antiangiogenic treatments and mechanisms of action in renal cell carcinoma

11
REVIEW Antiangiogenic treatments and mechanisms of action in renal cell carcinoma Sylvie Négrier & Eric Raymond Received: 4 March 2011 /Accepted: 26 April 2011 /Published online: 15 May 2011 # Springer Science+Business Media, LLC 2011 Summary Several angiogenic mechanisms are involved in the pathology of renal cell carcinoma (RCC). Increasing knowledge of angiogenesis and the associated signalling pathways has led to the development of targeted antiangio- genic agents for the treatment of metastatic RCC and the introduction of these agents has significantly improved outcomes for these patients. This article provides an overview of the angiogenic mechanisms implicated in RCC, focusing on the main vascular endothelial growth factor (VEGF), platelet-derived growth factor (PDGF) and mammalian target of rapamycin (mTOR) signalling path- ways. Targeted antiangiogenic agents for the treatment of mRCC include receptor tyrosine kinase inhibitors (such as sunitinib, sorafenib, pazopanib, axitinib, cediranib and tivozanib), monoclonal antibodies (such as bevacizumab) and mTOR inhibitors (such as temsirolimus and ever- olimus). In this article, we consider the modes of action of these targeted agents and their differing target receptor profiles and we also evaluate how these correlate with their clinical efficacy and tolerability profiles. Keywords Renal cell carcinoma . Angiogenesis . Targeted therapy . Mode of action . Tolerability Introduction Historically, metastatic renal cell carcinoma (mRCC) has been associated with poor prognosis and resistance to chemother- apy. The introduction of immunotherapy achieved efficacy in a small subgroup of patients but carried toxicity concerns [1, 2]. In recent years, elucidation of angiogenic mechanisms has led to the development of targeted antiangiogenic agents. Targeted agents have greatly improved outcomes in mRCC patients and several have been approved multinationally for the treatment of advanced and/or metastatic RCC. Currently licensed agents for the treatment of mRCC include sunitinib malate (SUTENT®; Pfizer Inc.), temsirolimus (Torisel®; Pfizer Inc.), sorafenib (Nexavar®; Bayer Healthcare AG), bevacizumab (Avastin®; F. Hoffmann-La Roche Ltd) in combination with interferon-alpha (IFN-α), and everolimus (Afinitor®; Novartis Pharmaceuticals). Pazopanib (Votrient®; GlaxoSmithKline) recently received approval in the US and Europe. In addition, a number of novel agentsincluding axitinib, cediranib and tivozanibare under investigation in clinical trials for the treatment of advanced RCC. This paper provides an overview of the angiogenic mechanisms implicated in RCC, and discusses the modes of action of antiangiogenic agents and how these correlate with their clinical profiles. Angiogenic mechanisms underlying the pathology of RCC Several angiogenic mechanisms underlie the pathology of solid tumours. The switch to a pro-angiogenic environment S. Négrier (*) Université de Lyon-Centre Léon Berard, 28, rue Laennec, 69373 Lyon cedex 08, France e-mail: [email protected] E. Raymond Department of Medical Oncology and Laboratory of Pharmacobiology of Anticancer Drugs (RayLab), Bichat-Beaujon University Hospital, Paris, France E. Raymond Hôpital BEAUJON, 100, Boulevard du Général Leclerc, 92118 Clichy Cedex, France Invest New Drugs (2012) 30:17911801 DOI 10.1007/s10637-011-9677-6

Upload: eric-raymond

Post on 26-Aug-2016

221 views

Category:

Documents


6 download

TRANSCRIPT

REVIEW

Antiangiogenic treatments and mechanisms of action in renalcell carcinoma

Sylvie Négrier & Eric Raymond

Received: 4 March 2011 /Accepted: 26 April 2011 /Published online: 15 May 2011# Springer Science+Business Media, LLC 2011

Summary Several angiogenic mechanisms are involved inthe pathology of renal cell carcinoma (RCC). Increasingknowledge of angiogenesis and the associated signallingpathways has led to the development of targeted antiangio-genic agents for the treatment of metastatic RCC and theintroduction of these agents has significantly improvedoutcomes for these patients. This article provides anoverview of the angiogenic mechanisms implicated inRCC, focusing on the main vascular endothelial growthfactor (VEGF), platelet-derived growth factor (PDGF) andmammalian target of rapamycin (mTOR) signalling path-ways. Targeted antiangiogenic agents for the treatment ofmRCC include receptor tyrosine kinase inhibitors (such assunitinib, sorafenib, pazopanib, axitinib, cediranib andtivozanib), monoclonal antibodies (such as bevacizumab)and mTOR inhibitors (such as temsirolimus and ever-olimus). In this article, we consider the modes of action ofthese targeted agents and their differing target receptorprofiles and we also evaluate how these correlate with theirclinical efficacy and tolerability profiles.

Keywords Renal cell carcinoma . Angiogenesis . Targetedtherapy .Mode of action . Tolerability

Introduction

Historically, metastatic renal cell carcinoma (mRCC) has beenassociated with poor prognosis and resistance to chemother-apy. The introduction of immunotherapy achieved efficacy ina small subgroup of patients but carried toxicity concerns [1,2]. In recent years, elucidation of angiogenic mechanismshas led to the development of targeted antiangiogenic agents.Targeted agents have greatly improved outcomes in mRCCpatients and several have been approved multinationally forthe treatment of advanced and/or metastatic RCC. Currentlylicensed agents for the treatment of mRCC include sunitinibmalate (SUTENT®; Pfizer Inc.), temsirolimus (Torisel®;Pfizer Inc.), sorafenib (Nexavar®; Bayer Healthcare AG),bevacizumab (Avastin®; F. Hoffmann-La Roche Ltd) incombination with interferon-alpha (IFN-α), and everolimus(Afinitor®; Novartis Pharmaceuticals). Pazopanib (Votrient®;GlaxoSmithKline) recently received approval in the US andEurope. In addition, a number of novel agents—includingaxitinib, cediranib and tivozanib—are under investigation inclinical trials for the treatment of advanced RCC.

This paper provides an overview of the angiogenicmechanisms implicated in RCC, and discusses the modes ofaction of antiangiogenic agents and how these correlatewith their clinical profiles.

Angiogenic mechanisms underlying the pathologyof RCC

Several angiogenic mechanisms underlie the pathology ofsolid tumours. The switch to a pro-angiogenic environment

S. Négrier (*)Université de Lyon-Centre Léon Berard,28, rue Laennec,69373 Lyon cedex 08, Francee-mail: [email protected]

E. RaymondDepartment of Medical Oncology and Laboratory ofPharmacobiology of Anticancer Drugs (RayLab),Bichat-Beaujon University Hospital,Paris, France

E. RaymondHôpital BEAUJON,100, Boulevard du Général Leclerc,92118 Clichy Cedex, France

Invest New Drugs (2012) 30:1791–1801DOI 10.1007/s10637-011-9677-6

can be induced by tumour-associated hypoxia, the activationof oncogenes, the inactivation of tumour-suppressor genes,and the secretion of a number of growth factors and cytokines[3, 4]. The pathways involved in RCC development includemainly the vascular endothelial growth factor (VEGF),platelet-derived growth factor (PDGF) and mammalian targetof rapamycin (mTOR) signalling pathways (Fig. 1) [5–7].

Vascular endothelial growth factor

VEGF expression is induced under hypoxic conditions [8]triggering several mechanisms that promote angiogenesis[9]. Members of the VEGF family regulate angiogenesisthrough binding to the related family of receptor tyrosinekinases (RTKs): VEGF receptors (VEGFR)-1, -2 and -3.The pro-angiogenic mechanisms of the VEGF signalingpathway have been well documented and are beyond thescope of this paper; readers are referred to a review by Ellisand Hicklin for a detailed discussion of this topic [9].

