.

Any patient treated with an anti-VEGF agent, particularly bevacizumab, should be considered at risk for GIP. Mortality rates may be as high as 50 percent [111], and early detection might help reduce the morbidity and mortality of this complication.

Perforation may be asymptomatic [112], or it can present with abdominal pain due to peritoneal contamination, free air, hemoperitoneum, or intraabdominal abscess. Postoperative patients and those being treated for rectal cancer can present with a fistula or anastomotic leak.

For all patients with new-onset abdominal pain while receiving bevacizumab, the potential diagnosis of GIP should be evaluated urgently. The evaluation should include a complete history, physical examination (to rule out signs of peritonitis), and abdominal imaging (ie, radiograph to evaluate for free air, or non-contrasted computed tomography). Evaluation and diagnosis of suspected GIP are discussed in detail separately. (See "Evaluation of the adult with abdominal pain in the emergency department", section on 'Ancillary studies'.)

Any case of GIP should result in the immediate and permanent discontinuation of VEGF-targeted therapy. Beyond that, there are no specific recommendations for management of documented GIP in patients receiving VEGF-targeted therapies. Patients with confirmed or highly suspected GIP whose overall condition is unstable secondary to the GIP should be considered for immediate surgical repair or diversion. Those who are more stable can be considered for less invasive management strategies such as bowel rest and broad-spectrum antibiotics with or without percutaneous drainage of concurrent abscesses [112,113]. The timing of the presentation, the patient's overall condition, their goals and wishes, and overall prognosis are important factors in the decision to explore these patients surgically.

Fatigue — Fatigue is a common side effect with all antiangiogenic TKIs, and bevacizumab may enhance fatigue caused by other agents given in combination, eg, interferon or chemotherapy. While mild fatigue is common, severe fatigue is seldom seen. In a meta-analysis of randomized trials, the RR of all-grade and high-grade fatigue with single agent VEGFR TKIs was 1.35 (95% CI 1.22-1.49, p<0.001) and 1.33 (95% CI 0.97-1.82, p = 0.08), respectively [114]. The mechanism of fatigue due to angiogenesis inhibitors is unclear. Contributing factors may be other drugs, hypothyroidism (especially with sunitinib), anemia, dehydration (secondary to diarrhea, nausea, or vomiting), and cardiac dysfunction, which should be addressed. (See "Cancer-related fatigue: Prevalence, screening, and clinical assessment" and 'Thyroid dysfunction' below.)

Supportive care and psychostimulants may be employed. Severe fatigue may warrant dose modification or, rarely, discontinuation. (See "Cancer-related fatigue: Treatment".)

Dysphonia — Dysphonia has been observed with antiangiogenic TKIs, especially with newer more potent agents, such as axitinib, regorafenib, lenvatinib, and tivozanib [115-117]. Dysphonia was also reported in 25 percent of patients treated with aflibercept in a phase III trial conducted in mCRC (compared with 3 percent of the chemotherapy alone group) [13]. In contrast, dysphonia has not been reported in patients treated with bevacizumab.

Osteonecrosis of the jaw — Medication-related osteonecrosis of the jaw (MRONJ) is defined by areas of tissue breakdown and exposure of bone in the maxillofacial region that fail to heal within eight weeks in a patient who has not received jaw irradiation. (See "Medication-related osteonecrosis of the jaw in patients with cancer", section on 'Nomenclature and definition'.)

There are isolated case reports of MRONJ in patients treated with VEGF inhibitors, mostly bevacizumab, sunitinib, and lenvatinib [118-128]. However, the incidence appears to be low overall, at least in the absence of other risk factors. As examples:

In a retrospective study of 3560 patients receiving bevacizumab-containing therapy for advanced breast cancer in two double-blind, randomized trials and a large nonrandomized safety study, MRONJ occurred in 0.3 percent of patients receiving bevacizumab in the blinded phase of the two randomized trials and in 0.4 percent of patients in the single-arm study [129].

In a randomized trial in patients with thyroid cancer, there was only one case of MRONJ among 261 lenvatinib-treated patients (0.4 percent), versus none with placebo [116]. Furthermore, at least some data support the view that risk for MRONJ is increased with lenvatinib only in patients with concomitant exposure to other risk factors such as high potency bisphosphonates, everolimus, glucocorticoids, or invasive dental procedures [130].

As a result of these data, the US Food and Drug Administration (FDA) and the European Medicines Agency has issued an MRONJ advisory only for bevacizumab and sunitinib. Furthermore, the United States Prescribing Information for sunitinib also recommends that sunitinib be withheld for at least three weeks prior to scheduled dental surgery or invasive dental procedures, if possible. The United States Prescribing Information for lenvatinib suggests that the drug be withheld for at least one week prior to scheduled dental surgery or invasive dental procedures.

However, there is at least a potential risk of MRONJ associated with several other TKIs that target VEGF, including sorafenib, axitinib, vandetanib, regorafenib, and others. The incidence of MRONJ with these other agents is insufficiently characterized.

Concurrent use of antiangiogenic agents does represent a risk factor for MRONJ among patients with bone metastases who are also receiving therapy with an antiresorptive agent for prevention of skeletal related events. These data are discussed elsewhere. (See "Osteoclast inhibitors for patients with bone metastases from breast, prostate, and other solid tumors" and "Medication-related osteonecrosis of the jaw in patients with cancer", section on 'Concurrent antineoplastic therapy'.)

In view of the difficulty in treating established MRONJ, prevention is emphasized. Patients at risk could have a comprehensive dental examination and preventive dentistry (preemptive extraction of unsalvageable teeth and optimized periodontal health) before beginning therapy with an antiangiogenic agent, particularly if they are also receiving concomitant therapy with antiresorptive agent such as a bisphosphonate or denosumab for skeletal metastases. Oral examinations and hygiene status should be monitored during treatment. Invasive dental procedures (eg, placement of dental implants) should be avoided during therapy if at all possible. (See "Medication-related osteonecrosis of the jaw in patients with cancer", section on 'Prevention'.)

Treatment objectives for patients with an established diagnosis of MRONJ are to eliminate pain, control infection of the soft tissue and bone, and minimize the progression or occurrence of bone necrosis. Treatment has generally shifted away from aggressive surgical interventions and towards conservative therapy with limited debridement, antibiotics, and oral rinses with chlorhexidine or hydrogen peroxide. (See "Medication-related osteonecrosis of the jaw in patients with cancer", section on 'Treatment of established MRONJ'.)

Reversible posterior leukoencephalopathy and brain capillary leak syndrome — Reversible posterior leukoencephalopathy syndrome (RPLS) is a clinical radiographic syndrome of heterogeneous etiologies that are grouped together because of similar findings on neuroimaging studies. It is also often referred to as:

Posterior reversible encephalopathy syndrome (PRES)

Reversible posterior cerebral edema syndrome

Posterior leukoencephalopathy syndrome

Hyperperfusion encephalopathy

Brain capillary leak syndrome

None of these names is completely satisfactory; the syndrome is not always reversible, and it is often not confined to either the white matter or the posterior regions of the brain. (See "Reversible posterior leukoencephalopathy syndrome", section on 'Introduction and terminology'.)

The pathogenesis of RPLS remains unclear, but it appears to be related to disordered cerebral autoregulation and endothelial dysfunction. (See "Reversible posterior leukoencephalopathy syndrome", section on 'Pathogenesis'.)

The clinical syndrome of reversible posterior leukoencephalopathy syndrome (RPLS) is characterized by headaches, altered consciousness, visual disturbances, and seizures; hypertension is frequent but not invariable. (See "Reversible posterior leukoencephalopathy syndrome", section on 'Clinical manifestations'.)

A wide variety of medical conditions have been implicated as causes of RPLS (table 1), including rapidly developing, fluctuating, or intermittent hypertension, and therapy with a VEGF-targeted agent (bevacizumab, sunitinib, sorafenib, lenvatinib, and pazopanib [131-141]). (See "Reversible posterior leukoencephalopathy syndrome", section on 'Related conditions'.)

Prevention and management — Stringent control of blood pressure is critical to avert this rare complication. In suspected cases of RPLS, discontinuation of the offending agent is essential. (See "Reversible posterior leukoencephalopathy syndrome", section on 'Management'.)

CLASS SIDE EFFECTS OF VEGF RECEPTOR TYROSINE KINASE INHIBITORS

Thyroid dysfunction — Thyroid dysfunction is frequently observed in patients treated with sunitinib. Typically this has been manifested by hypothyroidism [142-144]. In one series, 15 of 42 euthyroid patients with intact thyroid glands (36 percent) became hypothyroid (as defined as a persistently elevated level of thyroid stimulating hormone [TSH]) while receiving sunitinib for advanced gastrointestinal stromal tumors (GIST) [143]. The risk increased with longer duration of therapy (18, 29, and 90 percent in patients treated for 36, 52, and 96 weeks, respectively). The mean time to development of hypothyroidism was 50 weeks.

The mechanism by which sunitinib causes hypothyroidism has yet to be determined [145]. Inhibition of iodine uptake may be responsible [146].

Several reports have also described transient thyrotoxicosis with sunitinib treatment in patients with metastatic renal cell carcinoma (RCC) [147-149]. In one report, six patients developed thyrotoxicosis after starting treatment with sunitinib, and four later became hypothyroid [147]. Some of these patients appeared to have a destructive thyroiditis, characterized by transient thyrotoxicosis (with low radioiodine uptake, which distinguishes it from Graves' disease) followed by hypothyroidism associated with atrophy of thyroid follicular cells [147,148]. However, a case of lymphocytic thyroiditis accompanied by transient thyrotoxicosis has also been reported [150]. (See "Overview of thyroiditis" and "Disorders that cause hyperthyroidism", section on 'Thyroiditis'.)

Thyroid dysfunction also occurs in patients treated with sorafenib for metastatic RCC, but this appears to be less frequent [144,151-156]. In an analysis of thyroid function in 39 patients, hypothyroidism was seen in seven (18 percent) and hyperthyroidism in one [151]. Two of the hypothyroid patients (5 percent of the total) were symptomatic and required thyroid replacement therapy. Others report a frequency of thyroid hormone replacement therapy among patients taking sorafenib of 2.6 and 6 percent, respectively [144,156]. As with sunitinib, patients treated with sorafenib have also been reported to develop thyrotoxicosis with associated thyroiditis [151,152,155,157,158].

Hypothyroidism is also reported among patients treated with pazopanib, axitinib, and tivozanib and hence appears to be a class-effect of these agents [159-161]. Pazopanib appears to have the lowest reported incidence, with <10 percent of patients developing hypothyroidism in a phase III trial [160]. Hypothyroidism rates are approximately 11 percent with tivozanib [162].

Early reports with axitinib suggest a very high incidence. In a phase I trial, 89 percent of patients had elevations in TSH [159].

Finally, another potential mechanism for hypothyroidism that may be seen in patients being treated with antiangiogenic tyrosine kinase inhibitors (TKIs) for GISTs is consumptive hypothyroidism due to excessive degradation of thyroid hormone; however, this appears to be caused by overexpression of the thyroid hormone inactivating enzyme type 3 iodothyronine deiodinase (D3) within large GISTs and not a drug-related adverse effect. This subject is discussed in detail elsewhere. (See "Disorders that cause hypothyroidism", section on 'Consumptive hypothyroidism' and "Clinical presentation, diagnosis, and prognosis of gastrointestinal stromal tumors", section on 'Adults'.)

Lenvatinib is a multitargeted TKI approved for advanced, radioiodine-refractory, differentiated thyroid cancer and in combination with everolimus for RCC after one prior vascular endothelial growth factor (VEGF) inhibitor [1]. It has been associated with impaired suppression of TSH in patients receiving exogenous thyroid hormone supplementation [163]. The United States (US) prescribing information recommends monitoring TSH levels monthly and adjusting thyroid replacement medication as needed. (See "Differentiated thyroid cancer refractory to standard treatment: Systemic therapy", section on 'Mutation not identified'.)

Management — Because of the high prevalence of hypothyroidism, regular surveillance of TSH levels is warranted during therapy with antiangiogenic TKIs, more frequently in patients receiving sunitinib and lenvatinib. We suggest that thyroid function be evaluated at baseline and monitored every 4 to 12 weeks thereafter and earlier if dictated by symptoms. (See "Diagnosis of and screening for hypothyroidism in nonpregnant adults" and "Treatment of primary hypothyroidism in adults".)

Thyroid hormone supplementation should be given to symptomatic patients with hypothyroidism. Discontinuations and dose modifications of the VEGF receptor TKI is usually not necessary.

Transient thyrotoxicosis may be seen preceding hypothyroidism and there are no reports suggesting that therapeutic intervention is warranted.

Myelosuppression — Myelosuppression has been observed more commonly when utilizing antiangiogenic TKIs that more potently target Flt-3 (FMS-related tyrosine kinase 3 receptor) and KIT, which are required for hematopoiesis [164]. When comparing across different trials, sunitinib and sorafenib appear more myelosuppressive than more selective TKIs such as pazopanib, axitinib, and cabozantinib. A meta-analysis of trials of single agent sunitinib revealed high-grade neutropenia in 12.8 percent, thrombocytopenia in 10.7 percent, and anemia in 6.2 percent, respectively [165]. Indeed, the combination of sunitinib with relatively non-myelosuppressive chemotherapy regimens, eg, carboplatin plus paclitaxel, has appeared prohibitively myelosuppressive [166]. Moreover, thrombocytopenia and leukopenia induced by sunitinib appeared to be Flt-3 genotype and CYP1A polymorphism dependent, respectively, in retrospective studies [167,168]. The incidences of sorafenib-associated high-grade anemia, neutropenia, and thrombocytopenia in a meta-analysis were 2.0, 5.1, and 4.0 percent, respectively [169].

Bevacizumab is not known to cause significant myelosuppression.

Recurring grade 3 or 4 neutropenia or thrombocytopenia (table 2) persisting for at least five days and/or febrile neutropenia/bleeding require dose alterations.

Oral toxicity — Stomatitis and other manifestations of oral toxicity may be seen with all the antiangiogenic TKIs. Rates are variable, but it appears to be more common with sunitinib (29 to 48 percent), sorafenib (25 to 28 percent), cabozantinib (51 percent), and lenvatinib (40 percent) than with pazopanib (4 to 12 percent) and axitinib (15 percent) [115,170,171]. Symptoms of oral pain, dysesthesia/mucosal sensitivity, dysgeusia, dysphagia, dry mouth, and aphthous ulcers may occur rather than conventional oral mucositis [172]. The mechanism is unclear but may be related to poor healing of microtrauma (similar to the proposed mechanism for hand-foot skin reaction). (See 'Cutaneous toxicity' below and "Oral toxicity associated with systemic anticancer therapy", section on 'Etiology and risk factors'.)

In a phase III trial of FOLFIRI with or without aflibercept in metastatic colorectal cancer, rates of mucositis were higher in the combined therapy arm (55 versus 35 percent for all-grade, and 14 versus 5 percent for severe) [13], suggesting that anti-VEGF agents may exacerbate the risk of mucositis from chemotherapy; however, this has not been seen with bevacizumab.

Gastrointestinal toxicities — Diarrhea, nausea, and emesis have been observed with all antiangiogenic TKIs and are generally mild.

In clinical trials, diarrhea of any grade has been reported in 30 to 79 percent of patients (highest rates with vandetanib), with severe diarrhea (grade 3 or 4) in 3 to 17 percent [16,32,115,173-177]. Vandetanib is a moderately potent epidermal growth factor receptor (EGFR) inhibitor in addition to being a vascular endothelial growth factor receptor (VEGFR) inhibitor, which explains the somewhat higher incidence of diarrhea associated with this agent [173,174].

In clinical trials, nausea of any grade has been reported in 23 to 58 percent of treated patients, and rates are highest in patients treated with sunitinib, lenvatinib, cabozantinib [116,170,178,179]. Vomiting of any grade has been reported in 10 to 48 percent of treated patients; rates are lowest with pazopanib and sorafenib and highest with sunitinib, lenvatinib, and cabozantinib [116,170,178,179].

Pancreatic atrophy been reported in patients receiving long-term sorafenib. The possibility of pancreatic insufficiency should be considered if a patient treated with sorafenib has refractory diarrhea with clinical features suggesting of pancreatic exocrine insufficiency. (See 'Pancreatic atrophy' below and "Exocrine pancreatic insufficiency", section on 'Clinical manifestations'.)

Management — Symptomatic management of grade 1 or 2 diarrhea with an oral antidiarrheal agent, such as loperamide, and avoidance of foods and supplements (eg fiber) that increase gastrointestinal motility generally suffices [180]. Treatment interruption is necessary for grade 3 or 4 diarrhea, and dose modifications may be necessary to control diarrhea if the drug is continued.

Nausea and vomiting is rarely severe and can usually be controlled with oral antiemetics. However, caution should be exercised in combining vandetanib, lenvatinib, sunitinib, and sorafenib with serotonin antagonists such as granisetron and ondansetron, due to the potential for QTc interval prolongation and torsade de pointes. (See "Toxicity of molecularly targeted antiangiogenic agents: Cardiovascular effects", section on 'Prolongation of the QTc interval and cardiac arrhythmias' and "Prevention of chemotherapy-induced nausea and vomiting in adults", section on 'Cardiac issues'.)

Cutaneous toxicity — A range of cutaneous toxicities has been reported in patients receiving antiangiogenic TKIs (table 3). (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'BCR-ABL tyrosine kinase inhibitors'.)

Hand-foot skin reaction (HFSR) is a common cutaneous manifestation of toxicity with oral antiangiogenic TKIs. HFSRs appear to be more common with sorafenib than sunitinib, and more common with axitinib, lenvatinib, regorafenib, and vandetanib, as compared with pazopanib [16,115,116,181]. In a meta-analysis that examined the types of cutaneous toxicities, vandetanib exhibited the highest incidence of rash (41 percent), while sorafenib was most commonly associated with HFSR (37 percent) and pruritus (14 percent) [182]. A meta-analysis of 831 patients who received cabozantinib demonstrated an overall incidence of HFSR of 35 percent and grade 3 HFSR in 10 percent [183]. Interestingly, as has been seen with hypertension, an association between skin toxicity and improved outcomes has been described [184,185]. The mechanisms of HFSR include inhibition of pericyte-mediated endothelial survival, resulting in damage to the capillary endothelium in the hands and feet. Notably, KIT is strongly expressed in the ductal epithelium of eccrine glands, where the drug can be excreted [186]. (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'BCR-ABL tyrosine kinase inhibitors' and "Toxicity of molecularly targeted antiangiogenic agents: Cardiovascular effects", section on 'Association with antitumor efficacy'.)

Multiple case reports have also identified an increased risk of keratoacanthomas and squamous cell carcinoma in patients treated with sunitinib or sorafenib. (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'Squamoproliferative lesions'.)

Vandetanib also inhibits the EGFR and has been associated with a generalized maculopapular erythematous acneiform rash that is typical of other EGFR inhibitors (eg, gefitinib, erlotinib, cetuximab). (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'VEGFR/PDGFR inhibitors'.)

Prevention and management — The United States Prescribing Information provides suggested dose modifications for sorafenib based on the grade of HFSR (table 4) during therapy [187]. Guidelines are not available for the other agents, although the institution of supportive measures, dose reductions, or permanent discontinuations may be necessary depending on the severity and persistence of dermatologic toxicity.

Prevention and management strategies for HFSR in patients receiving sunitinib or sorafenib are also available from an international consensus group [180]. This subject is presented in detail elsewhere. (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'Hand-foot skin reaction'.)

Prevention and management strategies for the acneiform eruption in patients receiving vandetanib and other EGFR inhibitors are provided in detail elsewhere. (See "Acneiform eruption secondary to epidermal growth factor receptor (EGFR) and MEK inhibitors", section on 'Prevention' and "Acneiform eruption secondary to epidermal growth factor receptor (EGFR) and MEK inhibitors", section on 'Management'.)

Given the early onset of skin toxicities and the association with the dose of regorafenib, a randomized phase II trial demonstrated that a dose-escalation strategy (starting dose 80 mg/day orally with weekly escalation by 40 mg increments to 160 mg/day if no significant drug-related adverse events occurred) may be an acceptable alternative to a standard-dose strategy (160 mg/day orally) for 21 days of a 28-day cycle [188]. Interestingly, the trough concentration of M5, a major metabolite of regorafenib, was associated with the incidence of skin toxicities [189]. However, pharmacokinetic monitoring is not used clinically. (See "Systemic therapy for nonoperable metastatic colorectal cancer: Approach to later lines of systemic therapy", section on 'Regorafenib'.)