In RCC, VEGF is also a powerful tumour growth factor.Renal carcinoma cells over-express the different VEGFreceptors and also produce, as paracrine and autocrinegrowth factors, large amounts of VEGF [10].

Platelet-derived growth factor

The PDGF family (PDGF-A, -B, -C and -D) mediate theireffects through binding to the RTKs PDGF receptor-alpha

and -beta (PDGFR-α, PDGFR-β), leading to theactivation of intracellular signalling pathways that canpromote tumour growth [11, 12] Additionally, PDGFR-βis thought to be involved in the recruitment of pericytes tocapillaries [12]. Pericytes are required for microvascularstability and are important for maintaining tumourvasculature [13].

Few data have been published on PDGF and PDGFR inRCC. However, human RCC has been shown to expresshigh levels of PDGF-D, and PDGF-D over-expressionpromotes tumour growth, angiogenesis and metastasis inRCC [14]. Studies have also shown a relationship betweenRCC progression and PDGF-D/PDGFR-β signalling [14]and PDGFR-α expression [15].

Mammalian target of rapamycin

The mTOR pathway, including its role in RCC, has beenreviewed in several publications, to which readers arereferred for a more detailed discussion [16, 17] Hypoxia-induced activation of the mTOR pathway induces theexpression of several vascular growth factors, includingVEGF, VEGFR and PDGF, thus promoting tumourangiogenesis and endothelial proliferation [17, 18]. Inaddition to activation of the VEGF pathway, mTOR islargely involved in the AKT pathway, which is alsoderegulated in a number of tumour types including RCC[18, 19].

Fig. 1 Angiogenic pathways contributing to RCC growth and development, and the targeted agents that inhibit them [5–7]

1792 Invest New Drugs (2012) 30:1791–1801

Modes of action of antiangiogenic agentsfor the treatment of mRCC

A number of antiangiogenic agents are in use or underinvestigation for the treatment of mRCC. Many of theseagents are multitargeted, inhibiting a range of targetsinvolved in tumour growth and angiogenesis (see Fig. 1)[5–7].

RTK inhibitors

The RTK inhibitors—sunitinib, sorafenib, pazopanib, axi-tinib, cediranib and tivozanib—inhibit multiple angiogenicpathways including the RTKs VEGFR and PDGFR and theRaf serine/threonine kinases (see Table 1) [20–34]. Theseagents share some similarities in their efficacy andtolerability profiles, as might be expected from their similarmodes of action. The aspects in which they differ mayreflect their varying target receptor profiles.

Sunitinib Sunitinib inhibits RTK autophosphorylation andligand-induced cell proliferation. It is thought that by inhibit-ing both VEGFR and PDGFR, ‘dual’ attack on the vasculature(via endothelial cells and pericytes) is achieved, thus leadingto greater antiangiogenic activity [9]. Oral sunitinib isapproved multinationally for the first- and second-linetreatment of advanced and/or metastatic RCC [35].

Sorafenib Sorafenib inhibits VEGFR, PDGFR, FMS-liketyrosine kinase 3 (Flt-3), c-Kit and RET receptor tyrosine

kinases. A study in RCC models reported significanttumour growth inhibition and a reduction in tumourvasculature with sorafenib, and antiangiogenic activitycorrelated with an increased level of tumour apoptosis andcentral necrosis [36]. Oral sorafenib is approved for thetreatment of advanced RCC in the US and for the treatmentof advanced RCC after cytokine failure in Europe [37].

Pazopanib Pazopanib inhibits VEGFR, PDGFR and c-Kit.In contrast to sunitinib and sorafenib (but similar toaxitinib), pazopanib demonstrates inhibitory activity atlow nanomolar concentrations [24, 28]. Studies in murinemodels have shown that pazopanib inhibits angiogenesisand tumour growth in a broad range of tumour types,including RCC [28, 38]. Pazopanib has recently beenapproved in the US for the treatment of advanced RCC[39] and has received conditional approval in Europe.

Axitinib Axitinib is a potent small-molecule RTK inhibitor;it inhibits VEGFRs at subnanomolar concentrations, andPDGFR-β and KIT at low nanomolar concentrations [29].Axitinib dose-dependently blocks VEGF-stimulated recep-tor autophosphorylation, leading to the inhibition ofendothelial cell proliferation and survival [29]. Axitinibalso reduces the extent of VEGF-mediated endothelial cellmigration and tube formation [29] Studies in mouse modelshave demonstrated the antitumour, antiangiogenic and anti-metastatic activity of axitinib and its ability to inducecentral necrosis [29, 40]. Axitinib is under investigation forthe second-line treatment of advanced RCC.

Table 1 Principal receptor targets of the antiangiogenic receptor tyrosine kinase inhibitors with activity in renal cell carcinoma

Receptor target

VEGFR-1

VEGFR-2

VEGFR-3

PDGFR-α

PDGFR-β

KIT FLT1 FLT3 FLT4 CSF-1R

RET Raf FGFR2

Approved agents

Sunitinib[20, 23,25–27, 35]

√ √ √ √ √ √ √ √ √

Sorafenib[21, 31]

√ √ √ √ √ √ √

Pazopanib[24, 28, 33]

√ √ √ √ √ √

Novel agents

Axitinib[29]

√ √ √ √ √

Cediranib[22, 30, 32]

√ √ √ √ √ √ √ √

Tivozanib[34]

√ √ √ √ √

VEGFR vascular endothelial growth factor receptor; PDGFR platelet derived growth factor receptor; KIT stem-cell factor receptor; FLT3 FMS-like tyrosine kinase-3 receptor, CSF-1R colony-stimulating factor 1 receptor; RET glial cell-line derived neurotrophic factor receptor (Rearrangedduring Transfection); FGFR2 fibroblast growth factor receptor 2

Invest New Drugs (2012) 30:1791–1801 1793

Cediranib Cediranib (AZD2171) is an ATP-competitiveinhibitor of RTKs and, like axitinib, is potent atsubnanomolar concentrations [22, 30, 32]. Cediranibprevents VEGF-induced angiogenesis and exhibits dose-dependent antitumor activity through the prevention ofnew vessel formation and vascular regression [22, 32].Cediranib is under investigation for the treatment ofadvanced RCC.

Tivozanib Tivozanib (AV-951) is an RTK inhibitor, whichinhibits VEGFR-1, VEGFR-2 and VEGFR-3 at picomolarconcentrations. In xenograft tumour models, tivozanib hasbeen shown to inhibit tumour growth and angiogenesis[41]. Tivozanib is currently under investigation for the first-and second-line treatment of advanced RCC.

Monoclonal antibodies

Bevacizumab Bevacizumab, a recombinant humanisedmonoclonal antibody, binds directly to all biologicallyactive forms of VEGF [7] and shares some similaritieswith the RTK inhibitors in terms of clinical activity.Studies using xenograft models of a range of tumour typeshave shown dose-dependent inhibition of primary tumourgrowth with bevacizumab [42]. Bevacizumab has no effecton rates of tumour cell proliferation, supporting thehypothesis that this agent targets endothelial cell proliferationand disrupts neovascularisation [42, 43]. Bevacizumab isapproved multinationally, in combination with IFN-α, forthe first-line treatment of advanced and/or metastatic RCC[44].

mTOR inhibitors

Temsirolimus and everolimus are derivatives of the immu-nosuppressant agent rapamycin and both rapalogues inhibitthe mTOR angiogenic pathway. Preclinical data in RCCsuggest that the antitumour activity of mTOR inhibitorsmay be the result of a combination of two differentmechanisms: direct cytotoxic activity and indirect anti-angiogenic activity [17]. The cytotoxic activity of therapalogues against renal carcinoma cells is poor and dosedependent. In clinical trials with temsirolimus, toxicitylimited use of the higher doses tested using preclinicalmodels [17]. Antiangiogenic activity may be exerted byinterfering with the maintenance of endothelial cells andpericytes that are required for tumour angiogenesis [17]. Itis not clear which of these mechanisms predominates in theactivity of mTOR inhibitors in RCC, but it seems likely thatat current recommended doses, the efficacy of temsirolimusis mainly due to antiangiogenic activity.