Definitive guidelines for continuing versus discontinuing sorafenib in patients who develop squamoproliferative lesions while on sorafenib or sunitinib therapy have not been established. (See "Cutaneous adverse events of molecularly targeted therapy and other biologic agents used for cancer therapy", section on 'Squamoproliferative lesions'.)

Hepatotoxicity — Severe and occasionally fatal hepatotoxicity has been observed in clinical studies with all VEGFR TKIs. The risk for elevations in serum alanine transaminase (ALT), aspartate transaminase (AST), alkaline phosphatase (ALP), and bilirubin during therapy with VEGFR TKIs was addressed in a meta-analysis of 18,282 patients from 52 randomized controlled trials of a variety of VEGFR TKIs [190]. The incidence of hepatic failure with VEGFR TKIs was 0.8 percent overall. Compared with patients not treated with a VEGFR TKI, the relative risks (RRs) for all-grade elevations in ALT, AST, ALP and bilirubin in patients receiving a VEGFR TKI were 1.57 (95% CI 1.38-1.79, p<0.001), 1.57 (95% CI 1.36-1.81, p<0.001), 1.20 (95% CI 1.09-1.83, p<0.001), and 1.55 (95% CI 1.21-1.97, p<0.001), respectively. The RRs for high-grade elevations were 1.66 (95% CI 1.25-2.20, p = 0.001), 1.61 (95% CI 1.21-2.14, p = 0.001), 1.02 (95% CI 0.70-1.47, p = 0.932), and 1.34 (95% CI 1.0-1.81, p = 0.054), respectively. The RR of all-grade hepatotoxicity did not significantly differ between relatively more specific VEGFR TKIs, such as axitinib, as compared with less specific VEGFR TKIs (sunitinib, sorafenib, pazopanib, vandetanib, cabozantinib, ponatinib, regorafenib).

Monitoring and management — Patients treated with any of these agents should have a baseline evaluation of liver function tests (LFTs) and periodic re-evaluation during therapy. For patients treated with pazopanib, the United States Prescribing Information recommends assessing LFTs at baseline and at weeks 3, 5, 7, and 9, 12 and 16, and thereafter periodically as clinically indicated. Treatment should be interrupted or discontinued if evidence of hepatotoxicity is observed.

Specific recommendations for individual drugs include the following:

Pazopanib – Polymorphisms in the uridine diphospho-glucuronosyltransferase 1A1 (UGT 1A1) enzyme that cause Gilbert's syndrome (the UGT1A1*28 allele) may be associated with pazopanib-induced hyperbilirubinemia. Isolated hyperbilirubinemia in these patients may represent a benign manifestation of Gilbert's syndrome, and continuation of pazopanib monotherapy is reasonable in this setting. This subject is discussed in more detail elsewhere. (See "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'Pazopanib'.)

For patients with pre-existing moderate hepatic impairment, the United States Prescribing Information recommends that starting doses of pazopanib be reduced and that the drug be avoided in patients with pre-existing severe hepatic impairment, defined as total bilirubin >3 times the upper limit of normal (ULN) with any level of ALT/AST elevation.

Ponatinib – As hepatic elimination is a major route of excretion, hepatic impairment may result in increased ponatinib exposure. The United States Prescribing Information recommends avoiding use of the drug in patients with moderate to severe (Child-Pugh B or C (table 5)) hepatic impairment unless the benefit outweighs the possible risk of ponatinib overexposure. (See "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'Ponatinib'.)

Sorafenib – Sorafenib-induced hepatitis is characterized by a hepatocellular pattern of liver damage with significant increases of transaminases, which may result in hepatic failure and death. Increases in bilirubin and International Normalized Ratio (INR) may also occur. The United States prescribing information recommends that the drug be discontinued in case of significantly increased transaminases without alternative explanation such as viral hepatitis or progressing underlying malignancy. There are conflicting data on the need for dose reduction in patients with preexisting liver dysfunction. (See "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'Sorafenib'.)

Sunitinib – The United States Prescribing Information recommends treatment interruption for bilirubin levels >3X ULN or AST/ALT >5X ULN, and discontinuation of sunitinib if there is no resolution or if patients subsequently experience severe changes in LFTs or have other signs and symptoms of liver failure. (See "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'Sunitinib'.)

Lenvatinib – The United States Prescribing Information recommends discontinuing the drug for grade 3 or worse liver impairment during therapy, and that the drug be permanently discontinued for hepatic failure. A reduced starting dose is recommended for patients with preexisting severe hepatic impairment (Child-Pugh C cirrhosis (table 5)). (See "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'Lenvatinib'.)

Axitinib – When used in combination with avelumab or pembrolizumab, the United States Prescribing Information recommends treatment interruption for ALT or AST ≥3X ULN but <10X ULN without concurrent total bilirubin ≥2X ULN, and discontinuing the drug for ALT/AST increase to >3X ULN with concurrent total bilirubin ≥2X ULN or any ALT/AST increase >10X ULN. A reduced starting dose is recommended for all patients with preexisting moderate hepatic impairment (Child-Pugh class B cirrhosis). (See "Chemotherapy hepatotoxicity and dose modification in patients with liver disease: Molecularly targeted agents", section on 'Axitinib'.)

Pancreatitis — Elevations of the pancreatic enzymes lipase and amylase are frequently reported in patients treated with antiangiogenic TKIs, although overt pancreatitis appears to relatively rare, with the possible exception of ponatinib [175,191-193]:

In a phase II trial of ponatinib in 81 patients with refractory chronic myelogenous leukemia, pancreatitis developed in 11 (14 percent) and was severe (grade 3 or worse) in four (5 percent) [192].

The risk of pancreatitis in patients treated with VEGFR TKIs other than ponatinib, cabozantinib, or regorafenib was addressed in a meta-analysis that included 10,578 patients from 22 randomized trials [193]. Pancreatitis of any grade occurred in 25 of 5569 patients receiving a VEGFR TKI (0.4 percent), and high grade pancreatitis occurred in 22 (0.4 percent). The RR for all grade and high-grade pancreatitis for the TKI versus no TKI arms was 1.95 (p = 0.042, 95% CI 1.02 to 3.70) and 1.89 (p = 0.069, 95% CI 0.95 to 373), respectively. There was no differential impact of malignancy type or specific TKI agent on RR of either all grade or high-grade pancreatitis.

We closely monitor patients with pancreatic enzyme elevations and continue therapy with the TKI unless patients develop clinical pancreatitis.

Hypoglycemia — Blood glucose levels may be reduced in patients who are treated with antiangiogenic TKIs for a variety of malignancies. As an example, hypoglycemia has occurred in clinical trials in approximately 2 percent of patients receiving sunitinib for renal cell cancer or GIST, and in approximately 10 percent of patients treated with sunitinib for a pancreatic neuroendocrine tumor [194].

There are conflicting data as to whether hypoglycemia is more common in diabetic as compared with nondiabetic patients:

In a retrospective series of 19 type II diabetic patients whose blood glucose was monitored at baseline and during treatment with sunitinib, blood glucose levels fell from 149 mg/dL at baseline to 117 mg/dL at four weeks (8.3 to 6.5 mmol/L) [195]. In contrast, blood glucose levels fell only minimally in nine nondiabetic patients (106 to 95 mg/dL [5.89 to 5.26 mmol/L]) who were similarly monitored. No serious episodes of hypoglycemia were reported.

However, other data suggest that declines in blood glucose occur both in diabetic and nondiabetic patients [196]. In this retrospective review of diabetic (n = 17) and nondiabetic (n = 61) patients treated with sorafenib, sunitinib, imatinib, or dasatinib, all four drugs were associated with significant mean declines in blood glucose, which were greatest (52 mg/dL) with dasatinib. The magnitude of decline in blood glucose was similar in diabetic and nondiabetic patients. On the other hand, revised product labeling for sunitinib suggests that reductions in blood glucose levels may be worse among diabetic patients treated with sunitinib as compared with nondiabetic patients [194].

Blood glucose levels should be routinely checked during and after treatment for all patients who are treated with antiangiogenic TKIs. Antidiabetic medications should be adjusted if necessary to minimize the risk of hypoglycemia.

AGENT-SPECIFIC EFFECTS

Bevacizumab

Nasal septal perforation — Nasal septal perforation has been observed with bevacizumab administered in combination with chemotherapy [197,198]. The US Food and Drug Administration (FDA) recognized this complication in 2006 after seven postmarketing reports became available [199]. However, this is a rare complication. To date, only 18 cases have been published in the literature [200]. Although the data are sparse, some groups of patients may be at a higher risk of this complication. The perforations occurred in combination with taxanes and higher doses of bevacizumab (15 mg/kg once every three weeks) in many cases [197], but they have also been reported in patients treated with bevacizumab and fluorouracil (FU)-based chemotherapy [198]. The overall risk in this series of 100 patients treated for metastatic colorectal cancer was only 1 percent.

The mechanism may be related to underlying mucositis, delayed tissue repair, and additive or synergistic antiangiogenic activity of taxanes.

Management — When patients develop this complication, other potential causes of perforation should be excluded, even if the patient is receiving bevacizumab. The evaluation should include inspection of the nasal septum for evidence of infection and obtaining local cultures to exclude other causes of perforation [201]. Septal perforation is often seen in those who use intranasal cocaine. It may also result from septal surgery, atrophic rhinitis, granulomatosis with polyangiitis, and several other disorders (table 6). (See "Etiologies of nasal symptoms: An overview", section on 'Disorders affecting the septum'.)

Patients with a nasal septal perforation should be referred to an otolaryngologist for evaluation and management. Most perforations, particularly those that are posterior and asymptomatic, can be medically managed with frequent irrigation of the area with saline, and application of lubricant gels and other supportive deal with epistaxis or pain [200]. In most instances, surgical repair is not undertaken, but closing the perforation with a bridge flap, or more commonly, a button, is an option [202].