Temsirolimus Temsirolimus forms a complex with theintracellular protein FKBP-12, and this protein-drug com-plex inhibits the activity of mTOR [17]. Temsirolimus,administered by intravenous injection, is approved inEurope as a first-line therapy for mRCC patients with poorprognosis and is approved in the US for the treatment ofmRCC [45].

Everolimus Everolimus also forms a complex with FKBP-12to inhibit mTOR and downstream signalling events. This inturn leads to growth retardation and antiangiogenesis, andaccumulation of cells in the G1 phase of the cell cycle [46].Everolimus has been shown to be effective in patients withmRCC after failure of sunitinib and/or sorafenib [47] and hasrecently been approved in Europe and the US for use in thissetting [48].

Antiangiogenic agents for mRCC in clinical practice

In the clinical setting, targeted agents for advanced RCCexhibit some similarities but also a number of differences intheir efficacy and tolerability profiles. These may beexplained, at least in part, by their modes of action anddiffering target receptor profiles.

Efficacy

The targeted agents have not been compared directly inclinical trials and comparisons between trials should bemade with caution. In addition, it is important to note thatthe trials with targeted agents included different patientpopulations, for example, some were conducted incytokine-refractory patients, while another trial enrolledpatients with poor prognosis mRCC [49, 50]. Nevertheless,data from phase III trials in advanced RCC, in both thefirst- and second-line settings, highlight some interestingobservations (Table 2) [47, 49–55].

Objective response rates In phase III clinical trials, theobjective response rates (ORR) achieved with agents thattarget the VEGF signalling pathway—i.e., sunitinib (39%),pazopanib (30%), bevacizumab plus IFN-α (26–31%) and,to a lesser extent, sorafenib (10%)—were higher than thoseassociated with agents that target the mTOR pathway(temsirolimus and everolimus [each <9%]; Table 2) [47,49–55]. It has been postulated that loss of VEGF signalling,with the associated endothelial cell apoptosis and vesselgrowth inhibition, may occur with VEGF-targeted thera-pies, such as sunitinib, pazopanib and bevacizumab/IFN-α,which are associated with higher response rates [9]. Thelower ORR achieved with sorafenib may be due to the fact

1794 Invest New Drugs (2012) 30:1791–1801

that this agent was evaluated in the second-line setting (inpatients in whom previous therapy had failed). It could alsobe linked to the number of pathways (signal transduction)that are blocked by the drug, as illustrated by Karaman et alin a recent global analysis of multi-kinase activity [56].Additionally, it has been suggested that the lower ORR maybe due to the weaker binding affinity of sorafenib toVEGFR-2 and PDGFR-β compared with that of sunitinib[57].

Of note, preliminary data from phase II clinical trialsevaluating the efficacy of novel targeted agents in mRCCsuggest that these agents may have the potential to achievehigher ORRs than their older counterparts (Table 3) [58–60]. In the axitinib study in patients with cytokine-refractory RCC, the authors postulated that the level ofantitumour activity with axitinib (ORR 44%) may be due toits high potency against VEGFR1–3 [60].

Progression-freeandoverall survival Sunitinib (11.0 months),pazopanib (all patients: 9.2 months) and bevacizumab plusIFN-α (8.5–10.2 months) were associated with longermedian progression-free survival (PFS) than sorafenib(5.5 months) or the mTOR inhibitors (4.9–5.5 months) inclinical trials in mRCC (Table 2) [47, 49–55, 61]. Of note,the phase III randomised placebo-controlled study ofeverolimus was undertaken in patients with mRCC whohad progressed following other VEGF-targeted therapies[47]. The authors postulated that the efficacy of everolimusin this heavily pretreated population may have been due tothe distinct mechanism of action of mTOR inhibitors, and alack of clinical cross-resistance with VEGF inhibitors.

A meta-analysis evaluated investigator-assessed PFS inrandomised controlled trials of sunitinib, sorafenib, bev-acizumab and temsirolimus [62]. A total of five trials withtargeted agents as first-line therapy used IFN-α as thecomparator: the phase III sunitinib trial, the two phase IIItrials assessing bevacizumab plus IFN-α, the phase IIItemsirolimus trial in poor prognosis patients and thesorafenib phase II trial. Using indirect comparisons, withIFN-α as the common comparator, sunitinib was superior toboth sorafenib (HR: 0.58, 95% CI: 0.38–0.86, p<0.001)and bevacizumab plus IFN-α (HR: 0.75, 95% CI: 0.60–0.93, p=0.001), while sorafenib was not statisticallydifferent from bevacizumab plus IFN-α (HR:0.77, 95%CI: 0.52–1.13, p=0.21). Two trials used placebo as thecomparator: a phase II trial with bevacizumab alone incytokine-refractory patients and a phase III trial withsorafenib in cytokine-refractory patients. Using placebo asthe similar comparator, the authors found no significantdifference between sorafenib and bevacizumab alone (HR:0.81, 95% CI: 0.58–1.12, p=0.23). Temsirolimus providedsignificant PFS benefit in poor-prognosis patients (HR:0.69, 95% CI: 0.57–0.85).

In the first-line setting, sunitinib was associated withmedian overall survival (OS) of more than 2 years (Table 2)[54]. This represents the first time that this has beenachieved first-line in mRCC. Furthermore, when patientswho had received additional subsequent therapies wereomitted from the analysis, median OS was 28.1 monthswith sunitinib versus 14.1 months with IFN-α (p=0.0033)[54]. Median OS with bevacizumab plus IFN-α was 18.3and 23.3 months versus 17.4 and 21.3 months with IFN-α

Table 2 Efficacy data from phase III trials of targeted agents in the first- and second-line treatment of mRCC

Agent/comparator Setting N Median PFS, months Median OS, months ORR, %

Sunitinib vs. IFN-α [51, 54] First-line 750 11.0 vs. 5.1a 26.4 vs. 21.8 39 vs. 8a

Sorafenib vs. placebo [50] Second-line 903 5.5 vs. 2.8a 17.8 vs. 15.2 10 vs. 2a

Pazopanib vs. placebo [55, 65] First- and second-line 435 9.2 vs. 4.2a 22.9 vs. 20.5 30 vs. 3a

Bevacizumab+IFN-α vs. IFN-α [53, 63] First-line 649 10.2 vs. 5.4a 23.3 vs. 21.3 31 vs. 13a

Bevacizumab+IFN-α vs. IFN-α [52, 64] First-line 732 8.5 vs. 5.2a 18.3 vs. 17.4 26 vs. 13a

Temsirolimus vs. IFN-α [49] First-line 626 5.5 vs. 3.1a 10.9 vs. 7.3a 8.6 vs. 4.8

Everolimus vs. placebo [47, 61] Second-line 410 4.9 vs. 1.9a 14.8 vs. 14.4 1 vs. 0

PFS progression-free survival; OS overall survival; ORR objective response rate; IFN-α interferon-alpha; Bev bevacizumab; NR not reacheda Statistically significant

Targeted agent Patient population Response rate, % Disease control rate, %

Axitinib [60] Cytokine-refractory mRCC 44 69

Axitinib [59] Sorafenib-refractory mRCC 23 40

Cediranib [58] Treatment-naïve advanced RCC 38 84

Tivozanib [72] No prior VEGF-targeted therapy 27 84

Table 3 Response rates fromphase II clinical trials of axitiniband cediranib

Invest New Drugs (2012) 30:1791–1801 1795

alone (p=0.097 and p=0.1291) [63, 64]. Recently pre-sented data showed that median OS was 22.9 months withpazopanib in both treatment-naïve patients and those withcytokine-refractory disease versus 20.5 months with placebo(p=0.224). However, the OS analysis may have beenconfounded by frequent crossover of patients from theplacebo group to the pazopanib group [65].

Median OS achieved with sorafenib in the second-linesetting was 17.8 months versus 15.2 months with placebo(p=0.146) [66]. When these data were censored for patientswho switched from placebo to sorafenib, median OS was17.8 months with sorafenib versus 14.3 months withplacebo (p=0.0287) [66]. Median OS with everolimus inpatients who had progressed on VEGF-targeted therapieswas 14.8 months versus 14.4 months with placebo (p=0.162)[61]. Similar to other trials, patient crossover from placebo toeverolimus may have confounded the OS data [61].

In poor-prognosis patients, temsirolimus achieved medi-an OS of 10.9 months versus 7.3 months with IFN-α (p=0.008) [49]. A post-hoc analysis from the phase III ARCCstudy [49] indicated that temsirolimus also improves OS inpatients with histologies other than clear-cell RCC (themajority of these patients had papillary histologies),suggesting that it can be used for the treatment of all typesof RCC [67]. This finding could be explained by thedifference in carcinogenesis between papillary and clear-cell renal tumours. The importance of VEGF-driven growthin clear-cell tumours easily explains their sensitivity to anti-VEGF agents [68] whereas this mechanism is not predom-inant in the papillary subtype [69, 70]. On the contrary,papillary tumours over-express cMet and PTEN, two genesthat are closely related to mTOR activation status [71].

As noted for ORR, PFS and OS rates achieved withinvestigational drugs in phase II trials were longer thanthose achieved with more established agents in this class.Median OS with axitinib in cytokine-refractory mRCC was30 months, with a 1-year survival rate of 79% [60]. MedianPFS with cediranib in treatment-naïve patients was8.7 months, reflecting findings with comparable targetedagents in mRCC [58]. Similarly, tivozanib was associatedwith median PFS of 11.8 months in a phase II trial whichenrolled patients who had received no prior antiangiogenictherapy [72]. Further data from randomised phase III trialsare required to support these findings. Whilst direct compar-isons between agents are problematic due to inter-trialdifferences, these observations are interesting nonetheless.

Tolerability of the antiangiogenic agents in clinical practice

The tolerability profiles of the targeted agents bear anumber of similarities, as might be expected from theirmechanisms of action, but also some potentially clinically

significant differences (Table 4) [47, 49–55]. Adverseevents (AEs) that are believed to be due to VEGF inhibitioninclude hypertension [73, 74], haemorrhage [75], mucositis,skin toxicity (including hand–foot syndrome), hypothyroid-ism [76–79], and fatigue [80]. AEs attributable to theinhibition of PDGF include skin reactions [81]. Inhibitionof mTOR has been associated with a distinct pattern of skintoxicity (acne-like and maculopapular rashes) [82, 83].Specific AEs of particular interest are discussed furtherbelow.

Hypertension Anti-VEGF activity has been associated withhypertension, and clinical trials with the targeted agentssunitinib, sorafenib, bevacizumab, axitinib, pazopanib,cediranib and tivozanib have all shown incidences oftreatment-related hypertension [35, 37–39, 44, 52, 72, 84].

Several studies with VEGF inhibitors have demonstratedreduced tumour perfusion after treatment administration[85, 86], possibly representing vasoconstriction as a resultof decreased production of vasodilatory mediators. Suchvasoconstriction could lead to tumour ischemia andnecrosis, and could be important in RCC. As the vasocon-strictive effects of VEGF inhibitors are not limited to thetumour vasculature, researchers have postulated that hyper-tension may represent a surrogate marker of antitumouractivity in RCC, and preliminary data support this hypothesis[9]. Indeed, blood pressure was shown to be associated withlonger OS in a retrospective analysis of data from sixaxitinib studies [87]. Furthermore, a significant associationbetween hypertension and improved clinical outcome withsunitinib has been shown, suggesting that this may be a drugclass effect for tyrosine kinase inhibitors [88].

Haemorrhage/necrosis Both VEGF and mTOR inhibitionhave been associated with bleeding events and rarely withtumour haemorrhages in patients with mRCC [35, 37, 39,44, 48]. Bleeding events may manifest as epistaxis,hemoptysis, or rectal, gingival, upper gastrointestinal,genital, or wound bleeding. VEGF plays an important rolein maintaining vascular integrity by enhancing proliferationand survival of endothelial cells. Inhibition of VEGF maytherefore reduce the regeneration of endothelial cellsfollowing trauma, thus increasing the risk of bleeding [73].

Hand-foot syndrome Hand–foot syndrome is reportedcommonly in patients receiving treatment with sunitiniband, particularly, sorafenib [35, 37]. Neither the VEGF-binding agent bevacizumab nor the mTOR inhibitorstemsirolimus and everolimus are associated with this typeof toxicity [44, 45, 48].

Fatigue Fatigue is common in cancer patients treated withtargeted agents, but may also be due to disease and other co-

1796 Invest New Drugs (2012) 30:1791–1801

morbidities such as hypothyroidism, anaemia and depression.VEGF is known to be involved in thyroid functioning but it isnot known if inhibition leads to hypothyroidism and causesfatigue [23, 77, 89]. The development of hypothyroidismduring sunitinib and sorafenib treatment has been shown tobe an independent predictor of survival and may be a usefulas a clinical predictor of PFS [90].

Fatigue is commonly observed with the tyrosine kinaseinhibitors, sunitinib and sorafenib [50, 51, 54]. Grade 2–3fatigue has been reported with everolimus but not withtemsirolimus [91]. The potential relationship betweenmTOR inhibition and fatigue is not clear.

Cardiotoxicity Varying degrees of cardiotoxicity have beenreported with the tyrosine kinase inhibitors sunitinib andsorafenib [92]. HIF-1—a target for these agents—has beenshown to slow the progression of myocardial dysfunctionafter myocardial infarction, and inhibition may thereforeaffect cardiac function [93–95].

Pneumonitis Pneumonitis has been reported with bothtemsirolimus and everolimus, with a higher incidenceobserved in patients receiving everolimus [45, 47]. Themechanisms involved in the development of pneumonitisduring treatment with temsirolimus and everolimus havenot yet been determined.

Conclusions and future perspectives

Historically, mRCC has been associated with treatmentresistance and poor prognosis. However, increasing knowl-edge of angiogenesis and associated signalling pathways,and the subsequent development of antiangiogenic thera-pies have exerted a substantial impact on outcomes forpatients with mRCC. The targeted antiangiogenic agentssunitinib, sorafenib, pazopanib, bevacizumab (in combinationwith IFN-α), temsirolimus and everolimus are approved forthe treatment of advanced RCC. Each agent is unique in termsof its antiangiogenic and antitumor activities and the receptortargets with which it interacts, resulting in unique efficacy andsafety profiles. In addition, a number of novel agents are indevelopment, including axitinib, cediranib and tivozanib, withpreliminary data suggesting substantial antitumour activity inmRCC.