The decision as to whether to continue bevacizumab in a patient who has developed a nasal septal perforation must be individualized. In the published literature, a few patients continued with bevacizumab and seemed to do fine [197,203,204]. However, given the propensity of bevacizumab to delay wound healing, it seems prudent to wait for some evidence of perforation stability and healing prior to continuing the drug [200].

Sorafenib — Long-term treatment with sorafenib has been associated with muscle wasting and pancreatic atrophy.

Muscle wasting/sarcopenia — Cancer cachexia is characterized by diminished nutrient intake and progressive tissue depletion, both of which lead to weight loss. A disproportionate and excessive loss of lean body mass is the hallmark of cancer cachexia. (See "Pathogenesis, clinical features, and assessment of cancer cachexia", section on 'Changes in body composition'.)

Sarcopenia (skeletal muscle wasting) may also be an adverse effect of treatment with the antiangiogenic TKIs, particularly sorafenib:

Sarcopenia was linked to dose-limiting toxicities in a phase I study of sorafenib in renal cell carcinoma (RCC) [205].

In a subset analysis of 80 patients with RCC who were treated in the TARGET trial, body weight was measured and skeletal muscle mass was serially assessed by computed tomography (CT) [206]. At six months, sorafenib therapy was associated with a statistically significant decrease in total body weight and muscle mass compared with placebo (-2.1 versus + 0.8 kg and -7.4 versus -3.1 cm2, respectively). The loss in weight and muscle mass in patients treated with sorafenib was progressive during treatment from 6 to 12 months.

An associated between sarcopenia, sorafenib exposure, and dose limiting toxicity within the first month of treatment has also been seen in patients receiving sorafenib for hepatocellular carcinoma (HCC) [207].

The loss of muscle mass appears to be additive to that caused by advanced cancer and may contribute to asthenia and fatigue.

Whether or not sarcopenia represents a class effect of VEGF-targeted therapy is unclear. At least some data report muscle loss in patients treated with bevacizumab-containing chemotherapy for metastatic cancer, but there was no control group in either study, and the effects could have been attributable to the chemotherapy that was given in conjunction with bevacizumab or progression of the cancer itself [208,209].

Pancreatic atrophy — A single report describes two patients who developed irreversible pancreatic atrophy while on long-term treatment with sorafenib [210]. One patient developed intermittent diarrhea within three months of starting sorafenib, with remissions when treatment was interrupted and recurrence with rechallenge; pancreatic exocrine insufficiency was diagnosed 18 months after treatment initiation. The second developed diarrhea two months after treatment initiation that was responsive to pancreatic enzyme replacement and was shown to have a 35 percent decrease in the volume of the pancreas by CT by 37 months after treatment initiation. This complication has also been described with sunitinib [211].

Pazopanib and muscle pain — Myalgias and muscle spasms can occur in patients treated with pazopanib. In one study of 369 patients treated with pazopanib or placebo for advanced soft tissue sarcoma (STS), musculoskeletal pain of any grade occurred in 23 percent of pazopanib-treated patients versus 9 percent of the control group [212].

SUMMARY AND RECOMMENDATIONS

Antiangiogenic agents have unique toxicities that differ from those of traditional chemotherapy. Antiangiogenic agents available in the clinic include the vascular endothelial growth factor (VEGF) ligand-inhibiting agents (bevacizumab, aflibercept, ramucirumab) and tyrosine kinase inhibitors (TKIs) that target angiogenesis (sunitinib, sorafenib, pazopanib, vandetanib, cabozantinib, axitinib, ponatinib, regorafenib, and lenvatinib). Toxicities of antiangiogenic agents may be classified as VEGF class specific, drug class specific, or agent specific. (See 'Introduction' above.)

Many of the adverse effects seen with angiogenesis inhibitors, particularly the cardiovascular effects and hemorrhage, are serious and potentially fatal. (See 'Risk of fatality' above.)

Careful selection of patients for therapy with angiogenesis inhibitors (ie, reasonable performance status, controlled blood pressure, and lack of serious cardiovascular events within six months) as well as close monitoring and prompt intervention are necessary to alleviate the risks posed by these toxicities.

All VEGF-targeted agents cause proteinuria, which is rarely in the nephrotic range (>3.5 g/24 hours) and even more rarely associated with the nephrotic syndrome. The implications of asymptomatic proteinuria from VEGF inhibitors are unknown, and it is possible that the vast majority of cases have no clinical consequences. However, baseline and periodic urinalysis should be performed during treatment with all of these agents. (See 'Proteinuria/nephrotic syndrome' above.)

All VEGF-targeted agents have been associated with an increased risk of hemorrhage. This is most commonly grade 1 epistaxis, but serious and, in some cases, fatal hemorrhagic events, including hemoptysis (particularly in patients with squamous cell lung cancer), gastrointestinal bleeding, hematemesis, intracerebral hemorrhage, epistaxis, and vaginal bleeding, have occurred (risk of major bleeding approximately 2 to 3 percent). (See 'Bleeding' above.)

Because of the risk of massive hemoptysis, bevacizumab is contraindicated in patients with squamous cell lung carcinoma and in any patient with hemoptysis (>2.5 mL of blood) within three months. (See 'Pulmonary hemorrhage and cavitation' above.)

The risk of intracranial bleeding is low, even in patients with nonhemorrhagic brain metastases and recurrent gliomas. Patients with a history of treated nonhemorrhagic brain metastases or a recurrent primary brain tumor need not be excluded from systemic therapy with a VEGF inhibitor. (See 'Intracranial bleeding' above.)

Bevacizumab and antiangiogenic TKIs have been associated with impaired wound healing in a variety of settings. At least 28 days (preferably six to eight weeks) should elapse between surgery and last dose of bevacizumab, except when clinically necessary. (See 'Bevacizumab' above.)

If the clinical situation permits, all antiangiogenic TKIs should be interrupted for at least one week before surgery and not reinitiated until adequate wound healing has occurred, usually at least two to four weeks after major surgery.

Although best described with bevacizumab, all VEGF-targeted therapies can cause gastrointestinal perforation (GIP) leading to peritonitis, fistula formation, or intraabdominal abscess. Clinicians should maintain a high index of suspicion for GIP in patients who develop acute abdominal pain while receiving bevacizumab or ramucirumab, even if they have no apparent risk factors. (See 'Intestinal perforation/fistula formation' above.)

There are isolated case reports of medication-related osteonecrosis of the jaw (ONJ) in patients treated with VEGF inhibitors, mostly bevacizumab and sunitinib, but the overall incidence appears to be low. (See 'Osteonecrosis of the jaw' above.)

Antiangiogenic therapy can cause reversible posterior leukoencephalopathy syndrome (RPLS), a clinical-radiographic syndrome thought to be due to brain capillary leak and characterized by headaches, altered consciousness, visual disturbances, and seizures with or without hypertension. (See 'Reversible posterior leukoencephalopathy and brain capillary leak syndrome' above.)

Fatigue is a common side effect with all VEGF-targeted agents, but it is usually mild. (See 'Fatigue' above.)

Dysphonia has been reported with antiangiogenic TKIs and aflibercept, but not bevacizumab. (See 'Dysphonia' above.)

Thyroid dysfunction is a class effect of antiangiogenic TKIs, although it is most frequent with sunitinib and lenvatinib. (See 'Thyroid dysfunction' above.)

Mild myelosuppression is also a class effect of antiangiogenic TKIs and is seen most often with sunitinib and sorafenib because they target Flt-3 (FMS-related tyrosine kinase 3 receptor) and KIT, which are required for hematopoiesis. (See 'Myelosuppression' above.)

Stomatitis may be seen with all the antiangiogenic TKIs, but it appears to be most common with sunitinib, sorafenib, and lenvatinib. (See 'Oral toxicity' above.)

Diarrhea, nausea, and emesis have been observed with all the antiangiogenic TKIs; rates of diarrhea are higher with vandetanib, probably because it also inhibits epidermal growth factor receptor (EGFR). (See 'Gastrointestinal toxicities' above.)

Cutaneous toxicity with antiangiogenic TKIs includes hand-foot skin reaction (especially with sorafenib), an increased risk of keratoacanthomas and squamous cell carcinoma (with sunitinib or sorafenib), and an acneiform rash (with vandetanib). (See 'Cutaneous toxicity' above.)

Sunitinib, ponatinib, sorafenib, lenvatinib, and especially pazopanib have been associated with severe and occasionally fatal hepatic toxicities. Patients treated with these agents should have a baseline evaluation of liver function tests (LFTs) and periodic re-evaluation (at least monthly) during therapy. (See 'Hepatotoxicity' above.)

Elevations of the pancreatic enzymes lipase and amylase have been reported with antiangiogenic TKIs, although overt pancreatitis is rare, with the possible exception of ponatinib. (See 'Pancreatitis' above.)

Bevacizumab has been associated in rare cases with nasal septal perforation. (See 'Nasal septal perforation' above.)

Sarcopenia (skeletal muscle wasting) has been associated with sorafenib. (See 'Muscle wasting/sarcopenia' above.)