Ongoing and future trials will aim to further characterisethe efficacy and tolerability profiles of the approved andnovel agents in mRCC, investigating their activity acrossdifferent patient profiles and in combination and insequence in order to optimise patient management in thissetting. A number of clinical studies are in progress toinvestigate the efficacy and safety of such strategies. A lackof cross resistance between the targeted antiangiogenicinhibitors has been observed, suggesting that sequential

Table 4 Incidence and severity of commonly reported adverse events associated with targeted agents for the treatment of mRCC

Selected toxicity Percentage incidence for each targeted agent, all grades (grades 3–4)a

Sunitinib[54]

Sorafenib[50]

Pazopanib[55]

Bevacizumab+IFN-α[53]

Temsirolimus[49]

Everolimus[47]

Arterial hypertension 30 (12) 17 (4) 40 (4) 26 (3) – –

Asthenia 20 (7) – 14 (3) 32 (10) 51 (11) 18

Bleeding/Epistaxis 18 (1) – – 33 (3) – –

Diarrhea 61 (9) 43 (2) 52 (4) 20 (2) 27 (1) 17

Fatigue 54 (11) 37 (5) 19 (2) 33 (12) – 20 (3)

Hand–foot syndrome 29 (9) 30 (6) – – – –

Mucositis 26 (2) – – – – 14

Pneumonitis – – – – – 8 (3)

Hematological abnormalities/renal toxicities

Anemia 79 (8) 8 (3) – 10 (3) 45 (20) 91 (9)

Leucopenia 78 (8) – 37 (0) – 6 (1) 26 (0)

Neutropenia 77 (18) – 34 (1) 7 (4) 7 (3) 11 (0)

Thrombocytopenia 68 (9) – 32 (1) 6 (2) 14 (1) 20 (<1)

Proteinuria – – – 18 (7) – –

Increased creatinine 70 (<1) – – – 14 (3) 46 (<1)

Increased creatinekinase

49 (3) – – – – –

NR not reporteda Reported in ≥10% of patients for sunitinib, pazopanib and everolimus; ≥10% for grade 1–2, ≥2% for grade 3–4 for sorafenib; ≥10 of and ≥30%for clinical laboratory abnormalities for pazopanib; ≥2% for bevacizumab+IFN-α; ≥20% for temsirolimus

Invest New Drugs (2012) 30:1791–1801 1797

therapy may be effective [96]. Several trials, including theAGILE 1032 trial comparing axitinib and sorafenib assecond-line treatment and the 404 study comparing temsir-olimus and sorafenib in patients who have progressedfollowing first-line sunitinib, are ongoing and may providefurther information regarding optimal sequencing of agents[97]. In contrast, previous studies have suggested thatcombination treatments are not always well tolerated [98].However, the simultaneous inhibition of both VEGF andmTOR with bevacizumab and temsirolimus is beingassessed in an ongoing trial [97].

Acknowledgements Editorial support for this manuscript wasprovided by Cherry Bwalya and Caroline Masterman at ACUMED(Tytherington, UK) and was funded by Pfizer, Inc.

References

1. Motzer RJ, Bander NH, Nanus DM (1996) Renal-cell carcinoma.N Engl J Med 335:865–875

2. Motzer RJ, Bacik J, Schwartz LH, Reuter V, Russo P, Marion S,Mazumdar M (2004) Prognostic factors for survival in previouslytreated patients with metastatic renal cell carcinoma. J Clin Oncol22:454–463

3. Hanahan D, Folkman J (1996) Patterns and emerging mechanismsof the angiogenic switch during tumorigenesis. Cell 86:353–364

4. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell100:57–70

5. Duensing A, Heinrich MC, Fletcher CD, Fletcher JA (2004)Biology of gastrointestinal stromal tumors: KIT mutations andbeyond. Cancer Invest 22:106–116

6. Marmor MD, Skaria KB, Yarden Y (2004) Signal transductionand oncogenesis by ErbB/HER receptors. Int J Radiat Oncol BiolPhys 58:903–913

7. Rini BI, Small EJ (2005) Biology and clinical development ofvascular endothelial growth factor-targeted therapy in renal cellcarcinoma. J Clin Oncol 23:1028–1043

8. Shweiki D, Itin A, Soffer D, Keshet E (2008) Vascular endothelialgrowth factor induced by hypoxia may mediate hypoxia-initiatedangiogenesis. Nature 359:(abstr)

9. Ellis LM, Hicklin DJ (2008) VEGF-targeted therapy: mechanismsof anti-tumour activity. Nat Rev Cancer 8:579–591

10. Qian CN, Huang D, Wondergem B, Teh BT (2009) Complexity oftumor vasculature in clear cell renal cell carcinoma. Cancer115:2282–2289

11. Guo P, Hu B, Gu W, Xu L, Wang D, Huang HJ, Cavenee WK,Cheng SY (2003) Platelet-derived growth factor-B enhancesglioma angiogenesis by stimulating vascular endothelial growthfactor expression in tumor endothelia and by promoting pericyterecruitment. Am J Pathol 162:1083–1093

12. Pietras K, Sjoblom T, Rubin K, Heldin CH, Ostman A (2003)PDGF receptors as cancer drug targets. Cancer Cell 3:439–443

13. Pietras K, Hanahan D (2005) A multitargeted, metronomic, andmaximum-tolerated dose “chemo-switch” regimen is antiangiogenic,producing objective responses and survival benefit in a mouse modelof cancer. J Clin Oncol 23:939–952

14. Xu L, Tong R, Cochran DM, Jain RK (2005) Blocking platelet-derived growth factor-D/platelet-derived growth factor receptorbeta signaling inhibits human renal cell carcinoma progression inan orthotopic mouse model. Cancer Res 65:5711–5719

15. Sulzbacher I, Birner P, Traxler M, Marberger M, Haitel A (2003)Expression of platelet-derived growth factor-alpha alpha receptoris associated with tumor progression in clear cell renal cellcarcinoma. Am J Clin Pathol 120:107–112

16. Bousquet G, Dreyer C, Faivre RE (2007) TOR (target ofrapamycin) as an anti-cancer target. Drug Discov Today: TherStrateg 4:211–217

17. Le Tourneau C, Faivre S, Serova M, Raymond E (2008) mTORC1inhibitors: is temsirolimus in renal cancer telling us how theyreally work? Br J Cancer 99:1197–1203

18. Cho D, Signoretti S, Regan M, Meir JW, Atkins MB (2007) Therole of mammalian target of rapamycin inhibitors in the treatmentof advanced renal cancer. Clin Cancer Res 13: (abstr)

19. Velickovic M, Delahunt B, McIver B, Grebe SK (2002) IntragenicPTEN/MMAC1 loss of heterozygosity in conventional (clear-cell)renal cell carcinoma is associated with poor patient prognosis.Mod Pathol 15:479–485

20. Abrams TJ, Lee LB, Murray LJ, Pryer NK, Cherrington JM(2003) SU11248 inhibits KIT and platelet-derived growth factorreceptor beta in preclinical models of human small cell lungcancer. Mol Cancer Ther 2:471–478

21. Beeram M, Patnaik A, Rowinsky EK (2005) Raf: a strategic targetfor therapeutic development against cancer. J Clin Oncol23:6771–6790

22. Gomez-Rivera F, Santillan-Gomez AA, Younes MN, Kim S,Fooshee D, Zhao M, Jasser SA, Myers JN (2007) The tyrosinekinase inhibitor, AZD2171, inhibits vascular endothelial growthfactor receptor signaling and growth of anaplastic thyroid cancer inan orthotopic nude mouse model. Clin Cancer Res 13:4519–4527

23. Kim DW, Jo YS, Jung HS, Chung HK, Song JH, Park KC, ParkSH, Hwang JH, Rha SY, Kweon GR, Lee SJ, Jo KW, Shong M(2006) An orally administered multitarget tyrosine kinase inhibitor,SU11248, is a novel potent inhibitor of thyroid oncogenic RET/papillary thyroid cancer kinases. J Clin Endocrinol Metab 91:4070–4076

24. Hurwitz HI, Dowlati A, Saini S, Savage S, Suttle AB, GibsonDM, Hodge JP, Merkle EM, Pandite L (2009) Phase I trial ofpazopanib in patients with advanced cancer. Clin Cancer Res15:4220–4227