  1. Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971; 285:1182.
  2. Jayson GC, Kerbel R, Ellis LM, Harris AL. Antiangiogenic therapy in oncology: current status and future directions. Lancet 2016; 388:518.
  3. Ranpura V, Hapani S, Wu S. Treatment-related mortality with bevacizumab in cancer patients: a meta-analysis. JAMA 2011; 305:487.
  4. Schutz FA, Je Y, Richards CJ, Choueiri TK. Meta-analysis of randomized controlled trials for the incidence and risk of treatment-related mortality in patients with cancer treated with vascular endothelial growth factor tyrosine kinase inhibitors. J Clin Oncol 2012; 30:871.
  5. Sivendran S, Liu Z, Portas LJ Jr, et al. Treatment-related mortality with vascular endothelial growth factor receptor tyrosine kinase inhibitor therapy in patients with advanced solid tumors: a meta-analysis. Cancer Treat Rev 2012; 38:919.
  6. Sandler AB, Johnson DH, Herbst RS. Anti-vascular endothelial growth factor monoclonals in non-small cell lung cancer. Clin Cancer Res 2004; 10:4258s.
  7. Gressett SM, Shah SR. Intricacies of bevacizumab-induced toxicities and their management. Ann Pharmacother 2009; 43:490.
  8. Costero O, Picazo ML, Zamora P, et al. Inhibition of tyrosine kinases by sunitinib associated with focal segmental glomerulosclerosis lesion in addition to thrombotic microangiopathy. Nephrol Dial Transplant 2010; 25:1001.
  9. Izzedine H, Massard C, Spano JP, et al. VEGF signalling inhibition-induced proteinuria: Mechanisms, significance and management. Eur J Cancer 2010; 46:439.
  10. Tomita Y, Uemura H, Fujimoto H, et al. Key predictive factors of axitinib (AG-013736)-induced proteinuria and efficacy: a phase II study in Japanese patients with cytokine-refractory metastatic renal cell Carcinoma. Eur J Cancer 2011; 47:2592.
  11. Wu S, Kim C, Baer L, Zhu X. Bevacizumab increases risk for severe proteinuria in cancer patients. J Am Soc Nephrol 2010; 21:1381.
  12. Shah SR, Gressett Ussery SM, Dowell JE, et al. Shorter bevacizumab infusions do not increase the incidence of proteinuria and hypertension. Ann Oncol 2013; 24:960.
  13. Van Cutsem E, Tabernero J, Lakomy R, et al. Addition of aflibercept to fluorouracil, leucovorin, and irinotecan improves survival in a phase III randomized trial in patients with metastatic colorectal cancer previously treated with an oxaliplatin-based regimen. J Clin Oncol 2012; 30:3499.
  14. Arnold D, Fuchs CS, Tabernero J, et al. Meta-analysis of individual patient safety data from six randomized, placebo-controlled trials with the antiangiogenic VEGFR2-binding monoclonal antibody ramucirumab. Ann Oncol 2017; 28:2932.
  15. Zhang ZF, Wang T, Liu LH, Guo HQ. Risks of proteinuria associated with vascular endothelial growth factor receptor tyrosine kinase inhibitors in cancer patients: a systematic review and meta-analysis. PLoS One 2014; 9:e90135.
  16. Grothey A, Van Cutsem E, Sobrero A, et al. Regorafenib monotherapy for previously treated metastatic colorectal cancer (CORRECT): an international, multicentre, randomised, placebo-controlled, phase 3 trial. Lancet 2013; 381:303.
  17. Zhu X, Wu S, Dahut WL, Parikh CR. Risks of proteinuria and hypertension with bevacizumab, an antibody against vascular endothelial growth factor: systematic review and meta-analysis. Am J Kidney Dis 2007; 49:186.
  18. Zhu X, Stergiopoulos K, Wu S. Risk of hypertension and renal dysfunction with an angiogenesis inhibitor sunitinib: systematic review and meta-analysis. Acta Oncol 2009; 48:9.
  19. Shord SS, Bressler LR, Tierney LA, et al. Understanding and managing the possible adverse effects associated with bevacizumab. Am J Health Syst Pharm 2009; 66:999.
  20. Cappagli V, Moriconi D, Bonadio AG, et al. Proteinuria is a late-onset adverse event in patients treated with cabozantinib. J Endocrinol Invest 2021; 44:95.
  21. Eremina V, Jefferson JA, Kowalewska J, et al. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med 2008; 358:1129.
  22. Johnson DH, Fehrenbacher L, Novotny WF, et al. Randomized phase II trial comparing bevacizumab plus carboplatin and paclitaxel with carboplatin and paclitaxel alone in previously untreated locally advanced or metastatic non-small-cell lung cancer. J Clin Oncol 2004; 22:2184.
  23. Maynard SE, Min JY, Merchan J, et al. Excess placental soluble fms-like tyrosine kinase 1 (sFlt1) may contribute to endothelial dysfunction, hypertension, and proteinuria in preeclampsia. J Clin Invest 2003; 111:649.
  24. Bollée G, Patey N, Cazajous G, et al. Thrombotic microangiopathy secondary to VEGF pathway inhibition by sunitinib. Nephrol Dial Transplant 2009; 24:682.
  25. George BA, Zhou XJ, Toto R. Nephrotic syndrome after bevacizumab: case report and literature review. Am J Kidney Dis 2007; 49:e23.
  26. Eremina V, Quaggin SE. The role of VEGF-A in glomerular development and function. Curr Opin Nephrol Hypertens 2004; 13:9.
  27. Ostendorf T, Kunter U, Eitner F, et al. VEGF(165) mediates glomerular endothelial repair. J Clin Invest 1999; 104:913.
  28. Yeh J, Frieze D, Martins R, Carr L. Clinical utility of routine proteinuria evaluation in treatment decisions of patients receiving bevacizumab for metastatic solid tumors. Ann Pharmacother 2010; 44:1010.
  29. Patel TV, Morgan JA, Demetri GD, et al. A preeclampsia-like syndrome characterized by reversible hypertension and proteinuria induced by the multitargeted kinase inhibitors sunitinib and sorafenib. J Natl Cancer Inst 2008; 100:282.
  30. Halimi JM, Azizi M, Bobrie G, et al. [Vascular and renal effects of anti-angiogenic therapy]. Nephrol Ther 2008; 4:602.
  31. Robinson BG, Paz-Ares L, Krebs A, et al. Vandetanib (100 mg) in patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Endocrinol Metab 2010; 95:2664.
  32. Wells SA Jr, Gosnell JE, Gagel RF, et al. Vandetanib for the treatment of patients with locally advanced or metastatic hereditary medullary thyroid cancer. J Clin Oncol 2010; 28:767.
  33. Tam BY, Wei K, Rudge JS, et al. VEGF modulates erythropoiesis through regulation of adult hepatic erythropoietin synthesis. Nat Med 2006; 12:793.
  34. Alexandrescu DT, McClure R, Farzanmehr H, Dasanu CA. Secondary erythrocytosis produced by the tyrosine kinase inhibitors sunitinib and sorafenib. J Clin Oncol 2008; 26:4047.
  35. Harshman LC, Kuo CJ, Wong BY, et al. Increased hemoglobin associated with VEGF inhibitors in advanced renal cell carcinoma. Cancer Invest 2009; 27:851.
  36. van der Veldt AA, Boven E, Vroling L, et al. Sunitinib-induced hemoglobin changes are related to the dosing schedule. J Clin Oncol 2009; 27:1339.
  37. Alexandre I, Billemont B, Meric JB, et al. Axitinib induces paradoxical erythropoietin synthesis in metastatic renal cell carcinoma. J Clin Oncol 2009; 27:472.
  38. Harshman LC KC, Srinivas S. Bevacizumab-associated erythrocytosis in metastatic renal cell carcinoma. J Clin Oncol 26: 2008 (May 20 suppl; abstr 16096).
  39. Hang XF, Xu WS, Wang JX, et al. Risk of high-grade bleeding in patients with cancer treated with bevacizumab: a meta-analysis of randomized controlled trials. Eur J Clin Pharmacol 2011; 67:613.
  40. Hapani S, Sher A, Chu D, Wu S. Increased risk of serious hemorrhage with bevacizumab in cancer patients: a meta-analysis. Oncology 2010; 79:27.
  41. Scappaticci FA, Skillings JR, Holden SN, et al. Arterial thromboembolic events in patients with metastatic carcinoma treated with chemotherapy and bevacizumab. J Natl Cancer Inst 2007; 99:1232.
  42. Kozloff M, Yood MU, Berlin J, et al. Clinical outcomes associated with bevacizumab-containing treatment of metastatic colorectal cancer: the BRiTE observational cohort study. Oncologist 2009; 14:862.
  43. Fuchs CS, Tomasek J, Yong CJ, et al. Ramucirumab monotherapy for previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (REGARD): an international, randomised, multicentre, placebo-controlled, phase 3 trial. Lancet 2014; 383:31.
  44. Qi WX, Tang LN, Sun YJ, et al. Incidence and risk of hemorrhagic events with vascular endothelial growth factor receptor tyrosine-kinase inhibitors: an up-to-date meta-analysis of 27 randomized controlled trials. Ann Oncol 2013; 24:2943.
  45. Gordon MS, Margolin K, Talpaz M, et al. Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J Clin Oncol 2001; 19:843.
  46. Besse B, Lasserre SF, Compton P, et al. Bevacizumab safety in patients with central nervous system metastases. Clin Cancer Res 2010; 16:269.
  47. Socinski MA, Langer CJ, Huang JE, et al. Safety of bevacizumab in patients with non-small-cell lung cancer and brain metastases. J Clin Oncol 2009; 27:5255.
  48. Crinò L, Dansin E, Garrido P, et al. Safety and efficacy of first-line bevacizumab-based therapy in advanced non-squamous non-small-cell lung cancer (SAiL, MO19390): a phase 4 study. Lancet Oncol 2010; 11:733.
  49. Carden CP, Larkin JM, Rosenthal MA. What is the risk of intracranial bleeding during anti-VEGF therapy? Neuro Oncol 2008; 10:624.
  50. Kreisl TN, Kim L, Moore K, et al. Phase II trial of single-agent bevacizumab followed by bevacizumab plus irinotecan at tumor progression in recurrent glioblastoma. J Clin Oncol 2009; 27:740.
  51. Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol 2009; 27:4733.
  52. Norden AD, Bartolomeo J, Tanaka S, et al. Safety of concurrent bevacizumab therapy and anticoagulation in glioma patients. J Neurooncol 2012; 106:121.