25. Mendel DB, Laird AD, Xin X, Louie SG, Christensen JG, Li G,Schreck RE, Abrams TJ, Ngai TJ, Lee LB, Murray LJ, Carver J,Chan E, Moss KG, Haznedar JO, Sukbuntherng J, Blake RA, SunL, Tang C, Miller T, Shirazian S, McMahon G, Cherrington JM(2003) In vivo antitumor activity of SU11248, a novel tyrosinekinase inhibitor targeting vascular endothelial growth factor andplatelet-derived growth factor receptors: determination of apharmacokinetic/pharmacodynamic relationship. Clin Cancer Res9:327–337

26. Murray LJ, Abrams TJ, Long KR, Ngai TJ, Olson LM, Hong W,Keast PK, Brassard JA, O’Farrell AM, Cherrington JM, Pryer NK(2003) SU11248 inhibits tumor growth and CSF-1R-dependentosteolysis in an experimental breast cancer bone metastasis model.Clin Exp Metastasis 20:757–766

27. O’Farrell AM, Abrams TJ, Yuen HA, Ngai TJ, Louie SG, YeeKW, Wong LM, Hong W, Lee LB, Town A, Smolich BD,Manning WC, Murray LJ, Heinrich MC, Cherrington JM (2003)SU11248 is a novel FLT3 tyrosine kinase inhibitor with potentactivity in vitro and in vivo. Blood 101:3597–3605

28. Podar K, Tonon G, Sattler M, Tai YT, Legouill S, Yasui H,Ishitsuka K, Kumar S, Kumar R, Pandite LN, Hideshima T,Chauhan D, Anderson KC (2006) The small-molecule VEGFreceptor inhibitor pazopanib (GW786034B) targets both tumorand endothelial cells in multiple myeloma. Proc Natl Acad SciUSA 103:19478–19483

29. Hu-Lowe DD, Zou HY, Grazzini ML, Hallin ME, Wickman GR,Amundson K, Chen JH, Rewolinski DA, Yamazaki S, Wu EY,

1798 Invest New Drugs (2012) 30:1791–1801

McTigue MA, Murray BW, Kania RS, O’Connor P, ShalinskyDR, Bender SL (2008) Nonclinical antiangiogenesis and anti-tumor activities of axitinib (AG-013736), an oral, potent, andselective inhibitor of vascular endothelial growth factor receptortyrosine kinases 1, 2, 3. Clin Cancer Res 14:7272–7283

30. Takeda M, Arao T, Yokote H, Komatsu T, Yanagihara K, SasakiH, Yamada Y, Tamura T, Fukuoka K, Kimura H, Saijo N, NishioK (2007) AZD2171 shows potent antitumor activity againstgastric cancer over-expressing fibroblast growth factor receptor2/keratinocyte growth factor receptor. Clin Cancer Res 13:3051–3057

31. Wilhelm SM, Carter C, Tang L, Wilkie D, McNabola A, Rong H,Chen C, Zhang X, Vincent P, McHugh M, Cao Y, Shujath J,Gawlak S, Eveleigh D, Rowley B, Liu L, Adnane L, Lynch M,Auclair D, Taylor I, Gedrich R, Voznesensky A, Riedl B, Post LE,Bollag G, Trail PA (2004) BAY 43–9006 exhibits broad spectrumoral antitumor activity and targets the RAF/MEK/ERK pathwayand receptor tyrosine kinases involved in tumor progression andangiogenesis. Clin Cancer Res 64:7099–7109

32. Wedge SR, Kendrew J, Hennequin LF, Valentine PJ, Barry ST,Brave SR, Smith NR, James NH, Dukes M, Curwen JO, ChesterR, Jackson JA, Boffey SJ, Kilburn LL, Barnett S, Richmond GH,Wadsworth PF, Walker M, Bigley AL, Taylor ST, Cooper L, BeckS, Jurgensmeier JM, Ogilvie DJ (2005) AZD2171: a highlypotent, orally bioavailable, vascular endothelial growth factorreceptor-2 tyrosine kinase inhibitor for the treatment of cancer.Cancer Res 65:4389–4400

33. Harris PA, Boloor A, Cheung M, Kumar R, Crosby RM, vis-WardRG, Epperly AH, Hinkle KW, Hunter RN III, Johnson JH, KnickVB, Laudeman CP, Luttrell DK, Mook RA, Nolte RT, Rudolph SK,Szewczyk JR, Truesdale AT, Veal JM, Wang L, Stafford JA (2008)Discovery of 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-m ethyl-benzenesulfonamide (Pazopanib), anovel and potent vascular endothelial growth factor receptorinhibitor. J Med Chem 51:4632–4640

34. Schmidinger M, Bellmunt J (2010) Plethora of agents, plethora oftargets, plethora of side effects in metastatic renal cell carcinoma.Cancer Treat Rev 36:416–424

35. Pfizer Inc; SUTENT, Summary of product characteristics;December 2010

36. Chang YS, Adnane J, Trail PA, Levy J, Henderson A, Xue D,Bortolon E, Ichetovkin M, Chen C, McNabola A, Wilkie D,Carter CA, Taylor IC, Lynch M, Wilhelm S (2007) Sorafenib(BAY 43–9006) inhibits tumor growth and vascularization andinduces tumor apoptosis and hypoxia in RCC xenograft models.Cancer Chemother Pharmacol 59:561–574

37. Bayer Heathcare AG (December 2010) Sorafenib, Summary ofproduct characteristics

38. Kumar R, Knick VB, Rudolph SK, Johnson JH, Crosby RM,Crouthamel MC, Hopper TM, Miller CG, Harrington LE, OnoriJA, Mullin RJ, Gilmer TM, Truesdale AT, Epperly AH, Boloor A,Stafford JA, Luttrell DK, Cheung M (2007) Pharmacokinetic-pharmacodynamic correlation frommouse to human with pazopanib,a multikinase angiogenesis inhibitor with potent antitumor andantiangiogenic activity. Mol Cancer Ther 6:2012–2021

39. GlaxoSmithKline (2011) Votrient® (pazopanib), Prescribinginformation, June 2010

40. Inai T, Mancuso M, Hashizume H, Baffert F, Haskell A, Baluk P,Hu-Lowe DD, Shalinsky DR, Thurston G, Yancopoulos GD,McDonald DM (2004) Inhibition of vascular endothelial growthfactor (VEGF) signaling in cancer causes loss of endothelialfenestrations, regression of tumor vessels, and appearance ofbasement membrane ghosts. Am J Pathol 165:35–52

41. De LA, Normanno N (2010) Tivozanib, a pan-VEGFR tyrosinekinase inhibitor for the potential treatment of solid tumors. IDrugs13:636–645

42. Gerber HP, Ferrara N (2005) Pharmacology and pharmacodynamicsof bevacizumab as monotherapy or in combination with cytotoxictherapy in preclinical studies. Cancer Res 65:671–680

43. Kim KJ, Li B, Winer J, Armanini M, Gillett N, Phillips HS,Ferrara N (1993) Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumour growth in vivo. Nature362:841–844

44. Roche Products Ltd (January 2011) Avastin, Summary of productcharacteristics

45. Pfizer Inc (November 2010) Torisel, Summary of productcharacteristics

46. Boulay A, Zumstein-Mecker S, Stephan C, Beuvink I, ZilbermannF, Haller R, Tobler S, Heusser C, O’Reilly T, Stolz B, Marti A,Thomas G, Lane HA (2004) Antitumor efficacy of intermittenttreatment schedules with the rapamycin derivative RAD001correlates with prolonged inactivation of ribosomal protein S6kinase 1 in peripheral blood mononuclear cells. Cancer Res64:252–261

47. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C,Bracarda S, Grunwald V, Thompson JA, Figlin RA, HollaenderN, Urbanowitz G, Berg WJ, Kay A, Lebwohl D, Ravaud A(2008) Efficacy of everolimus in advanced renal cell carcinoma: adouble-blind, randomised, placebo-controlled phase III trial. Lancet372:449–456