  53. Harshman LC, Yu RJ, Allen GI, et al. Surgical outcomes and complications associated with presurgical tyrosine kinase inhibition for advanced renal cell carcinoma (RCC). Urol Oncol 2013; 31:379.
  54. Sandler A, Hirsh V, Reck M, et al. An evidence-based review of the incidence of CNS bleeding with anti-VEGF therapy in non-small cell lung cancer patients with brain metastases. Lung Cancer 2012; 78:1.
  55. Pouessel D, Culine S. High frequency of intracerebral hemorrhage in metastatic renal carcinoma patients with brain metastases treated with tyrosine kinase inhibitors targeting the vascular endothelial growth factor receptor. Eur Urol 2008; 53:376.
  56. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with bevacizumab for non-small-cell lung cancer. N Engl J Med 2006; 355:2542.
  57. Crabb SJ, Patsios D, Sauerbrei E, et al. Tumor cavitation: impact on objective response evaluation in trials of angiogenesis inhibitors in non-small-cell lung cancer. J Clin Oncol 2009; 27:404.
  58. Reck M, von Pawel J, Zatloukal P, et al. Phase III trial of cisplatin plus gemcitabine with either placebo or bevacizumab as first-line therapy for nonsquamous non-small-cell lung cancer: AVAil. J Clin Oncol 2009; 27:1227.
  59. Marom EM, Martinez CH, Truong MT, et al. Tumor cavitation during therapy with antiangiogenesis agents in patients with lung cancer. J Thorac Oncol 2008; 3:351.
  60. Sandler AB, Schiller JH, Gray R, et al. Retrospective evaluation of the clinical and radiographic risk factors associated with severe pulmonary hemorrhage in first-line advanced, unresectable non-small-cell lung cancer treated with Carboplatin and Paclitaxel plus bevacizumab. J Clin Oncol 2009; 27:1405.
  61. Socinski MA, Novello S, Brahmer JR, et al. Multicenter, phase II trial of sunitinib in previously treated, advanced non-small-cell lung cancer. J Clin Oncol 2008; 26:650.
  62. Blumenschein GR Jr, Gatzemeier U, Fossella F, et al. Phase II, multicenter, uncontrolled trial of single-agent sorafenib in patients with relapsed or refractory, advanced non-small-cell lung cancer. J Clin Oncol 2009; 27:4274.
  63. Reck M, Barlesi F, Crinò L, et al. Predicting and managing the risk of pulmonary haemorrhage in patients with NSCLC treated with bevacizumab: a consensus report from a panel of experts. Ann Oncol 2012; 23:1111.
  64. Valipour A, Kreuzer A, Koller H, et al. Bronchoscopy-guided topical hemostatic tamponade therapy for the management of life-threatening hemoptysis. Chest 2005; 127:2113.
  65. Kvale PA, Selecky PA, Prakash UB, American College of Chest Physicians. Palliative care in lung cancer: ACCP evidence-based clinical practice guidelines (2nd edition). Chest 2007; 132:368S.
  66. Scappaticci FA, Fehrenbacher L, Cartwright T, et al. Surgical wound healing complications in metastatic colorectal cancer patients treated with bevacizumab. J Surg Oncol 2005; 91:173.
  67. Cortés J, Caralt M, Delaloge S, et al. Safety of bevacizumab in metastatic breast cancer patients undergoing surgery. Eur J Cancer 2012; 48:475.
  68. Chamberlain MC. Bevacizumab plus irinotecan in recurrent glioblastoma. J Clin Oncol 2008; 26:1012.
  69. Clark AJ, Butowski NA, Chang SM, et al. Impact of bevacizumab chemotherapy on craniotomy wound healing. J Neurosurg 2011; 114:1609.
  70. Erinjeri JP, Fong AJ, Kemeny NE, et al. Timing of administration of bevacizumab chemotherapy affects wound healing after chest wall port placement. Cancer 2011; 117:1296.
  71. Zawacki WJ, Walker TG, DeVasher E, et al. Wound dehiscence or failure to heal following venous access port placement in patients receiving bevacizumab therapy. J Vasc Interv Radiol 2009; 20:624.
  72. Spigel DR, Hainsworth JD, Yardley DA, et al. Tracheoesophageal fistula formation in patients with lung cancer treated with chemoradiation and bevacizumab. J Clin Oncol 2010; 28:43.
  73. Gore E, Currey A, Choong N. Tracheoesophageal fistula associated with bevacizumab 21 months after completion of radiation therapy. J Thorac Oncol 2009; 4:1590.
  74. Goodgame B, Veeramachaneni N, Patterson A, Govindan R. Tracheo-esophageal fistula with bevacizumab after mediastinal radiation. J Thorac Oncol 2008; 3:1080.
  75. US Food and Drug Administration (FDA)-approved prescribing information for ramucirumab available online at http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/125477lbl.pdf (Accessed on April 25, 2014).
  76. Feyerabend S, Schilling D, Wicke C, Stenzl A. Toxic dermatolysis, tissue necrosis and impaired wound healing due to sunitinib treatment leading to forefoot amputation. Urol Int 2009; 82:246.
  77. Chapin BF, Delacroix SE Jr, Culp SH, et al. Safety of presurgical targeted therapy in the setting of metastatic renal cell carcinoma. Eur Urol 2011; 60:964.
  78. Jonasch E, Wood CG, Matin SF, et al. Phase II presurgical feasibility study of bevacizumab in untreated patients with metastatic renal cell carcinoma. J Clin Oncol 2009; 27:4076.
  79. Powles T, Kayani I, Blank C, et al. The safety and efficacy of sunitinib before planned nephrectomy in metastatic clear cell renal cancer. Ann Oncol 2011; 22:1041.
  80. Han ES, Monk BJ. What is the risk of bowel perforation associated with bevacizumab therapy in ovarian cancer? Gynecol Oncol 2007; 105:3.
  81. Mir O, Mouthon L, Alexandre J, et al. Bevacizumab-induced cardiovascular events: a consequence of cholesterol emboli syndrome? J Natl Cancer Inst 2007; 99:85.
  82. Verheul HM, Pinedo HM. Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nat Rev Cancer 2007; 7:475.
  83. Ganapathi AM, Westmoreland T, Tyler D, Mantyh CR. Bevacizumab-associated fistula formation in postoperative colorectal cancer patients. J Am Coll Surg 2012; 214:582.
  84. Kabbinavar FF, Flynn PJ, Kozloff M, et al. Gastrointestinal perforation associated with bevacizumab use in metastatic colorectal cancer: results from a large treatment observational cohort study. Eur J Cancer 2012; 48:1126.
  85. Hapani S, Chu D, Wu S. Risk of gastrointestinal perforation in patients with cancer treated with bevacizumab: a meta-analysis. Lancet Oncol 2009; 10:559.
  86. Schellhaas E, Loddenkemper C, Schmittel A, et al. Bowel perforation in non-small cell lung cancer after bevacizumab therapy. Invest New Drugs 2009; 27:184.
  87. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med 2004; 350:2335.
  88. Bège T, Lelong B, Viret F, et al. Bevacizumab-related surgical site complication despite primary tumor resection in colorectal cancer patients. Ann Surg Oncol 2009; 16:856.
  89. Ley EJ, Vukasin P, Kaiser AM, et al. Delayed rectovaginal fistula: a potential complication of bevacizumab (avastin). Dis Colon Rectum 2007; 50:930.
  90. Liu SV, Gollard R, Iqbal S. Case report of perforation of an ileal neobladder after treatment of rectal cancer with bevacizumab and comment on mechanisms of intestinal perforation associated with bevacizumab. J Clin Pharm Ther 2012; 37:607.
  91. Tol J, Cats A, Mol L, et al. Gastrointestinal ulceration as a possible side effect of bevacizumab which may herald perforation. Invest New Drugs 2008; 26:393.
  92. Tarnawski AS. Cellular and molecular mechanisms of gastrointestinal ulcer healing. Dig Dis Sci 2005; 50 Suppl 1:S24.
  93. Gutin PH, Iwamoto FM, Beal K, et al. Safety and efficacy of bevacizumab with hypofractionated stereotactic irradiation for recurrent malignant gliomas. Int J Radiat Oncol Biol Phys 2009; 75:156.
  94. Cannistra SA, Matulonis UA, Penson RT, et al. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. J Clin Oncol 2007; 25:5180.
  95. Richardson DL, Backes FJ, Hurt JD, et al. Which factors predict bowel complications in patients with recurrent epithelial ovarian cancer being treated with bevacizumab? Gynecol Oncol 2010; 118:47.
  96. Aghajanian C. The role of bevacizumab in ovarian cancer--an evolving story. Gynecol Oncol 2006; 102:131.
  97. Garcia AA, Hirte H, Fleming G, et al. Phase II clinical trial of bevacizumab and low-dose metronomic oral cyclophosphamide in recurrent ovarian cancer: a trial of the California, Chicago, and Princess Margaret Hospital phase II consortia. J Clin Oncol 2008; 26:76.
  98. Nimeiri HS, Oza AM, Morgan RJ, et al. Efficacy and safety of bevacizumab plus erlotinib for patients with recurrent ovarian, primary peritoneal, and fallopian tube cancer: a trial of the Chicago, PMH, and California Phase II Consortia. Gynecol Oncol 2008; 110:49.
  99. Simpkins F, Belinson JL, Rose PG. Avoiding bevacizumab related gastrointestinal toxicity for recurrent ovarian cancer by careful patient screening. Gynecol Oncol 2007; 107:118.
  100. Burger RA, Brady MF, Bookman MA, et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med 2011; 365:2473.
  101. Burger RA, Brady MF, Bookman MA, et al. Risk factors for GI adverse events in a phase III randomized trial of bevacizumab in first-line therapy of advanced ovarian cancer: A Gynecologic Oncology Group Study. J Clin Oncol 2014; 32:1210.
  102. Perren TJ, Swart AM, Pfisterer J, et al. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med 2011; 365:2484.
  103. Aghajanian C, Blank SV, Goff BA, et al. OCEANS: a randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer. J Clin Oncol 2012; 30:2039.
  104. Walraven M, Witteveen PO, Lolkema MP, et al. Antiangiogenic tyrosine kinase inhibition related gastrointestinal perforations: a case report and literature review. Angiogenesis 2011; 14:135.
  105. Flaig TW, Kim FJ, La Rosa FG, et al. Colonic pneumatosis and intestinal perforations with sunitinib treatment for renal cell carcinoma. Invest New Drugs 2009; 27:83.