48. Novartis Pharmaceuticals UK Ltd.; Afinitor (everolimus), Summaryof Product Characteristics, May 2010. –>

49. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A,Staroslawska E, Sosman J, McDermott D, Bodrogi I, KovacevicZ, Lesovoy V, Schmidt-Wolf IG, Barbarash O, Gokmen E, O’TooleT, Lustgarten S, Moore L, Motzer RJ (2007) Temsirolimus,interferon alfa, or both for advanced renal-cell carcinoma. N Engl JMed 356:2271–2281

50. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, SiebelsM, Negrier S, Chevreau C, Solska E, Desai AA, Rolland F,Demkow T, Hutson TE, Gore M, Freeman S, Schwartz B, ShanM, Simantov R, Bukowski RM (2007) Sorafenib in advancedclear-cell renal-cell carcinoma. N Engl J Med 356:125–134

51. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, BukowskiRM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I,Bycott PW, Baum CM, Figlin RA (2007) Sunitinib versusinterferon alfa in metastatic renal-cell carcinoma. N Engl J Med356:115–124

52. Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Ou SS,Archer L, Atkins JN, Picus J, Czaykowski P, Dutcher J, Small EJ(2008) Bevacizumab plus interferon alfa compared with interferonalfa monotherapy in patients with metastatic renal cell carcinoma:CALGB 90206. J Clin Oncol 26:5422–5428

53. Escudier B, Pluzanska A, Koralewski P, Ravaud A, Bracarda S,Szczylik C, Chevreau C, Filipek M, Melichar B, Bajetta E,Gorbunova V, Bay JO, Bodrogi I, Jagiello-Gruszfeld A, Moore N(2007) Bevacizumab plus interferon alfa-2a for treatment ofmetastatic renal cell carcinoma: a randomised, double-blind phaseIII trial. Lancet 370:2103–2111

54. Motzer RJ, Hutson TE, Tomczak P, Dror Michaelson M,Bukowski RM, Oudard S, Negrier S, Szczylik C, Pili R,Bjarnason GA, Garcia-del-Muro X, Sosman JA, Solska E,Wilding G, Thompson JA, Kim ST, Chen I, Huang X, FiglinRA (2009) Overall survival and updated results for sunitinibversus interferon alfa in first-line treatment of patients withmetastatic renal cell carcinoma. J Clin Oncol 27:3584–3590

55. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, WagstaffJ, Barrios CH, Salman P, Gladkov OA, Kavina A, Zarba JJ, ChenM, McCann L, Pandite L, Roychowdhury DF, Hawkins RE(2010) Pazopanib in locally advanced or metastatic renal cellcarcinoma: results of a randomized phase III trial. J Clin Oncol28:1061–1068

Invest New Drugs (2012) 30:1791–1801 1799

56. Karaman MW, Herrgard S, Treiber DK, Gallant P, Atteridge CE,Campbell BT, Chan KW, Ciceri P, Davis MI, Edeen PT, FaraoniR, Floyd M, Hunt JP, Lockhart DJ, Milanov ZV, Morrison MJ,Pallares G, Patel HK, Pritchard S, Wodicka LM, Zarrinkar PP(2008) A quantitative analysis of kinase inhibitor selectivity. NatBiotechnol 26:127–132

57. Patel PH, Chaganti RS, Motzer RJ (2006) Targeted therapy formetastatic renal cell carcinoma. Br J Cancer 94:614–619

58. Sridhar SS,MackenzieMJ, Hotte SJ,Mukherjee SD,KollmannsbergerC, Haider MA, Chen EX, Wang R, Srinivasan R, Ivy SP, Moore MJ;Activity of cediranib (AZD2171) in patients (pts) with previouslyuntreated metastatic renal cell cancer (RCC). A phase II trial of thePMH Consortium. 2008, pp (Abstract 5047)

59. Rini BI, Wilding G, Hudes G, Stadler WM, Kim S, Tarazi J,Rosbrook B, Trask PC, Wood L, Dutcher JP (2009) Phase II studyof axitinib in sorafenib-refractory metastatic renal cell carcinoma.J Clin Oncol 27:4462–4468

60. Rixe O, Bukowski RM, Michaelson MD, Wilding G, Hudes GR,Bolte O, Motzer RJ, Bycott P, Liau KF, Freddo J, Trask PC, KimS, Rini BI (2007) Axitinib treatment in patients with cytokine-refractory metastatic renal-cell cancer: a phase II study. LancetOncol 8:975–984

61. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, BracardaS, Grunwald V, Thompson JA, Figlin RA, Hollaender N, Kay A,Ravaud A (2010) Phase 3 trial of everolimus for metastatic renalcell carcinoma: final results and analysis of prognostic factors.Cancer 116:4256–4265

62. Mills EJ, Rachlis B, O’Regan C, Thabane L, Perri D (2009)Metastatic renal cell cancer treatments: An indirect comparisonmeta-analysis. BMC Cancer 9:34

63. Escudier B, Bellmunt J, Negrier S, Bajetta E, Melichar B,Bracarda S, Ravaud A, Golding S, Jethwa S, Sneller V (2010)Phase III trial of bevacizumab plus Interferon alfa-2a in patientswith metastatic renal cell carcinoma (AVOREN): final analysis ofoverall survival. J Clin Oncol 28:2144–2150

64. Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, ArcherL, Atkins JN, Picus J, Czaykowski P, Dutcher J, Small EJ (2010)Phase III trial of bevacizumab plus interferon alfa versusinterferon alfa monotherapy in patients with metastatic renal cellcarcinoma: final results of CALGB 90206. J Clin Oncol 28:2137–2143

65. Sternberg CN, Hawkins RE, Szczylik C, Davis ID, Wagstaff J,McCann L, Chen M, Rubin SD (2010) Randomized, double blindphase III study of pazopanib in patients with advanced/metastaticrenal cell carcinoma (MRCC): Final overall survival (OS) results.Oral presentation at the 35th European Society of MedicalOncology Congress, Milan, Italy

66. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, StaehlerM, Negrier S, Chevreau C, Desai AA, Rolland F, Demkow T,Hutson TE, Gore M, Anderson S, Hofilena G, Shan M, Pena C,Lathia C, Bukowski RM (2009) Sorafenib for treatment of renalcell carcinoma: Final efficacy and safety results of the phase IIItreatment approaches in renal cancer global evaluation trial. J ClinOncol 27:3312–3318

67. Dutcher JP, de Souza P, McDermott D, Figlin RA, Berkenblit A,Thiele A, Krygowski M, Strahs A, Feingold J, Hudes G (2009)Effect of temsirolimus versus interferon-alpha on outcome ofpatients with advanced renal cell carcinoma of different tumorhistologies. Med Oncol 26:202–209

68. Atkins MB, Choueiri TK, Cho D, Regan M, Signoretti S (2009)Treatment selection for patients with metastatic renal cellcarcinoma. Cancer 115:2327–2333

69. Jacobsen J, Grankvist K, Rasmuson T, Ljungberg B (2006)Different isoform patterns for vascular endothelial growth factorbetween clear cell and papillary renal cell carcinoma. BJU Int97:1102–1108

70. Ljungberg BJ, Jacobsen J, Rudolfsson SH, Lindh G, Grankvist K,Rasmuson T (2006) Different vascular endothelial growth factor(VEGF), VEGF-receptor 1 and -2 mRNA expression profilesbetween clear cell and papillary renal cell carcinoma. BJU Int98:661–667

71. Maulik G, Shrikhande A, Kijima T, Ma PC, Morrison PT, SalgiaR (2002) Role of the hepatocyte growth factor receptor, c-Met, inoncogenesis and potential for therapeutic inhibition. CytokineGrowth Factor Rev 13:41–59