  106. Hur H, Park AR, Jee SB, et al. Perforation of the colon by invading recurrent gastrointestinal stromal tumors during sunitinib treatment. World J Gastroenterol 2008; 14:6096.
  107. Peters NA, Richel DJ, Verhoeff JJ, Stalpers LJ. Bowel perforation after radiotherapy in a patient receiving sorafenib. J Clin Oncol 2008; 26:2405.
  108. Park SG, Chung CH, Park CY. Colon perforation during sorafenib therapy for advanced hepatocelluar carcinoma. A case report. Tumori 2011; 97:794.
  109. Kindler H, Campbell NP, Wroblewski K, et al. Final results of a University of Chicago phase II consortium trial of sorafenib (SOR) in patients (pts) with imatinib (IM)- and sunitinib (SU)-resistant (RES) gastrointestinal stromal tumors (GIST) (abstract). Data presented at the 2011 ASCO GI Cancers Symposium, January 20-22, San Francisco, CA. Abstract available online at https://ascopubs.org/doi/abs/10.1200/jco.2011.29.15_suppl.10009 (Accessed on April 27, 2020).
  110. Qi WX, Sun YJ, Tang LN, et al. Risk of gastrointestinal perforation in cancer patients treated with vascular endothelial growth factor receptor tyrosine kinase inhibitors: a systematic review and meta-analysis. Crit Rev Oncol Hematol 2014; 89:394.
  111. Diaz JP, Tew WP, Zivanovic O, et al. Incidence and management of bevacizumab-associated gastrointestinal perforations in patients with recurrent ovarian carcinoma. Gynecol Oncol 2010; 116:335.
  112. Shinagare AB, Howard SA, Krajewski KM, et al. Pneumatosis intestinalis and bowel perforation associated with molecular targeted therapy: an emerging problem and the role of radiologists in its management. AJR Am J Roentgenol 2012; 199:1259.
  113. Badgwell BD, Camp ER, Feig B, et al. Management of bevacizumab-associated bowel perforation: a case series and review of the literature. Ann Oncol 2008; 19:577.
  114. Ghatalia P, Je Y, Nguyen PL, et al. Fatigue with vascular endothelial growth factor receptor tyrosine kinase inhibitors and mammalian target of rapamycin inhibitors in patients with renal cell carcinoma (RCC) and other malignancies: A meta-analysis of randomized clinical trials. Crit Rev Oncol Hematol 2015; 95:251.
  115. Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 2011; 378:1931.
  116. Schlumberger M, Tahara M, Wirth LJ, et al. Lenvatinib versus placebo in radioiodine-refractory thyroid cancer. N Engl J Med 2015; 372:621.
  117. Motzer RJ, Nosov D, Eisen T, et al. Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: results from a phase III trial. J Clin Oncol 2013; 31:3791.
  118. Estilo CL, Fornier M, Farooki A, et al. Osteonecrosis of the jaw related to bevacizumab. J Clin Oncol 2008; 26:4037.
  119. Fleissig Y, Regev E, Lehman H. Sunitinib related osteonecrosis of jaw: a case report. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 113:e1.
  120. Nicolatou-Galitis O, Migkou M, Psyrri A, et al. Gingival bleeding and jaw bone necrosis in patients with metastatic renal cell carcinoma receiving sunitinib: report of 2 cases with clinical implications. Oral Surg Oral Med Oral Pathol Oral Radiol 2012; 113:234.
  121. Pakosch D, Papadimas D, Munding J, et al. Osteonecrosis of the mandible due to anti-angiogenic agent, bevacizumab. Oral Maxillofac Surg 2013; 17:303.
  122. Bevacizumab, sunitinib: osteonecrosis of the jaw. Prescrire Int 2011; 20:155.
  123. Van Poznak C. Osteonecrosis of the jaw and bevacizumab therapy. Breast Cancer Res Treat 2010; 122:189.
  124. Greuter S, Schmid F, Ruhstaller T, Thuerlimann B. Bevacizumab-associated osteonecrosis of the jaw. Ann Oncol 2008; 19:2091.
  125. Koch FP, Walter C, Hansen T, et al. Osteonecrosis of the jaw related to sunitinib. Oral Maxillofac Surg 2011; 15:63.
  126. Antonuzzo L, Lunghi A, Giommoni E, et al. Regorafenib Also Can Cause Osteonecrosis of the Jaw. J Natl Cancer Inst 2016; 108.
  127. Nicolatou-Galitis O, Kouri M, Papadopoulou E, et al. Osteonecrosis of the jaw related to non-antiresorptive medications: a systematic review. Support Care Cancer 2019; 27:383.
  128. Mauceri R, Panzarella V, Morreale I, Campisi G. Medication-related osteonecrosis of the jaw in a cancer patient receiving lenvatinib. Int J Oral Maxillofac Surg 2019; 48:1530.
  129. Guarneri V, Miles D, Robert N, et al. Bevacizumab and osteonecrosis of the jaw: incidence and association with bisphosphonate therapy in three large prospective trials in advanced breast cancer. Breast Cancer Res Treat 2010; 122:181.
  130. Lu W, Guo Q, Ma Z, et al. Lenvatinib and osteonecrosis of the jaw: A pharmacovigilance study. Eur J Cancer 2021; 150:211.
  131. Sclafani F, Giuseppe G, Mezynksi J, et al. Reversible posterior leukoencephalopathy syndrome and bevacizumab in breast cancer. J Clin Oncol 2012; 30:e257.
  132. Lou E, Turner S, Sumrall A, et al. Bevacizumab-induced reversible posterior leukoencephalopathy syndrome and successful retreatment in a patient with glioblastoma. J Clin Oncol 2011; 29:e739.
  133. Glusker P, Recht L, Lane B. Reversible posterior leukoencephalopathy syndrome and bevacizumab. N Engl J Med 2006; 354:980.
  134. Ozcan C, Wong SJ, Hari P. Reversible posterior leukoencephalopathy syndrome and bevacizumab. N Engl J Med 2006; 354:980.
  135. Martín G, Bellido L, Cruz JJ. Reversible posterior leukoencephalopathy syndrome induced by sunitinib. J Clin Oncol 2007; 25:3559.
  136. Govindarajan R, Adusumilli J, Baxter DL, et al. Reversible posterior leukoencephalopathy syndrome induced by RAF kinase inhibitor BAY 43-9006. J Clin Oncol 2006; 24:e48.
  137. Chelis L, Souftas V, Amarantidis K, et al. Reversible posterior leukoencephalopathy syndrome induced by pazopanib. BMC Cancer 2012; 12:489.
  138. Dogan E, Aksoy S, Arslan C, et al. Probable sorafenib-induced reversible encephalopathy in a patient with hepatocellular carcinoma. Med Oncol 2010; 27:1436.
  139. Kapiteijn E, Brand A, Kroep J, Gelderblom H. Sunitinib induced hypertension, thrombotic microangiopathy and reversible posterior leukencephalopathy syndrome. Ann Oncol 2007; 18:1745.
  140. Allen JA, Adlakha A, Bergethon PR. Reversible posterior leukoencephalopathy syndrome after bevacizumab/FOLFIRI regimen for metastatic colon cancer. Arch Neurol 2006; 63:1475.
  141. Seet RC, Rabinstein AA. Clinical features and outcomes of posterior reversible encephalopathy syndrome following bevacizumab treatment. QJM 2012; 105:69.
  142. Rini BI, Tamaskar I, Shaheen P, et al. Hypothyroidism in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst 2007; 99:81.
  143. Desai J, Yassa L, Marqusee E, et al. Hypothyroidism after sunitinib treatment for patients with gastrointestinal stromal tumors. Ann Intern Med 2006; 145:660.
  144. Feldt S, Schüssel K, Quinzler R, et al. Incidence of thyroid hormone therapy in patients treated with sunitinib or sorafenib: a cohort study. Eur J Cancer 2012; 48:974.
  145. Torino F, Corsello SM, Longo R, et al. Hypothyroidism related to tyrosine kinase inhibitors: an emerging toxic effect of targeted therapy. Nat Rev Clin Oncol 2009; 6:219.
  146. Mannavola D, Coco P, Vannucchi G, et al. A novel tyrosine-kinase selective inhibitor, sunitinib, induces transient hypothyroidism by blocking iodine uptake. J Clin Endocrinol Metab 2007; 92:3531.
  147. Grossmann M, Premaratne E, Desai J, Davis ID. Thyrotoxicosis during sunitinib treatment for renal cell carcinoma. Clin Endocrinol (Oxf) 2008; 69:669.
  148. Faris JE, Moore AF, Daniels GH. Sunitinib (sutent)-induced thyrotoxicosis due to destructive thyroiditis: a case report. Thyroid 2007; 17:1147.
  149. Sakurai K, Fukazawa H, Arihara Z, Yoshida K. Sunitinib-induced thyrotoxicosis followed by persistent hypothyroidism with shrinkage of thyroid volume. Tohoku J Exp Med 2010; 222:39.
  150. Alexandrescu DT, Popoveniuc G, Farzanmehr H, et al. Sunitinib-associated lymphocytic thyroiditis without circulating antithyroid antibodies. Thyroid 2008; 18:809.
  151. Tamaskar I, Bukowski R, Elson P, et al. Thyroid function test abnormalities in patients with metastatic renal cell carcinoma treated with sorafenib. Ann Oncol 2008; 19:265.
  152. Miyake H, Kurahashi T, Yamanaka K, et al. Abnormalities of thyroid function in Japanese patients with metastatic renal cell carcinoma treated with sorafenib: a prospective evaluation. Urol Oncol 2010; 28:515.
  153. Escudier B, Eisen T, Stadler WM, et al. Sorafenib for treatment of renal cell carcinoma: Final efficacy and safety results of the phase III treatment approaches in renal cancer global evaluation trial. J Clin Oncol 2009; 27:3312.
  154. Robinson SI, Hobday TJ, Sathananthan A, et al. Can sorafenib cause hypothyroidism? J Chemother 2007; 19:352.
  155. Haraldsdottir S, Li Q, Villalona-Calero MA, et al. Case of sorafenib-induced thyroid storm. J Clin Oncol 2013; 31:e262.
  156. Walko CM, Aubert RE, La-Beck NM, et al. Pharmacoepidemiology of Clinically Relevant Hypothyroidism and Hypertension from Sunitinib and Sorafenib. Oncologist 2017; 22:208.
  157. Barbaro D. Sorafenib and thyrotoxicosis. J Endocrinol Invest 2010; 33:436.
  158. van Doorn L, Eskens FA, Visser TJ, et al. Sorafenib induced thyroiditis in two patients with hepatocellular carcinoma. Thyroid 2011; 21:197.
  159. Fujiwara Y, Kiyota N, Chayahara N, et al. Management of axitinib (AG-013736)-induced fatigue and thyroid dysfunction, and predictive biomarkers of axitinib exposure: results from phase I studies in Japanese patients. Invest New Drugs 2012; 30:1055.