72. Bhargava P, Nosov DA, Esteves B, Al-Adhami M, Lipatov O,Lyulko A, Anishchenko AA, Chacko RT, Doval DC, SlichenmyerW (2010) Phase 2 randomized discontinuation trial (RDT) oftivozanib in patients with renal cell carcinoma (RCC): Results inpatients randomized to tivozanib vs placebo. Oral presentation atthe 35th European Society of Medical Oncology Congress, Milan,Italy

73. Kamba T, McDonald DM (2007) Mechanisms of adverse effectsof anti-VEGF therapy for cancer. Br J Cancer 96:1788–1795

74. Verheul HM, Pinedo HM (2007) Possible molecular mechanismsinvolved in the toxicity of angiogenesis inhibition. Nat RevCancer 7:475–485

75. Robert C, Faivre S, Raymond E, Armand JP, Escudier B (2005)Subungual splinter hemorrhages: a clinical window to inhibitionof vascular endothelial growth factor receptors? Ann Intern Med143:313–314

76. Iitaka M, Miura S, Yamanaka K, Kawasaki S, Kitahama S,Kawakami Y, Kakinuma S, Oosuga I, Wada S, Katayama S (1998)Increased serum vascular endothelial growth factor levels andintrathyroidal vascular area in patients with Graves’ disease andHashimoto’s thyroiditis. J Clin Endocrinol Metab 83:3908–3912

77. Ramsden JD (2000) Angiogenesis in the thyroid gland. JEndocrinol 166:475–480

78. Viglietto G, Romano A, Manzo G, Chiappetta G, Paoletti I,Califano D, Galati MG, Mauriello V, Bruni P, Lago CT, Fusco A,Persico MG (1997) Upregulation of the angiogenic factors PlGF,VEGF and their receptors (Flt-1, Flk-1/KDR) by TSH in culturedthyrocytes and in the thyroid gland of thiouracil-fed rats suggest aTSH-dependent paracrine mechanism for goiter hypervasculariza-tion. Oncogene 15:2687–2698

79. Wang JF, Milosveski V, Schramek C, Fong GH, Becks GP, HillDJ (1998) Presence and possible role of vascular endothelialgrowth factor in thyroid cell growth and function. J Endocrinol157:5–12

80. Eskens FA, Verweij J (2006) The clinical toxicity profile ofvascular endothelial growth factor (VEGF) and vascular endothelialgrowth factor receptor (VEGFR) targeting angiogenesis inhibitors; areview. Eur J Cancer 42:3127–3139

81. Ferraresi V, Catricala C, Ciccarese M, Ferrari A, Zeuli M,Cognetti F (2006) Severe skin reaction in a patient withgastrointestinal stromal tumor treated with imatinib mesylate.Anticancer Res 26:4771–4774

82. O’Donnell A, Faivre S, Burris HA III, Rea D, PapadimitrakopoulouV, Shand N, Lane HA, Hazell K, Zoellner U, Kovarik JM, Brock C,Jones S, Raymond E, Judson I (2008) Phase I pharmacokinetic andpharmacodynamic study of the oral mammalian target of rapamycininhibitor everolimus in patients with advanced solid tumors. J ClinOncol 26:1588–1595

83. Raymond E, Alexandre J, Faivre S, Vera K, Materman E, Boni J,Leister C, Korth-Bradley J, Hanauske A, Armand JP (2004)Safety and pharmacokinetics of escalated doses of weeklyintravenous infusion of CCI-779, a novel mTOR inhibitor, inpatients with cancer. J Clin Oncol 22:2336–2347

84. Drevs J, Siegert P, Medinger M, Mross K, Strecker R, ZirrgiebelU, Harder J, Blum H, Robertson J, Jurgensmeier JM, PuchalskiTA, Young H, Saunders O, Unger C (2007) Phase I clinical studyof AZD2171, an oral vascular endothelial growth factor signaling

1800 Invest New Drugs (2012) 30:1791–1801

inhibitor, in patients with advanced solid tumors. J Clin Oncol25:3045–3054

85. Siemann DW, Brazelle WD, Jurgensmeier JM (2009) Thevascular endothelial growth factor receptor-2 tyrosine kinaseinhibitor cediranib (Recentin; AZD2171) inhibits endothelial cellfunction and growth of human renal tumor xenografts. Int J RadiatOncol Biol Phys 73:897–903

86. Willett CG, Boucher Y, di Tomaso E, Duda DG, Munn LL, TongRT, Chung DC, Sahani DV, Kalva SP, Kozin SV, Mino M, CohenKS, Scadden DT, Hartford AC, Fischman AJ, Clark JW, Ryan DP,Zhu AX, Blaszkowsky LS, Chen HX, Shellito PC, Lauwers GY,Jain RK (2004) Direct evidence that the VEGF-specific antibodybevacizumab has antivascular effects in human rectal cancer. NatMed 10:145–147

87. Rini BI, Schiller JH, Fruehauf JP, Cohen EE, Tarazi JC, RosbrookB, Ricart AD, Olszanski AJ, Kim S, Spano J-P (2008) Associationof diastolic blood pressure (dBP) ≥90 mmHg with overall survival(OS) in patients treated with axitinib (AG- 013736). J Clin Oncol26:(abstr)

88. Rini BI, Cohen DP, Lu D, Chen I, Hariharan S, Gore ME, FiglinRA, Baum MS, Motzer RJ (2010) Hypertension (HTN) as abiomarker of efficacy in patients (pts) with metastatic renal cellcarcinoma (mRCC) treated with sunitinib. American Socierty forClinical Oncology 2010 Genitourinary Cancers SymposiumAbstract 312

89. Carlomagno F, Anaganti S, Guida T, Salvatore G, Troncone G,Wilhelm SM, Santoro M (2006) BAY 43–9006 inhibition ofoncogenic RET mutants. J Natl Cancer Inst 98:326–334

90. Schmidinger M, Vogl UM, Bojic M, Lamm W, Heinzl H, HaitelA, Clodi M, Kramer G, Zielinski CC (2010) Hypothyroidism inpatients with renal cell carcinoma: blessing or curse? Cancer Epubahead of print

91. Novartis Pharmaceuticals UK Ltd. (2009) Afinitor (everolimus),Summary of Product Characteristics, March 2009

92. Schmidinger M, Zielinski CC, Vogl UM, Bojic A, Bojic M,Schukro C, Ruhsam M, Hejna M, Schmidinger H (2008) Cardiactoxicity of sunitinib and sorafenib in patients with metastatic renalcell carcinoma. J Clin Oncol 26:5204–5212

93. Kido M, Du L, Sullivan CC, Li X, Deutsch R, Jamieson SW,Thistlethwaite PA (2005) Hypoxia-inducible factor 1-alpha reducesinfarction and attenuates progression of cardiac dysfunction aftermyocardial infarction in themouse. J AmColl Cardiol 46:2116–2124

94. Parisi Q, Biondi-Zoccai GG, Abbate A, Santini D, Vasaturo F,Scarpa S, Bussani R, Leone AM, Petrolini A, Silvestri F, BiasucciLM, Baldi A (2005) Hypoxia inducible factor-1 expressionmediates myocardial response to ischemia late after acutemyocardial infarction. Int J Cardiol 99:337–339

95. Waltenberger J (1997) Modulation of growth factor action: implica-tions for the treatment of cardiovascular diseases. Circulation96:4083–4094

96. Merseburger AS, Simon A, Waalkes S, Kuczyk MA (2009)Sorafenib reveals efficacy in sequential treatment of metastaticrenal cell cancer. Expert Rev Anticancer Ther 9:1429–1434

97. www.clinicaltrials.gov (2010)98. Sosman J, Puzanov I (2009) Combination targeted therapy in

advanced renal cell carcinoma. Cancer 115:2368–2375

Invest New Drugs (2012) 30:1791–1801 1801