  160. Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 2010; 28:1061.
  161. Wu J, Huang H. Acquired Hypothyroidism in Patients with Metastatic Renal Cell Carcinoma Treated with Tyrosine Kinase Inhibitors. Drug Des Devel Ther 2020; 14:3977.
  162. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/212904s000lbl.pdf (Accessed on April 14, 2021).
  163. http://www.accessdata.fda.gov/drugsatfda_docs/label/2015/206947s000lbl.pdf?et_cid=35470087&et_rid=907466112&linkid=http%3a%2f%2fwww.accessdata.fda.gov%2fdrugsatfda_docs%2flabel%2f2015%2f206947s000lbl.pdf (Accessed on February 20, 2015).
  164. Kumar R, Crouthamel MC, Rominger DH, et al. Myelosuppression and kinase selectivity of multikinase angiogenesis inhibitors. Br J Cancer 2009; 101:1717.
  165. Funakoshi T, Latif A, Galsky MD. Risk of hematologic toxicities in cancer patients treated with sunitinib: a systematic review and meta-analysis. Cancer Treat Rev 2013; 39:818.
  166. Heath EI, Blumenschein GR Jr, Cohen RB, et al. Sunitinib in combination with paclitaxel plus carboplatin in patients with advanced solid tumors: phase I study results. Cancer Chemother Pharmacol 2011; 68:703.
  167. van Erp NP, Mathijssen RH, van der Veldt AA, et al. Myelosuppression by sunitinib is flt-3 genotype dependent. Br J Cancer 2010; 103:757.
  168. van Erp NP, Eechoute K, van der Veldt AA, et al. Pharmacogenetic pathway analysis for determination of sunitinib-induced toxicity. J Clin Oncol 2009; 27:4406.
  169. Schutz FA, Je Y, Choueiri TK. Hematologic toxicities in cancer patients treated with the multi-tyrosine kinase sorafenib: a meta-analysis of clinical trials. Crit Rev Oncol Hematol 2011; 80:291.
  170. Smith DC, Smith MR, Sweeney C, et al. Cabozantinib in patients with advanced prostate cancer: results of a phase II randomized discontinuation trial. J Clin Oncol 2013; 31:412.
  171. Boers-Doets CB, Epstein JB, Raber-Durlacher JE, et al. Oral adverse events associated with tyrosine kinase and mammalian target of rapamycin inhibitors in renal cell carcinoma: a structured literature review. Oncologist 2012; 17:135.
  172. Yuan A, Kurtz SL, Barysauskas CM, et al. Oral adverse events in cancer patients treated with VEGFR-directed multitargeted tyrosine kinase inhibitors. Oral Oncol 2015; 51:1026.
  173. Lee JS, Hirsh V, Park K, et al. Vandetanib Versus placebo in patients with advanced non-small-cell lung cancer after prior therapy with an epidermal growth factor receptor tyrosine kinase inhibitor: a randomized, double-blind phase III trial (ZEPHYR). J Clin Oncol 2012; 30:1114.
  174. Leboulleux S, Bastholt L, Krause T, et al. Vandetanib in locally advanced or metastatic differentiated thyroid cancer: a randomised, double-blind, phase 2 trial. Lancet Oncol 2012; 13:897.
  175. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007; 356:115.
  176. Demetri GD, van Oosterom AT, Garrett CR, et al. Efficacy and safety of sunitinib in patients with advanced gastrointestinal stromal tumour after failure of imatinib: a randomised controlled trial. Lancet 2006; 368:1329.
  177. Natale RB, Thongprasert S, Greco FA, et al. Phase III trial of vandetanib compared with erlotinib in patients with previously treated advanced non-small-cell lung cancer. J Clin Oncol 2011; 29:1059.
  178. Raymond E, Dahan L, Raoul JL, et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N Engl J Med 2011; 364:501.
  179. Motzer RJ, Hutson TE, Tomczak P, et al. Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol 2009; 27:3584.
  180. Eisen T, Sternberg CN, Robert C, et al. Targeted therapies for renal cell carcinoma: review of adverse event management strategies. J Natl Cancer Inst 2012; 104:93.
  181. Fischer A, Wu S, Ho AL, Lacouture ME. The risk of hand-foot skin reaction to axitinib, a novel VEGF inhibitor: a systematic review of literature and meta-analysis. Invest New Drugs 2013; 31:787.
  182. Massey PR, Okman JS, Wilkerson J, Cowen EW. Tyrosine kinase inhibitors directed against the vascular endothelial growth factor receptor (VEGFR) have distinct cutaneous toxicity profiles: a meta-analysis and review of the literature. Support Care Cancer 2015; 23:1827.
  183. Belum VR, Serna-Tamayo C, Wu S, Lacouture ME. Incidence and risk of hand-foot skin reaction with cabozantinib, a novel multikinase inhibitor: a meta-analysis. Clin Exp Dermatol 2016; 41:8.
  184. Poprach A, Pavlik T, Melichar B, et al. Skin toxicity and efficacy of sunitinib and sorafenib in metastatic renal cell carcinoma: a national registry-based study. Ann Oncol 2012; 23:3137.
  185. Grothey A, Huang L, Wagner A, et al. Hand-foot skin reaction (HFSR) and outcomes in the phase 3 CORRECT trial of regorafenib for metastatic colorectal cancer (mCRC) (abstract). J Clin Oncol 35, 2017 (suppl; abstr 3551). Abstract available online at http://abstracts.asco.org/199/AbstView_199_186659.html (Accessed on July 25, 2017).
  186. Lammie A, Drobnjak M, Gerald W, et al. Expression of c-kit and kit ligand proteins in normal human tissues. J Histochem Cytochem 1994; 42:1417.
  187. FDA-approved manufacturer's package insert for sorafenib available online at http://dailymed.nlm.nih.gov/dailymed/lookup.cfm?setid=b50667e4-5ebc-4968-a646-d605058dbef0#section-2 (Accessed on January 31, 2013).
  188. Bekaii-Saab TS, Ou FS, Ahn DH, et al. Regorafenib dose-optimisation in patients with refractory metastatic colorectal cancer (ReDOS): a randomised, multicentre, open-label, phase 2 study. Lancet Oncol 2019; 20:1070.
  189. Taguchi D, Inoue M, Fukuda K, et al. Therapeutic drug monitoring of regorafenib and its metabolite M5 can predict treatment efficacy and the occurrence of skin toxicities. Int J Clin Oncol 2020; 25:531.
  190. Ghatalia P, Je Y, Mouallem NE, et al. Hepatotoxicity with vascular endothelial growth factor receptor tyrosine kinase inhibitors: A meta-analysis of randomized clinical trials. Crit Rev Oncol Hematol 2015; 93:257.
  191. Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2007; 356:125.
  192. Cortes JE, Kantarjian H, Shah NP, et al. Ponatinib in refractory Philadelphia chromosome-positive leukemias. N Engl J Med 2012; 367:2075.
  193. Ghatalia P, Morgan CJ, Choueiri TK, et al. Pancreatitis with vascular endothelial growth factor receptor tyrosine kinase inhibitors. Crit Rev Oncol Hematol 2015; 94:136.
  194. http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/021938s027lbl.pdf (Accessed on January 05, 2015).
  195. Billemont B, Medioni J, Taillade L, et al. Blood glucose levels in patients with metastatic renal cell carcinoma treated with sunitinib. Br J Cancer 2008; 99:1380.
  196. Agostino NM, Chinchilli VM, Lynch CJ, et al. 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.
  197. Mailliez A, Baldini C, Van JT, et al. Nasal septum perforation: a side effect of bevacizumab chemotherapy in breast cancer patients. Br J Cancer 2010; 103:772.
  198. Ramiscal JA, Jatoi A. Bevacizumab-induced nasal septal perforation: incidence of symptomatic, confirmed event(s) in colorectal cancer patients. Acta Oncol 2011; 50:578.
  199. http://www.medscape.com/viewarticle/545083 (Accessed on January 31, 2013).
  200. Ramiscal JA, Jatoi A. Nasal septal perforation from bevacizumab: a discussion of outcomes, management, and pharmacovigilance. Curr Oncol Rep 2012; 14:307.
  201. Diamantopoulos II, Jones NS. The investigation of nasal septal perforations and ulcers. J Laryngol Otol 2001; 115:541.
  202. Watson D, Barkdull G. Surgical management of the septal perforation. Otolaryngol Clin North Am 2009; 42:483.
  203. Fakih MG, Lombardo JC. Bevacizumab-induced nasal septum perforation. Oncologist 2006; 11:85.
  204. Ruiz N, Fernandez-Martos C, Romero I, et al. Invasive fungal infection and nasal septum perforation with bevacizumab-based therapy in advanced colon cancer. J Clin Oncol 2007; 25:3376.
  205. Antoun S, Baracos VE, Birdsell L, et al. Low body mass index and sarcopenia associated with dose-limiting toxicity of sorafenib in patients with renal cell carcinoma. Ann Oncol 2010; 21:1594.
  206. Antoun S, Birdsell L, Sawyer MB, et al. Association of skeletal muscle wasting with treatment with sorafenib in patients with advanced renal cell carcinoma: results from a placebo-controlled study. J Clin Oncol 2010; 28:1054.
  207. Mir O, Coriat R, Blanchet B, et al. Sarcopenia predicts early dose-limiting toxicities and pharmacokinetics of sorafenib in patients with hepatocellular carcinoma. PLoS One 2012; 7:e37563.
  208. Poterucha T, Burnette B, Jatoi A. A decline in weight and attrition of muscle in colorectal cancer patients receiving chemotherapy with bevacizumab. Med Oncol 2012; 29:1005.
  209. Parsons HA, Tsimberidou AM, Pontikos M, et al. Evaluation of the clinical relevance of body composition parameters in patients with cancer metastatic to the liver treated with hepatic arterial infusion chemotherapy. Nutr Cancer 2012; 64:206.
  210. Hescot S, Vignaux O, Goldwasser F. Pancreatic atrophy--a new late toxic effect of sorafenib. N Engl J Med 2013; 369:1475.
  211. Shinagare AB, Steele E, Braschi-Amirfarzan M, et al. Sunitinib-associated Pancreatic Atrophy in Patients with Gastrointestinal Stromal Tumor: A Toxicity with Prognostic Implications Detected at Imaging. Radiology 2016; 281:140.
  212. http://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid=f5b7b3a4-c3a4-4722-8ca5-8f6a5c622553 (Accessed on February 21, 2019).
Topic 86366 Version 58.0

References